Rapid review on protective immunity post COVID-19 vaccination: update 3
October 2021
- Download in PDF format (1.79 MB, 73 pages)
Table of contents
- Introduction
- Key points
- Overview of the evidence
- Methods
- Evidence tables
- Table 1: Randomized controlled trials, prospective cohort and case control studies evaluating vaccine breakthrough infections (n=42)
- Table 2: Immune responses ≥6 months after the primary series of COVID-19 vaccination in individuals with no history of prior COVID-19 (n=26)
- Table 3: Immune responses in ≥6 months after primary series of COVID-19 vaccination in individuals with a history of COVID-19 (n=10)
- Table 4: Systematic and rapid reviews relevant to vaccinated immunity (n=3)
- References
Introduction
What do we know about protective immunity acquired from vaccination evidenced by breakthrough infections and markers of immunity ≥6 months post vaccination?
Understanding the extent and limits of protective immunity against COVID-19 has important implications for the COVID-19 pandemic and response. Immunity arising from infection with coronaviruses in general varies tremendously, from a few months for the seasonal coronaviruses associated with the common cold, to 2-3 years for the emerging coronaviruses such as SARS-CoV-1 and MERSFootnote 1. For SARS-CoV-2 (COVID-19), it is known that most people develop immune responses after receiving a full primary vaccination series (2 doses unless the primary series is a 1 dose vaccine e.g. Janssen), however, for how long and to what extent immune responses protect individuals from infection is not yet clear.
Previous versions of this report from February, April and August 2021 summarized the evidence on protective immunity post infection and post vaccination together and can be requested through ocsoevidence-bcscdonneesprobantes@phac-apsc.gc.ca. Due to the expanding evidence base, reviews on protective immunity post infection and post vaccination have been done separately in update 3 (Oct 2021) and will be done separately for subsequent updates. A separate review has been completed to look at protective immunity from SARS-CoV-2 infection, including the evidence on reinfection and correlates of long-term immunity post-infection (≥12 months). The current review addresses protective immunity from the primary series of vaccines, including the evidence on breakthrough infections and correlates of long-term immunity (≥6 months) from vaccination in individuals with or without a history of prior infection.
With the number of partially and fully vaccinated individuals increasing around the world, real-world data and research on infections is starting to emerge. There is some heterogeneity across studies in how “fully-vaccinated” and “breakthrough infection” are defined. In this review, articles were included if they reported data based on the CDC case definition of breakthrough infection: a person has SARS-CoV-2 RNA or antigen detected on a respiratory specimen ≥14 days after completing the primary series of a COVID-19 vaccine (e.g., two weeks post second dose of a two-dose vaccine or two weeks post first dose of a one-dose vaccine)Footnote 2. Since the last update of this review, research on third doses and “booster doses” have been accumulating. As such, this review also captured data on breakthrough infections following third doses or boosters following the primary series.
There are also challenges in assessing long-term immunity against COVID-19 post-vaccination. This arises because immune responses are variable, not everyone vaccinated for COVID-19 develops detectable antibody levels and not all people with antibodies specific to SARS