Assessing Healthy Vaccinee Bias in COVID-19 Vaccine Effectiveness Studies: A National Cohort Study in Qatar =========================================================================================================== * Hiam Chemaitelly * Houssein H. Ayoub * Peter Coyle * Patrick Tang * Mohammad R. Hasan * Hadi M. Yassine * Asmaa A. Al Thani * 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 * Hamad Eid Al-Romaihi * Mohamed H. Al-Thani * Abdullatif Al-Khal * Roberto Bertollini * Adeel A. Butt * Laith J. Abu-Raddad ## Abstract **Background:** This study investigated the presence of the healthy vaccinee bias in two COVID-19 vaccine effectiveness studies, involving primary series and booster vaccinations, as well as its temporal patterns and variability across different subpopulations, by examining the association between COVID-19 vaccination and non-COVID-19 mortality in Qatar. **Methods:** Two matched, retrospective cohort studies assessed the incidence of non-COVID-19 death in national cohorts of individuals with a primary series vaccination versus no vaccination (two-dose analysis), and individuals with three-dose (booster) vaccination versus primary series vaccination (three-dose analysis), from January 5, 2021, to April 9, 2024. **Results:** The adjusted hazard ratio (aHR) for non-COVID-19 death was 0.76 (95% CI: 0.64-0.90) in the two-dose analysis and 0.85 (95% CI: 0.67-1.07) in the three-dose analysis. In the first six months of follow-up in the two-dose analysis, the aHR was 0.35 (95% CI: 0.27-0.46); however, the combined analysis of all subsequent periods showed an aHR of 1.52 (95% CI: 1.19- 1.94). In the first six months of follow-up in the three-dose analysis, the aHR was 0.31 (95% CI: 0.20-0.50); however, the combined analysis of all subsequent periods showed an aHR of 1.37 (95% CI: 1.02-1.85). The overall effectiveness of the primary series and third-dose vaccinations against severe, critical, or fatal COVID-19 was 95.9% (95% CI: 94.0-97.1) and 34.1% (95% CI:-46.4-76.7), respectively. Subgroup analyses showed that the healthy vaccinee bias is pronounced among those aged 50 years and older and among those more clinically vulnerable to severe COVID-19. **Conclusion:** A strong healthy vaccinee bias was observed in the first six months following vaccination. This bias may have stemmed from a lower likelihood of vaccination among seriously ill, end-of-life individuals, and less mobile elderly populations. Despite this bias, vaccination provided strong protection against severe COVID-19. Keywords * COVID-19 * vaccine * all-cause mortality * bias * cohort study * epidemiology ## Introduction While randomized controlled trials remain the gold standard for determining vaccine efficacy, they often have short follow-up durations, primarily involve healthy participants, and may cover only a limited range of clinical outcomes [1–3]. Real-world observational studies are frequently utilized to evaluate vaccine effectiveness beyond the controlled trial environment [2, 4, 5]. In these settings, diverse health statuses, variable health behaviors, and structural determinants can influence vaccine uptake, potentially biasing estimates of effectiveness [2, 4, 5]. The accuracy of vaccine effectiveness estimates from observational studies can be impacted by two opposite forms of bias: indication bias and healthy vaccinee bias [2, 6]. Indication bias occurs when individuals with underlying health conditions are more likely to receive vaccination, leading to an underestimation of vaccine effectiveness [6]. Conversely, healthy vaccinee bias occurs when healthier or health-conscious individuals are more likely to receive vaccination, leading to an overestimation of vaccine effectiveness [6]. Both forms of bias can skew effectiveness results by conflating health status with the protective effects of the vaccine [2, 6]. Such biases have been documented in observational studies of influenza vaccine effectiveness [2, 6–8], coronavirus disease 2019 (COVID-19) vaccine effectiveness [9–11], and prescriptive medication effectiveness [2]. In particular, studies have documented a strong effect of healthy vaccinee bias in estimations of influenza vaccine effectiveness among the elderly [2, 8]. While it is common for studies to control for such bias by adjusting for coexisting conditions based on administrative healthcare utilization databases, this approach may not sufficiently or properly adjust for bias, as it may not capture the illness severity, recency, duration, or the functional status of individuals—factors that can confound the association between vaccination and health outcomes, particularly in the elderly [2, 6, 8]. Assessing coexisting conditions based on database variables can also be affected by differential misclassification, as these variables not only reflect chronic diseases but also inherently measure utilization of health services [2]. A notable feature of the healthy vaccinee bias is its potentially strong time dependence, most pronounced immediately after vaccination but gradually diminishing [2, 8]. This trend is observed because seriously ill individuals, those with deteriorating health, and frail, less mobile elderly persons are less likely to be vaccinated, resulting in a higher short-term mortality risk among the unvaccinated [2, 8]. Studies show that elderly individuals who are more mobile or have fewer functional limitations are more likely to be vaccinated [2, 12]. For instance, one study demonstrated that the inability to bathe independently was associated with a 13-fold increase in mortality risk and a 52% reduced likelihood of receiving a vaccination [12]. Over time, as the less functional and seriously ill individuals in the unvaccinated group die, the disparities between the vaccinated and unvaccinated groups diminish [2, 8]. Additional changes in health status over time among members of both groups also contribute to this equilibration [2, 8]. In this national retrospective cohort study, the presence of indication or healthy vaccinee bias was investigated within a conventionally designed and well-controlled COVID-19 vaccine effectiveness study covering both primary series and booster mRNA vaccinations. Three aspects of this bias were explored: its existence, its temporal pattern, and its variability across different subpopulations. This was accomplished by assessing the association between COVID-19 vaccination and non-COVID-19 mortality, which serves as a suitable control outcome to gauge the degree of potential residual bias in well-controlled vaccine effectiveness estimates against SARS-CoV-2 infection or severe forms of COVID-19 [2]. ## Methods ### **Study population, data sources, and vaccination** This study was conducted among the resident population of Qatar from January 5, 2021, which marks the earliest record of a completed COVID-19 primary series vaccination, to April 9, 2024, the study’s end date. Data on COVID-19 laboratory testing, vaccination, hospitalization, and death were retrieved from the integrated, nationwide digital health information platform (Section S1 in Supplementary Appendix). Deaths not related to COVID-19 were sourced from the national federated mortality database, which captures all deaths in the country, occurring in healthcare facilities and elsewhere, including forensic deaths investigated by Qatar’s Ministry of Interior. The national digital health information platform includes all SARS-CoV-2-related records, encompassing COVID-19 vaccinations, hospitalizations, and polymerase chain reaction (PCR) tests, irrespective of location or facility, and, from January 5, 2022, medically supervised rapid antigen tests (Section S2). Until October 31, 2022, Qatar maintained an extensive testing approach, testing 5% of the population weekly, primarily for routine purposes such as screening or travel-related requirements [13, 14]. From November 1, 2022, onwards, testing was reduced to below 1% of the population weekly [15]. Most COVID-19 infections in Qatar were identified through routine testing rather than symptomatic presentation (Section S1) [13, 14]. The national platform further contains data on coexisting conditions for individuals who have accessed care through the universal public health care system since the establishment of the digital health platform in 2013 (Section S3). COVID-19 vaccination in Qatar was almost exclusively done utilizing mRNA vaccines [15–17] and was administered throughout the pandemic according to the United States Food and Drug Administration approved protocols. Vaccination was provided free of charge to all individuals, irrespective of citizenship, exclusively through the public healthcare system [18]. The rollout strategy prioritized frontline healthcare workers, individuals with severe or multiple chronic conditions, and individuals aged 50 years or older [13]. Demographic information was obtained from the national health registry. Qatar’s demographic composition is distinct, with only 9% of the population aged 50 years or older and 89% being resident expatriates from over 150 countries [19]. Further details on Qatar’s population and COVID-19 databases have been previously published [13, 14, 19–23]. ### Study design Two national, matched, retrospective cohort studies were conducted to investigate the potential for indication bias or healthy vaccinee bias influencing the estimated effectiveness of COVID-19 primary series (two-dose) and booster (three-dose) vaccinations in Qatar’s population. Given the objective of exploring these biases, the studies were designed as vaccine effectiveness studies, adhering to cohort designs developed and implemented in Qatar’s population since the pandemic’s onset [15, 20, 22, 24–28]. In the first study (two-dose analysis), the incidence of non-COVID-19 death in the national cohort of individuals who received the primary series vaccination (designated as the two-dose cohort) was compared with that in the national cohort of unvaccinated individuals (designated as the unvaccinated cohort). In the second study (three-dose analysis), the incidence of non-COVID-19 death in the national cohort of individuals who received a third (booster) dose of vaccination (designated as the three-dose cohort) was compared with that in the two-dose cohort. For both studies, vaccine effectiveness was also estimated by comparing the incidence of SARS-CoV-2 infection and of severe forms of COVID-19 between the study cohorts. Severe forms of COVID-19 were classified by trained medical personnel independent of the study investigators. The classifications were based on individual chart reviews, adhering to the World Health Organization (WHO) guidelines for defining COVID-19 case severity (acute-care hospitalization) [29], criticality (intensive-care-unit hospitalization) [29], and fatality [30] (Section S4). These evaluations were implemented throughout the pandemic as part of a national protocol, under which every individual with a SARS-CoV-2-positive test and a concurrent COVID-19 hospital admission was assessed for infection severity at regular intervals until discharge or death, regardless of the hospital length of stay. All deaths in the population not classified as COVID-19 deaths were deemed non-COVID-19 deaths. Incidence of infection was defined as any PCR-positive or rapid-antigen-positive test after the start of follow-up, irrespective of symptomatic presentation. Individuals whose infection progressed to severe, critical, or fatal COVID-19 were classified based on their worst outcome, starting with COVID-19 death [30], followed by critical disease [29], and then severe disease [29] (Section S4). Incidence of outcomes of severe forms of COVID-19 was recorded on the date of the SARS-CoV-2-positive test confirming the infection. ### Cohorts’ eligibility and matching Individuals qualified for inclusion in the two-dose cohort if they received two doses of an mRNA vaccine, and in the three-dose cohort if they received three doses of an mRNA vaccine. Those who were administered the ChAdOx1 nCoV-19 (AZD1222) vaccine, a small proportion of the population, or the pediatric 10-µg BNT162b2 vaccine were excluded. Individuals qualified for inclusion in the unvaccinated cohort if they had no vaccination record at the start of follow-up. Cohorts were matched exactly one-to-one by sex, 10-year age group, nationality, exact coexisting conditions (Section S3), and prior documented SARS-CoV-2 infection status (no prior infection, prior pre-omicron infection, prior omicron infection, or prior pre-omicron and omicron infections). Since prior infection may affect the health status of an individual, such as by causing Long COVID [31] or affecting vaccination uptake [32], the matching by prior infection status aimed to balance this confounder across cohorts. Prior infections were classified as pre-omicron if they occurred before December 19, 2021, the onset of the omicron wave in Qatar [14], and as omicron thereafter. For the two-dose analysis, individuals who received their second vaccine dose in a specific calendar week in the two-dose cohort were additionally matched to individuals who had a record of a SARS-CoV-2-negative test in that same calendar week in the unvaccinated cohort. This matching was done to ensure that matched pairs had presence in Qatar over the same time period. Individuals who were tested after death or who had an unascertained or discrepant death date were excluded. Similarly, for the three-dose analysis, individuals who received their third vaccine dose in a specific calendar week in the three-dose cohort were matched to individuals who had a record of a SARS-CoV-2-negative test in that same calendar week in the two-dose cohort. Additionally, individuals in the three-dose cohort were matched to individuals in the two-dose cohort by the calendar week of the second vaccine dose. These matching criteria ensured that the paired individuals received their primary series vaccinations at the same time and were present in Qatar during the same period. Iterative matching was implemented so that, at the start of follow-up, individuals were alive, had maintained their vaccination status, had the same prior infection status as their match, and had no documented SARS-CoV-2 infection within the previous 90 days. The 90-day threshold was used to avoid misclassification of a previous (prolonged) SARS-CoV-2 infection as an incident infection [14, 33–35]. Consequently, a prior infection was defined as a SARS-CoV-2-positive test that occurred ≥90 days before the start of follow-up. The above-detailed matching approach aimed to balance observed confounders that could potentially affect the risk of non-COVID-19 death or the risk of infection across the exposure groups [19, 23, 36–39]. The matching factors were selected based on findings from earlier studies on Qatar’s population [13, 17, 18, 23, 40–42]. The matching algorithm was implemented using *ccmatch* command in Stata 18.0 supplemented with conditions to retain only controls that fulfilled the eligibility criteria, and was iterated using loops with as many replications as needed until exhaustion (i.e., no more matched pairs could be identified). According to this study design and matching approach, individuals in the matched unvaccinated cohort in the two-dose analysis may have contributed follow-up time before receiving the primary series vaccination and subsequently contributed follow-up time as part of the two-dose cohort after receiving the primary series vaccination. Similarly, in the three-dose analysis, individuals in the matched two-dose cohort may have contributed follow-up time before receiving the third (booster) dose, as part of the two-dose cohort, and subsequently contributed follow-up time as part of the three-dose cohort after receiving the third dose. ### Cohorts’ follow-up Follow-up started from the calendar date of the second dose in the two-dose analysis and from the calendar date of the third dose in the three-dose analysis. To ensure exchangeability [20, 43], both members of each matched pair were censored at the earliest occurrence of receiving an additional vaccine dose. Accordingly, individuals were followed until the first of any of the following events: a documented SARS-CoV-2 infection (irrespective of symptoms), first-dose vaccination for individuals in the unvaccinated cohort (with matched-pair censoring), third-dose vaccination for individuals in the two-dose cohort (with matched-pair censoring), fourth-dose vaccination for individuals in the three-dose cohort (with matched-pair censoring), death, or the administrative end of follow-up at the end of the study. ### 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; Table S1). ### 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 [44]. The cumulative incidence of non-COVID-19 death, defined as proportion of individuals at risk whose primary endpoint during follow-up was a non-COVID-19 death, was estimated using the Kaplan-Meier estimator method. Incidence rate of non-COVID-19 death in each cohort, defined as number of non-COVID-19 deaths divided by number of person-weeks contributed by all individuals in the cohort, was estimated along with the corresponding 95% confidence interval (CI), using a Poisson log-likelihood regression model with the Stata 18.0 *stptime* command. Overall adjusted hazard ratio (aHR), comparing incidence of non-COVID-19 death between the cohorts, and corresponding 95% CI, were calculated using Cox regression models with adjustment for the matching factors, via the Stata 18.0 *stcox* command. This adjustment was implemented to ensure precise and unbiased estimation of the standard variance [45]. CIs were not adjusted for multiplicity. Schoenfeld residuals and log-log plots for survival curves were used to examine the proportional hazards assumption. An aHR less than 1 indicated evidence of a healthy vaccinee bias. An aHR greater than 1 indicated evidence of an indication bias. The overall aHR provides a weighted average of the time-varying hazard ratio [46]. To explore differences in the risk of non-COVID-19 death over time, the aHR was also estimated by 6-month intervals from the start of follow-up, using separate Cox regressions, with "failure" restricted to specific time intervals. Subgroup analyses estimating the overall aHR stratified by age group (<50 years versus ≥50 years), clinical vulnerability status, and prior infection status were also conducted. Individuals were classified as less clinically vulnerable to severe COVID-19 if they were <50 years of age and had one or no coexisting conditions, and as more clinically vulnerable to severe COVID-19 if they were either ≥50 years of age or <50 years of age but with ≥2 coexisting conditions [21, 22]. The study analyzed non-COVID-19 mortality in the population of Qatar. However, some deaths may have occurred outside Qatar when expatriates were traveling abroad or had permanently left the country after the start of follow-up. The matching strategy aimed to mitigate any differential effects of these out-of-country deaths on the matched groups, for instance, by matching on a SARS-CoV-2-negative test among controls to ensure their presence in Qatar during the same period. To assess whether our results could have been affected by bias due to out-of-country deaths or the matching requirement of a SARS-CoV-2-negative test, two sensitivity analyses were conducted: first, by restricting the cohorts to only Qataris, where out-of-country deaths are unlikely, and second, by eliminating the requirement for matching by a SARS-CoV-2-negative test. Analogous methods were used to compare incidence of SARS-CoV-2 infection and of severe forms of COVID-19 between study cohorts. The overall aHR, comparing incidence of SARS-CoV-2 infection (or severe forms of COVID-19) between study cohorts, was calculated, including an additional adjustment for the testing rate. Vaccine effectiveness against infection and against severe forms of COVID-19, along with the associated 95% CIs, were derived from the aHR as 1-aHR if the aHR was <1, and as 1/aHR-1 if the aHR was ≥1 [22, 47]. This approach ensured a symmetric scale for both negative and positive effectiveness, spanning from-100%- 100%, resulting in a meaningful interpretation of effectiveness, regardless of the value being positive or negative. Statistical analyses were performed using Stata/SE version 18.0 (Stata Corporation, College Station, TX, USA). ## Results ### **Two-dose analysis** Fig. S1 illustrates the process of selecting the study cohorts. Table 1 outlines the cohorts’ baseline characteristics. Each matched cohort comprised 812,583 individuals. Median date of the second vaccine dose was June 21, 2021 for the two-dose cohort. Median duration of follow-up was 206 days (interquartile range (IQR), 41-925 days) in the two-dose cohort and 199 days (IQR, 36-933 days) in the unvaccinated cohort (Fig. 1A). ![Fig. 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/07/29/2024.07.28.24311115/F1.medium.gif) [Fig. 1:](http://medrxiv.org/content/early/2024/07/29/2024.07.28.24311115/F1) Fig. 1: Cumulative incidence of non-COVID-19 death in the matched A) two-dose cohort compared to the unvaccinated cohort and B) three-dose cohort compared to the two-dose cohort. View this table: [Table 1:](http://medrxiv.org/content/early/2024/07/29/2024.07.28.24311115/T1) Table 1: Baseline characteristics of the full and matched cohorts for investigating indication bias or healthy vaccinee bias among recipients of primary series or booster (third dose) vaccination in Qatar. During follow-up, 237 non-COVID-19 deaths occurred in the two-dose cohort compared to 306 in the unvaccinated cohort (Table 2A and Fig. S1). There were 54,427 SARS-CoV-2 infections recorded in the two-dose cohort, of which 23 progressed to severe, 6 to critical, and none to fatal COVID-19. Meanwhile, 57,974 SARS-CoV-2 infections were recorded in the unvaccinated cohort, of which 539 progressed to severe, 66 to critical, and 25 to fatal COVID-19. View this table: [Table 2:](http://medrxiv.org/content/early/2024/07/29/2024.07.28.24311115/T2) Table 2: Hazard ratios for incidence of non-COVID-19 death, SARS-CoV-2 infection, and severe, critical, or fatal COVID-19 in the A) two-dose analysis and B) three-dose analysis. View this table: [Table 3:](http://medrxiv.org/content/early/2024/07/29/2024.07.28.24311115/T3) Table 3: Subgroup analyses. Hazard ratios for incidence of non-COVID-19 death stratified by age group, clinical vulnerability status, and prior infection status in the A) two-dose analysis and B) three-dose analysis. The cumulative incidence of non-COVID-19 death was 0.070% (95% CI: 0.061-0.081%) for the two-dose cohort and 0.071% (95% CI: 0.062-0.080%) for the unvaccinated cohort after 990 days of follow-up (Fig. 1A). The overall aHR comparing the incidence of non-COVID-19 death in the two-dose cohort to that in the unvaccinated cohort was 0.76 (95% CI: 0.64-0.90), indicating evidence of a healthy vaccinee bias (Table 2A). In the first six months of follow-up, the aHR was 0.35 (95% CI: 0.27-0.46), indicating strong evidence of a healthy vaccinee bias (Fig. 2A). However, the combined analysis of all periods after the first six months showed an aHR of 1.52 (95% CI: 1.19-1.94), pointing to an indication bias. ![Fig. 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/07/29/2024.07.28.24311115/F2.medium.gif) [Fig. 2:](http://medrxiv.org/content/early/2024/07/29/2024.07.28.24311115/F2) Fig. 2: Adjusted hazard ratios for incidence of non-COVID-19 death in the A) two-dose analysis and B) three-dose analysis, by 6-month interval of follow-up. The subgroup analyses estimated the aHR at 0.89 (95% CI: 0.72-1.11) among individuals under 50 years of age and at 0.56 (95% CI: 0.42-0.75) among those 50 years of age and older (Table 2A). The aHR was 0.98 (95% CI: 0.79-1.22) for those less clinically vulnerable to severe COVID-19 and 0.51 (95% CI: 0.39-0.68) for the more clinically vulnerable group. The aHR by prior infection status was 0.74 (95% CI: 0.63-0.89) for no prior infection and 1.00 (95% CI: 0.45-2.20) for prior pre-omicron infection. In the two sensitivity analyses—one including only Qataris and the other also including only Qataris but without matching on a SARS-CoV-2-negative test among controls—the aHRs for non-COVID-19 death were 0.29 (95% CI: 0.19-0.43) and 0.38 (95% CI: 0.30-0.50), respectively (Table 4A). Both analyses are consistent with each other and with the main analysis results. However, the healthy vaccinee bias effect is more pronounced among Qataris, as the proportion of individuals above 50 years of age or those with serious coexisting conditions is substantially higher among Qataris compared to the rest of the population, which primarily comprises working-age male craft and manual workers.[19, 23, 39] View this table: [Table 4:](http://medrxiv.org/content/early/2024/07/29/2024.07.28.24311115/T4) Table 4: Sensitivity analyses. Hazard ratios for incidence of non-COVID-19 death among Qataris with and without matching on a SARS-CoV-2-negative test among controls in the A) two-dose analysis and B) three-dose analysis. The overall effectiveness of primary series vaccination compared to no vaccination was 10.7% (95% CI: 9.6-11.7) against infection and 95.9% (95% CI: 94.0-97.1) against severe, critical, or fatal COVID-19 (Table 2A). ### Three-dose analysis Fig. S2 illustrates the process of selecting the study cohorts. Table 1 outlines the cohorts’ baseline characteristics. Each matched cohort comprised 330,568 individuals. The median date of the second vaccine dose was May 15, 2021, for both the two-dose and three-dose cohorts. The median date of the third vaccine dose in the three-dose cohort was January 24, 2022. The median duration of follow-up was 695 days (IQR, 66-802 days) in the three-dose cohort and 685 days (IQR, 49-798 days) in the two-dose cohort (Fig. 1B). During follow-up, 132 non-COVID-19 deaths occurred in the three-dose cohort compared to 147 in the two-dose cohort (Table 2B and Fig. S2). There were 26,842 SARS-CoV-2 infections recorded in the three-dose cohort, of which 3 progressed to severe, 2 to critical, and 1 to fatal COVID-19. Meanwhile, 35,411 SARS-CoV-2 infections were recorded in the two-dose cohort, of which 8 progressed to severe, 1 to critical, and none to fatal COVID-19. The cumulative incidence of non-COVID-19 death was 0.064% (95% CI: 0.054-0.076%) for the three-dose cohort and 0.070% (95% CI: 0.059-0.083%) for the two-dose cohort, after 840 days of follow-up (Fig. 1B). The overall aHR comparing the incidence of non-COVID-19 death in the three-dose cohort to that in the two-dose cohort was 0.85 (95% CI: 0.67-1.07), indicating no overall evidence of a healthy vaccinee bias (Table 2B). In the first six months of follow-up, the aHR was 0.31 (95% CI: 0.20-0.50), indicating strong evidence of a healthy vaccinee bias (Fig. 2B). However, the combined analysis of all subsequent periods showed an aHR of 1.37 (95% CI: 1.02-1.85), pointing to an indication bias. The subgroup analyses estimated the aHR at 0.90 (95% CI: 0.67-1.20) among individuals under 50 years of age and at 0.76 (95% CI: 0.51-1.13) among those 50 years of age and older (Table 2A). The aHR was 0.91 (95% CI: 0.67-1.22) for those less clinically vulnerable to severe COVID-19 and 0.76 (95% CI: 0.52-1.12) for the more clinically vulnerable group. The aHR by prior infection status was 0.79 (95% CI: 0.61-1.01) for no prior infection, 1.63 (95% CI: 0.71- 3.72) for prior pre-omicron infection, and 1.32 (95% CI: 0.30-5.91) for prior omicron infection. In the two sensitivity analyses—one including only Qataris and the other also including only Qataris but without matching on a SARS-CoV-2-negative test among controls—the aHRs for non-COVID-19 death were 0.76 (95% CI: 0.43-1.32) and 0.77 (95% CI: 0.53-1.13), respectively (Table 4B). Both analyses are consistent with each other and with the main analysis results. The overall effectiveness of the third-dose (booster) vaccination compared to primary series vaccination was 26.3% (95% CI: 25.2-27.5) against infection and 34.1% (95% CI: -46.4-76.7) against severe, critical, or fatal COVID-19 (Table 2B). ## Discussion The results affirm the presence of a healthy vaccinee bias, evident during the first six months after vaccination. Notably, the same bias, with a similar magnitude, was observed in both primary series and booster vaccinations, suggesting a consistent underlying phenomenon. This bias, similar to that found in influenza vaccine effectiveness studies [2, 8], may stem from seriously ill and end-of-life individuals, such as terminal cancer patients, as well as frail and less mobile elderly persons, being less likely to be vaccinated [2, 8]. This leads to a higher short-term mortality risk among the unvaccinated [2, 8]. This is supported by this bias being only evident in the first six months, and specifically among those aged 50 years and older and those more clinically vulnerable to severe COVID-19. Given the strength of this bias, it seems unlikely that it can be attributed to an effect of vaccination-induced nonspecific immune activation or trained/bystander immunity that protects against a range of infectious and non-infectious outcomes [11, 48–50]. While we observed a healthy vaccinee bias, it is possible that both a healthy vaccinee bias and an indication bias were present, albeit at different strengths and times. The healthy vaccinee bias was evident in the first six months after vaccination. Subsequently, there was supporting evidence for an indication bias, potentially reflecting the depletion of seriously ill individuals during those six months among those who were not vaccinated, leaving this group enriched with relatively healthier individuals. Meanwhile, the vaccinated group continued to include those with less serious health conditions who received the vaccination to improve their health status. These findings raise a concern, as vaccine effectiveness is typically estimated for the first few months after vaccination for seasonal infections, or for infections with repeated waves, such as influenza and SARS-CoV-2 [2, 6, 8, 51]. These findings support a rationale for excluding seriously ill, immunosuppressed, or functionally impaired individuals in studies of vaccine effectiveness in the general population [2, 8]. Although a healthy vaccinee bias was observed in this study, the extent to which this bias may have skewed the estimated vaccine effectiveness remains uncertain. This bias is presumably more likely to affect vaccine effectiveness against severe forms of COVID-19 than against infection alone [10]. The impact of this bias might also have been mitigated somewhat by using specific infection outcomes—such as severe, critical, or fatal COVID-19—and by confirming infections through laboratory methods, rather than relying on broad non-specific outcomes like all-cause mortality, commonly used in influenza vaccine effectiveness studies [2, 6, 8]. Ironically, the overall healthy vaccinee bias over the entire duration of follow-up may have been partially mitigated by the indication bias observed after the first six months of vaccination. Despite the presence of a healthy vaccinee bias, the results still confirm strong protection from vaccination against severe forms of COVID-19, as the observed effectiveness for the primary series was extremely high, at 96%. However, vaccine effectiveness against infection was modest, which is expected given that this type of protection rapidly diminishes within the first few months after vaccination [13, 40, 51, 52], and effectiveness was estimated over three years of follow-up. This study has limitations. The specific causes of non-COVID-19 deaths were not available to the investigators, which limited the scope of additional analyses. A number of non-COVID-19 deaths had unascertained or discrepant death date; therefore, these individuals were excluded from the study from the onset. However, this exclusion is not likely to materially affect the analyses, as there were only 23 deaths with unascertained or discrepant death dates in the entire population of Qatar over the three years of this study. Documented COVID-19 deaths may not include all deaths that occurred because of COVID-19 [53, 54], and thus there could be some misclassification bias affecting the distinction between COVID-19 and non-COVID-19 deaths. However, the number of COVID-19 deaths was small (Fig. S1 and Fig. S2), and the COVID-19 death rate in the young and working-age population of Qatar has been one of the lowest worldwide, with less than 0.1% of documented infections resulting in death [21, 23, 55, 56]. Earlier studies suggest that the number of undocumented COVID-19 deaths in Qatar is too small to appreciably affect the analyses of this study [21, 23, 56]. The study analyzed all deaths occurring within Qatar; however, some deaths might have occurred outside the country. Data on deaths outside the country were not available for our analysis. Nevertheless, the matching process was designed to ensure that participants were present in Qatar during the same period and to balance the risk of out-of-country deaths across cohorts. Consequently, these out-of-country deaths are not likely to have influenced the comparative outcomes of the matched cohorts. Further supporting our results, the sensitivity analysis, which was restricted to only Qataris—a group very unlikely to experience out-of-Qatar deaths—corroborated the main study results. The national testing database served as a sampling frame for unvaccinated individuals in Qatar. However, this database does not capture individuals who have never had a SARS-CoV-2 test since the onset of the pandemic. Nevertheless, testing has been extensive in Qatar, with the vast majority conducted for routine reasons [13, 14]. Given the widespread testing mandates and the large volume of tests conducted, it is not likely that any citizen or resident in Qatar has not had at least one SARS-CoV-2 test since the onset of the pandemic [13, 14]. Matched unvaccinated individuals were required to have tested negative for SARS-CoV-2 in the week their matched vaccinated counterparts received their vaccine, ensuring that both groups were present in Qatar during the same time period. Different eligibility criteria between the two arms could bias the study if there was a correlation between testing and non-COVID-19 death. However, the sensitivity analysis for Qataris, which eliminated the requirement for matching by a SARS-CoV-2-negative test, confirmed similar results, suggesting that this matching requirement may not have biased the results. The study was conducted in a specific national population consisting mainly of healthy working-age adults, thus the generalizability of the findings to other populations remains uncertain. As an observational study, the investigated cohorts were neither blinded nor randomized, so unmeasured or uncontrolled confounding factors cannot be excluded. Although matching accounted for key factors affecting risks of death and infection [19, 36–39], it was not possible for other factors such as geography or occupation, for which data were unavailable. However, Qatar is essentially a city-state where infection incidence was broadly distributed across neighborhoods. Nearly 90% of Qatar’s population are expatriates from over 150 countries, primarily coming for employment [19]. In this context, nationality, age, and sex serve as powerful proxies for socioeconomic status [19, 36–39]. Nationality is also strongly associated with occupation [19, 37–39]. The matching procedure used in this study has been evaluated in previous studies with different epidemiologic designs and using control groups to test for null effects [13, 17, 18, 40, 57]. These prior studies demonstrated that this procedure balances differences in infection exposure to estimate vaccine effectiveness [13, 17, 18, 40, 57], suggesting that the matching strategy may also have mitigated differences in mortality risk. Lastly, the aHRs were estimated both overall and by 6-month intervals from the start of follow-up. However, the interval-based analysis can be susceptible to changes in the composition of the study population over time. The study has strengths. It was implemented on Qatar’s entire population and sizable cohorts, representing a diverse range of national backgrounds. Extensive, validated databases from numerous prior COVID-19 studies were utilized in this study. The availability of an integrated digital health information platform provided data on various confounding factors, facilitating rigorous matching based on specific coexisting conditions and prior infection statuses. The ascertainment of COVID-19 deaths was meticulously conducted by trained personnel, adhering to a national protocol and WHO guidelines for classifying COVID-19 case fatalities [30]. In conclusion, a healthy vaccinee bias was observed, but only in the first six months following COVID-19 vaccination and specifically among those aged 50 years and older and those more clinically vulnerable to severe COVID-19. The same bias, with similar magnitude, was observed for both primary series and booster vaccinations, suggesting a consistent underlying phenomenon, perhaps a lower likelihood of vaccination among seriously ill, end-of-life individuals, and less mobile elderly populations. COVID-19 booster vaccine policies should account for this bias when interpreting effectiveness estimates and formulating vaccine guidelines. Despite this bias, the results confirm strong protection from vaccination against severe forms of COVID-19. ## 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. The data are available under restricted access for preservation of confidentiality of patient data. Access can be obtained through a direct application for data access to Her Excellency the Minister of Public Health ([https://www.moph.gov.qa/english/OurServices/eservices/Pages/Governmental-HealthCommunication-Center.aspx](https://www.moph.gov.qa/english/OurServices/eservices/Pages/Governmental-HealthCommunication-Center.aspx)). The raw data are protected and are not available due to data privacy laws. Aggregate data are available within the paper and its supplementary information. ## Contributors 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. HC and LJA accessed and verified all the data. PVC designed mass PCR testing to allow routine capture of variants and conducted viral genome sequencing. PT and MRH designed and conducted multiplex, RT-qPCR variant screening and viral genome sequencing. HMY and AAAT 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. ## Data sharing statement 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. The data are available under restricted access for preservation of confidentiality of patient data. Access can be obtained through a direct application for data access to Her Excellency the Minister of Public Health ([https://www.moph.gov.qa/english/OurServices/eservices/Pages/Governmental-HealthCommunication-Center.aspx](http://www.moph.gov.qa/english/OurServices/eservices/Pages/Governmental-HealthCommunication-Center.aspx)). The raw data are protected and are not available due to data privacy laws. Aggregate data are available within the paper and its supplementary information. ## Declaration of 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. ## Supplementary Appendix ### Section S1: Study population and data sources Qatar’s national and universal public healthcare system uses the Cerner-system advanced digital health platform to track all electronic health record encounters of each individual in the country, including all citizens and residents registered in the national and universal public healthcare system. Registration in the public healthcare system is mandatory for citizens and residents. The databases analyzed in this study are data-extract downloads from the Cerner-system that have been implemented on a regular weekly schedule since the onset of pandemic by the Business Intelligence Unit at Hamad Medical Corporation (HMC). HMC is the national public healthcare provider in Qatar. At every download all severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) tests, coronavirus disease 2019 (COVID-19) vaccinations, hospitalizations related to COVID-19, and all death records regardless of cause are provided to the authors through.csv files. These databases have been analyzed throughout the pandemic not only for study-related purposes, but also to provide policymakers with summary data and analytics to inform the national response. Every health encounter in the Cerner-system is linked to an individual through the HMC Number, which serves as a unique identifier that links all records for this individual at the national level. Databases were merged and analyzed using the HMC Number to link all records pertaining to testing, vaccinations, hospitalizations, and deaths. All deaths in Qatar are recorded by the public healthcare system. COVID-19-related healthcare was provided exclusively in the public healthcare system. COVID-19 vaccination was also provided only through the public healthcare system. These health records were tracked throughout the COVID-19 pandemic using the Cerner system. This system has been implemented in 2013, before the onset of the pandemic. This pre-established system ensured that we had access to comprehensive health records related to this study for both citizens and residents throughout the entire pandemic, allowing us to follow each person over time. Demographic details for every HMC Number (individual) such as sex, age, and nationality are collected upon issuing of the universal health card, based on the Qatar Identity Card, which is a mandatory requirement by the Ministry of Interior to every citizen and resident in the country. Data extraction from the Qatar Identity Card to the digital health platform is performed electronically through scanning techniques. All SARS-CoV-2 testing in any facility in Qatar is tracked nationally in one database, the national testing database. This database covers all testing throughout the country, whether in public or private facilities. Every polymerase chain reaction (PCR) test and a 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, such as the presence of clinical symptoms, contact tracing, participation in surveys or random testing campaigns, individual requests for testing, routine healthcare testing, pre-travel requirements, at the point of entry into the country, or any other relevant reasons for testing. Before November 1, 2022, SARS-CoV-2 testing in Qatar was performed extensively with about 5% of the population were tested every week [1]. Based on the distribution of the reason for testing up to November 1, 2022, most of the tests in Qatar were conducted for routine reasons, such as travel-related purposes, and about 75% of infections were diagnosed not because of presence of symptoms [1, 2]. Starting from November 1, 2022, testing for SARS-CoV-2 was substantially reduced, but still close to 1% of the population are being tested every week [2]. This study factored all SARS-CoV-2-related testing included in the national testing database over the duration of follow-up. The first omicron wave that reached its peak in January of 2022 was massive and strained the testing capacity in the country [1, 3–5]. To alleviate the burden on PCR testing, rapid antigen testing was rapidly introduced. The swift change in testing policy precluded incorporating reason for testing for a number of rapid antigen tests. While the reason for testing is documented for all PCR tests, it is not uniformly available for all rapid antigen tests. Rapid antigen test kits are accessible for purchase at pharmacies in Qatar, but results of home-based testing are neither reported nor documented in the national databases. Since SARS-CoV-2-test outcomes were 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. Qatar launched its COVID-19 vaccination program in December 2020, employing mRNA vaccines and prioritizing individuals based on coexisting conditions and age criteria [2, 6]. COVID-19 vaccination was provided free of charge, regardless of citizenship or residency status, and was nationally tracked [2, 6]. 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 [7, 8]. Further descriptions of the study population and these national databases were reported previously [1, 2, 5, 8–12]. ### Section S2: Laboratory methods and variant ascertainment #### Real-time reverse-transcription polymerase chain reaction testing Nasopharyngeal and/or oropharyngeal swabs were collected for 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 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 SARS-CoV-2 variants in Qatar is based on viral genome sequencing and multiplex RT-qPCR variant screening [13] of weekly collected random positive clinical samples [2, 14–18], complemented by deep sequencing of wastewater samples [16, 19, 20]. 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, 4, 10, 14–18, 21–26]. ### Section S3: Classification of coexisting conditions Coexisting conditions were ascertained and classified based on the ICD-10 codes for the 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. The public healthcare system provides healthcare to the entire resident population of Qatar free of charge or at heavily subsidized costs, including prescription drugs. With the mass expansion of this sector in recent years, facilities have been built to cater to specific needs of subpopulations. For example, tens of facilities have been built, including clinics and hospitals, in localities with high density of craft and manual workers [27]. All encounters for each individual were analyzed to determine the coexisting-condition classification for that individual. 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 coexisting-condition diagnosis since 2013, this person was classified with this coexisting condition. Individuals who may have coexisting conditions but never sought care in the public healthcare system were classified as individuals with no coexisting condition due to absence of recorded encounters for them. The classification of coexisting conditions spanned the following conditions: 1) Behchet’s disease, 2) cancer, 3) cardiovascular diseases, 4) infectious and parasitic diseases, 5) Chron’s disease, 6) chronic kidney disease (CKD), 7) chronic liver disease (CLD), 8) chronic lung disease, 9) congenital malformations, deformations and chromosomal abnormalities, 10) diseases of the blood and blood-forming organs and certain disorders involving the immune mechanism, 11) diseases of the ear and mastoid process, 12) deep vein thrombosis (DVT), 13) dermatitis, 14) diabetes mellitus, 15) diseases of the circulatory system, 16) diseases of the digestive system, 17) diseases of the eye and adnex, 18) diseases of the genitourinary system, 19) diseases of the musculoskeletal system and connective tissue, 20) diseases of the nervous system, 21) diseases of the respiratory system, 22) diseases of the skin and subcutaneous tissue, 23) endocrine, nutritional and metabolic diseases, 24) gingivitis, 25) hypertension, 26) injury, poisoning and certain other consequences of external causes, 27) mental and behavioral disorders, 28) neoplasms, 29 periodontitis, 30) pregnancy, childbirth and the puerperium, 31) pulmonary tuberculosis, 32) rheumatoid arthritis, 33) Sjogren’s syndrome, 34) stroke or neural conditions, 35) symptoms, signs and abnormal clinical and laboratory findings, not elsewhere classified, 36) systemic lupus erythematosus, 37) systemic sclerosis, 38) organ transplant, and 39) other unspecified factors influencing health status and contact with health services. ### Section S4: COVID-19 severity, criticality, and fatality classification Classification of COVID-19 case severity (acute-care hospitalizations) [28], criticality (intensive-care-unit hospitalizations) [28], and fatality [29] 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 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 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]. #### Fatal COVID-19 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/2024/07/29/2024.07.28.24311115/T5) Table S1. Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) checklist for cohort studies. ![Fig. S1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/07/29/2024.07.28.24311115/F3.medium.gif) [Fig. S1:](http://medrxiv.org/content/early/2024/07/29/2024.07.28.24311115/F3) Fig. S1: Flowchart describing the study population selection process for investigating indication bias or healthy vaccinee bias among recipients of primary series vaccination compared to those with no vaccination in Qatar. ![Fig. S2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/07/29/2024.07.28.24311115/F4.medium.gif) [Fig. S2:](http://medrxiv.org/content/early/2024/07/29/2024.07.28.24311115/F4) Fig. S2: Flowchart describing the study population selection process for investigating indication bias or healthy vaccinee bias among recipients of booster (third dose) vaccination compared to recipients of primary series vaccination in Qatar. ## Acknowledgments 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. Statements made herein are solely the responsibility of the authors. * Received July 28, 2024. * Revision received July 28, 2024. * Accepted July 29, 2024. * © 2024, 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].Janiaud P, Hemkens LG, Ioannidis JPA. Challenges and Lessons Learned From COVID-19 Trials: Should We Be Doing Clinical Trials Differently? Can J Cardiol. 2021;37:1353–64. 2. [2].Nelson JC, Jackson ML, Weiss NS, Jackson LA. New strategies are needed to improve the accuracy of influenza vaccine effectiveness estimates among seniors. J Clin Epidemiol. 2009;62:687–94. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jclinepi.2008.06.014&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19124221&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000266685900003&link_type=ISI) 3. [3].Simonsen L, Taylor RJ, Viboud C, Miller MA, Jackson LA. Mortality benefits of influenza vaccination in elderly people: an ongoing controversy. Lancet Infect Dis. 2007;7:658–66. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1473-3099(07)70236-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17897608&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000249818900017&link_type=ISI) 4. [4].Chambers C. Using observational epidemiology to evaluate COVID-19 vaccines: integrating traditional methods with new data sources and tools. Can J Public Health. 2021;112:867–71. 5. [5].de Waure C, Gärtner BC, Lopalco PL, Puig-Barbera J, Nguyen-Van-Tam JS. Real world evidence for public health decision-making on vaccination policies: perspectives from an expert roundtable. Expert Review of Vaccines. 2024;23:27–38. 6. [6].Remschmidt C, Wichmann O, Harder T. Frequency and impact of confounding by indication and healthy vaccinee bias in observational studies assessing influenza vaccine effectiveness: a systematic review. BMC Infect Dis. 2015;15:429. 7. [7].Simonsen L, Reichert TA, Viboud C, Blackwelder WC, Taylor RJ, Miller MA. Impact of influenza vaccination on seasonal mortality in the US elderly population. Arch Intern Med. 2005;165:265–72. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archinte.165.3.265&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15710788&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000226952400002&link_type=ISI) 8. [8].Jackson LA, Jackson ML, Nelson JC, Neuzil KM, Weiss NS. Evidence of bias in estimates of influenza vaccine effectiveness in seniors. Int J Epidemiol. 2006;35:337–44. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/ije/dyi274&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16368725&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000236817900028&link_type=ISI) 9. [9].Fürst T, Bazalová A, Fryčák T, Janošek J. Does the healthy vaccinee bias rule them all? Association of COVID-19 vaccination status and all-cause mortality from an analysis of data from 2.2 million individual health records. Int J Infect Dis. 2024;142:106976. 10. [10].Høeg TB, Duriseti R, Prasad V. Potential "Healthy Vaccinee Bias" in a Study of BNT162b2 Vaccine against Covid-19. N Engl J Med. 2023;389:284–5. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMc2306683&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=37470285&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 11. [11].Xu S, Huang R, Sy LS, Hong V, Glenn SC, Ryan DS, et al. A safety study evaluating non-COVID-19 mortality risk following COVID-19 vaccination. Vaccine. 2023;41:844–54. 12. [12].Jackson LA, Nelson JC, Benson P, Neuzil KM, Reid RJ, Psaty BM, et al. Functional status is a confounder of the association of influenza vaccine and risk of all cause mortality in seniors. Int J Epidemiol. 2006;35:345–52. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/ije/dyi275&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16368724&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000236817900029&link_type=ISI) 13. [13].Chemaitelly H, Tang P, Hasan MR, AlMukdad S, Yassine HM, Benslimane FM, et al. Waning of BNT162b2 vaccine protection against SARS-CoV-2 infection in Qatar. The New England journal of medicine. 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%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 14. [14].Altarawneh HN, Chemaitelly H, Ayoub HH, Tang P, Hasan MR, Yassine HM, et al. Effects of previous infection and vaccination on symptomatic Omicron infections. The New England journal of medicine. 2022;387:21–34. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMOA2203965&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 15. [15].Chemaitelly H, Ayoub HH, AlMukdad S, Faust JS, Tang P, Coyle P, et al. Bivalent mRNA-1273.214 vaccine effectiveness against SARS-CoV-2 omicron XBB* infections. J Travel Med. 2023;30. 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%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 17. [17].Chemaitelly H, Yassine HM, Benslimane FM, Al Khatib HA, Tang P, Hasan MR, 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. Nature medicine. 2021;27:1614-21. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 18. [18].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. 19. [19].Abu-Raddad LJ, Chemaitelly H, Ayoub HH, Al Kanaani Z, Al Khal A, Al Kuwari E, et al. Characterizing the Qatar advanced-phase SARS-CoV-2 epidemic. Scientific reports. 2021;11:6233. 20. [20].Abu-Raddad LJ, Chemaitelly H, Ayoub HH, AlMukdad S, Yassine HM, Al-Khatib HA, et al. Effect of mRNA vaccine boosters against SARS-CoV-2 Omicron infection in Qatar. The New England journal of medicine. 2022;386:1804–16. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 21. [21].Chemaitelly H, Faust JS, Krumholz HM, Ayoub HH, Tang P, Coyle P, et al. Short-and longer-term all-cause mortality among SARS-CoV-2-infected individuals and the pull-forward phenomenon in Qatar: a national cohort study. International journal of infectious diseases: IJID: official publication of the International Society for Infectious Diseases. 2023;136:81–90. 22. [22].Chemaitelly H, Ayoub HH, Tang P, Coyle P, Yassine HM, Al Thani AA, et al. Long-term COVID-19 booster effectiveness by infection history and clinical vulnerability and immune imprinting: a retrospective population-based cohort study. The Lancet infectious diseases. 2023;23:816–27. 23. [23].AlNuaimi AA, Chemaitelly H, Semaan S, AlMukdad S, Al-Kanaani Z, Kaleeckal AH, et al. All-cause and COVID-19 mortality in Qatar during the COVID-19 pandemic. BMJ Glob Health. 2023;8. 24. [24].Abu-Raddad LJ, Chemaitelly H, Ayoub HH, Yassine HM, Benslimane FM, Al Khatib HA, et al. Association of prior SARS-CoV-2 infection with risk of breakthrough infection following mRNA vaccination in Qatar. Jama. 2021;326:1930–9. 25. [25].Chemaitelly H, AlMukdad S, Ayoub HH, Altarawneh HN, Coyle P, Tang P, et al. Covid-19 vaccine protection among children and adolescents in Qatar. The New England journal of medicine. 2022;387:1865–76. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2210058&link_type=DOI) 26. [26].Chemaitelly H, Ayoub HH, AlMukdad S, Coyle P, Tang P, Yassine HM, et al. Protection from previous natural infection compared with mRNA vaccination against SARS-CoV-2 infection and severe COVID-19 in Qatar: a retrospective cohort study. The Lancet Microbe. 2022;3:e944–e55. 27. [27].Chemaitelly H, Ayoub HH, Tang P, Coyle PV, Yassine HM, Al Thani AA, et al. History of primary-series and booster vaccination and protection against Omicron reinfection. Science advances. 2023;9:eadh0761. 28. [28].Mahmoud MA, Ayoub HH, Coyle P, Tang P, Hasan MR, Yassine HM, et al. SARS-CoV-2 infection and effects of age, sex, comorbidity, and vaccination among older individuals: A national cohort study. Influenza and other respiratory viruses. 2023;17:e13224. 29. [29].World Health Organization (WHO). Living guidance for clinical management of COVID-19. Aavailable from: [https://www.who.int/publications/i/item/WHO-2019-nCoV-clinical-2021-2](https://www.who.int/publications/i/item/WHO-2019-nCoV-clinical-2021-2). Accessed on: February 27, 2023. 2023. 30. [30].World Health Organization (WHO). International Guidelines for Certification and Classification (Coding) of COVID-19 as Cause of Death. Available from: [https://www.who.int/publications/m/item/international-guidelines-for-certification-and-classification-(coding)-of-covid-19-as-cause-of-death](https://www.who.int/publications/m/item/international-guidelines-for-certification-and-classification-(coding)-of-covid-19-as-cause-of-death). Accessed on: February 27, 2023. 2023. 31. [31].Al-Aly Z, Bowe B, Xie Y. Long COVID after breakthrough SARS-CoV-2 infection. Nat Med. 2022;28:1461–7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-022-01840-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 32. [32].Nguyen KH, Yankey D, Coy KC, Brookmeyer KA, Abad N, Guerin R, et al. COVID-19 Vaccination Coverage, Intent, Knowledge, Attitudes, and Beliefs among Essential Workers, United States. Emerg Infect Dis. 2021;27:2908–13. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3201/eid2711.211557&link_type=DOI) 33. [33].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. 34. [34].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) 35. [35].Chemaitelly H, Ayoub HH, Tang P, Yassine HM, Al Thani AA, Hasan MR, et al. Addressing bias in the definition of SARS-CoV-2 reinfection: implications for underestimation. Frontiers in medicine. 2024;11:1363045. 36. [36].Ayoub HH, Chemaitelly H, Seedat S, Makhoul M, Al Kanaani Z, Al Khal A, 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. 37. [37].Coyle PV, Chemaitelly H, Ben Hadj Kacem MA, Abdulla Al Molawi NH, El Kahlout RA, Gilliani I, 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. 38. [38].Jeremijenko A, Chemaitelly H, Ayoub HH, Alishaq M, Abou-Samra AB, Al Ajmi J, et al. Herd immunity against severe acute respiratory syndrome coronavirus 2 infection in 10 communities, Qatar. Emerging infectious diseases. 2021;27:1343–52. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3201/eid2705.204365&link_type=DOI) 39. [39].Al-Thani MH, Farag E, Bertollini R, Al Romaihi HE, Abdeen S, Abdelkarim A, 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) 40. [40].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. 41. [41].Chemaitelly H, Tang P, Coyle P, Yassine HM, Al-Khatib HA, Smatti MK, et al. Protection against Reinfection with the Omicron BA.2.75 Subvariant. N Engl J Med. 2023;388:665-7. 42. [42].Chemaitelly H, Ayoub HH, Coyle P, Tang P, Yassine HM, Al-Khatib HA, et al. Protection of Omicron sub-lineage infection against reinfection with another Omicron sub-lineage. Nature communications. 2022;13:4675. 43. [43].Barda N, Dagan N, Cohen C, Hernan MA, Lipsitch M, Kohane IS, 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%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 44. [44].Austin PC. Using the Standardized Difference to Compare the Prevalence of a Binary Variable Between Two Groups in Observational Research. Commun Stat-Simul C. 2009;38:1228–34. 45. [45].Sjolander A, Greenland S. Ignoring the matching variables in cohort studies-when is it valid and why? Stat Med. 2013;32:4696–708. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/sim.5879&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23761197&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 46. [46].Stensrud MJ, Hernan MA. Why Test for Proportional Hazards? JAMA. 2020;323:1401–2. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2020.1267&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 47. [47].Tseng HF, Ackerson BK, Bruxvoort KJ, Sy LS, Tubert JE, Lee GS, et al. Effectiveness of mRNA-1273 against infection and COVID-19 hospitalization with SARS-CoV-2 Omicron subvariants: BA.1, BA.2, BA.2.12.1, BA.4, and BA.5. medRxiv. 2022:2022.09.30.22280573. 48. [48].Benn CS, Netea MG, Selin LK, Aaby P. A small jab-a big effect: nonspecific immunomodulation by vaccines. Trends Immunol. 2013;34:431–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.it.2013.04.004&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23680130&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000324971600002&link_type=ISI) 49. [49].Netea MG, Dominguez-Andres J, Barreiro LB, Chavakis T, Divangahi M, Fuchs E, et al. Defining trained immunity and its role in health and disease. Nat Rev Immunol. 2020;20:375–88. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41577-020-0285-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 50. [50].Tayar E, Abdeen S, Abed Alah M, Chemaitelly H, Bougmiza I, Ayoub HH, et al. Effectiveness of influenza vaccination against SARS-CoV-2 infection among healthcare workers in Qatar. Journal of infection and public health. 2023;16:250–6. 51. [51].Feikin DR, Higdon MM, Abu-Raddad LJ, Andrews N, Araos R, Goldberg Y, et al. Duration of effectiveness of vaccines against SARS-CoV-2 infection and COVID-19 disease: results of a systematic review and meta-regression. Lancet. 2022;399:924–44. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(22)00152-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 52. [52].Chemaitelly H, Abu-Raddad LJ. Waning effectiveness of COVID-19 vaccines. Lancet. 2022;399:771–3. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(22)00277-X&link_type=DOI) 53. [53].Islam N, Shkolnikov VM, Acosta RJ, Klimkin I, Kawachi I, Irizarry RA, et al. Excess deaths associated with covid-19 pandemic in 2020: age and sex disaggregated time series analysis in 29 high income countries. BMJ. 2021;373:n1137. 54. [54].Kontis V, Bennett JE, Rashid T, Parks RM, Pearson-Stuttard J, Guillot M, et al. Magnitude, demographics and dynamics of the effect of the first wave of the COVID-19 pandemic on all-cause mortality in 21 industrialized countries. Nat Med. 2020;26:1919–28. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-020-1112-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 55. [55].Johns Hopkins Coronavirus Resource. Mortality Analyses. [https://coronavirus.jhu.edu/data/mortality](https://coronavirus.jhu.edu/data/mortality). Accessed on October 2,. 2022. 56. [56].Seedat S, Chemaitelly H, Ayoub HH, Makhoul M, Mumtaz GR, Al Kanaani Z, et al. SARS-CoV-2 infection hospitalization, severity, criticality, and fatality rates in Qatar. Scientific reports. 2021;11:18182. 57. [57].Abu-Raddad LJ, Chemaitelly H, Yassine HM, Benslimane FM, Al Khatib HA, Tang P, et al. Pfizer-BioNTech mRNA BNT162b2 Covid-19 vaccine protection against variants of concern after one versus two doses. Journal of travel medicine. 2021;28. ## References 1. [1].Altarawneh HN, Chemaitelly H, Ayoub HH, Tang P, Hasan MR, Yassine HM, et al. Effects of previous infection and vaccination on symptomatic Omicron infections. The New England journal of medicine. 2022;387:21–34. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMOA2203965&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 2. [2].Chemaitelly H, Tang P, Hasan MR, AlMukdad S, Yassine HM, Benslimane FM, et al. Waning of BNT162b2 vaccine protection against SARS-CoV-2 infection in Qatar. The New England journal of medicine. 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%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 3. [3].Chemaitelly H, Ayoub HH, AlMukdad S, Faust JS, Tang P, Coyle P, et al. Bivalent mRNA-1273.214 vaccine effectiveness against SARS-CoV-2 omicron XBB* infections. J Travel Med. 2023;30. 4. [4].Altarawneh HN, Chemaitelly H, Hasan MR, Ayoub HH, Qassim S, AlMukdad S, et al. Protection against the Omicron variant from previous SARS-CoV-2 infection. The New England journal of medicine. 2022;386:1288–90. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 5. [5].Chemaitelly H, Ayoub HH, Tang P, Coyle P, Yassine HM, Al Thani AA, et al. Long-term COVID-19 booster effectiveness by infection history and clinical vulnerability and immune imprinting: a retrospective population-based cohort study. The Lancet infectious diseases. 2023;23:816–27. 6. [6].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. 7. [7].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. 8. [8].Abu-Raddad LJ, Chemaitelly H, Ayoub HH, Al Kanaani Z, Al Khal A, Al Kuwari E, et al. Characterizing the Qatar advanced-phase SARS-CoV-2 epidemic. Scientific reports. 2021;11:6233. 9. [9].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) 10. [10].Abu-Raddad LJ, Chemaitelly H, Ayoub HH, AlMukdad S, Yassine HM, Al-Khatib HA, et al. Effect of mRNA vaccine boosters against SARS-CoV-2 Omicron infection in Qatar. The New England journal of medicine. 2022;386:1804–16. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 11. [11].Chemaitelly H, Faust JS, Krumholz HM, Ayoub HH, Tang P, Coyle P, et al. Short-and longer-term all-cause mortality among SARS-CoV-2-infected individuals and the pull-forward phenomenon in Qatar: a national cohort study. International journal of infectious diseases: IJID: official publication of the International Society for Infectious Diseases. 2023;136:81–90. 12. [12].AlNuaimi AA, Chemaitelly H, Semaan S, AlMukdad S, Al-Kanaani Z, Kaleeckal AH, et al. All-cause and COVID-19 mortality in Qatar during the COVID-19 pandemic. BMJ Glob Health. 2023;8. 13. [13].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. doi:10.17504/protocols.io.br9vm966. 2021. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.17504/protocols.io.br9vm966&link_type=DOI) 14. [14].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%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 15. [15].Chemaitelly H, Yassine HM, Benslimane FM, Al Khatib HA, Tang P, Hasan MR, 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. Nature medicine. 2021;27:1614-21. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 16. [16].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. 17. [17].Benslimane FM, Al Khatib HA, Al-Jamal O, Albatesh D, Boughattas S, Ahmed AA, et al. One year of SARS-CoV-2: genomic characterization of COVID-19 outbreak in Qatar. Frontiers in cellular and infection microbiology. 2021;11:768883. 18. [18].Hasan MR, Kalikiri MKR, Mirza F, Sundararaju S, Sharma A, Xaba T, 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) 19. [19].Saththasivam J, El-Malah SS, Gomez TA, Jabbar KA, Remanan R, Krishnankutty AK, et al. COVID-19 (SARS-CoV-2) outbreak monitoring using wastewater-based epidemiology in Qatar. Sci Total Environ. 2021;774:145608. 20. [20].El-Malah SS, Saththasivam J, Jabbar KA, K KA, Gomez TA, Ahmed AA, 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. 21. [21].Tang P, Hasan MR, Chemaitelly H, Yassine HM, Benslimane FM, Al Khatib HA, et al. BNT162b2 and mRNA-1273 COVID-19 vaccine effectiveness against the SARS-CoV-2 Delta variant in Qatar. Nature medicine. 2021;27:2136–43. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F29%2F2024.07.28.24311115.atom) 22. [22].Chemaitelly H, Ayoub HH, AlMukdad S, Coyle P, Tang P, Yassine HM, et al. Duration of mRNA vaccine protection against SARS-CoV-2 Omicron BA.1 and BA.2 subvariants in Qatar. Nature communications. 2022;13:3082. 23. [23].Qassim SH, Chemaitelly H, Ayoub HH, AlMukdad S, Tang P, Hasan MR, 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;29. 24. [24].Altarawneh HN, Chemaitelly H, Ayoub HH, Hasan MR, Coyle P, Yassine HM, et al. Protective effect of previous SARS-CoV-2 infection against Omicron BA.4 and BA.5 subvariants. The New England journal of medicine. 2022;387:1620-2. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMc2209306&link_type=DOI) 25. [25].Chemaitelly H, Tang P, Coyle P, Yassine HM, Al-Khatib HA, Smatti MK, et al. Protection against Reinfection with the Omicron BA.2.75 Subvariant. N Engl J Med. 2023;388:665-7. 26. [26].Chemaitelly H, Coyle P, Kacem MAB, Ayoub HH, Tang P, Hasan MR, et al. Protection of natural infection against reinfection with SARS-CoV-2 JN.1 variant. medRxiv. 2024:2024.02.22.24303193. 27. [27].Al-Thani MH, Farag E, Bertollini R, Al Romaihi HE, Abdeen S, Abdelkarim A, 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. 28. [28].World Health Organization (WHO). Living guidance for clinical management of COVID-19. Aavailable from: [https://www.who.int/publications/i/item/WHO-2019-nCoV-clinical-2021-2](https://www.who.int/publications/i/item/WHO-2019-nCoV-clinical-2021-2). Accessed on: February 27, 2023. 2021. 29. [29].World Health Organization (WHO). International Guidelines for Certification and Classification (Coding) of COVID-19 as Cause of Death. Available from: [https://www.who.int/publications/m/item/international-guidelines-for-certification-and-classification-(coding)-of-covid-19-as-cause-of-death](https://www.who.int/publications/m/item/international-guidelines-for-certification-and-classification-(coding)-of-covid-19-as-cause-of-death). Accessed on: February 27, 2023. 2020.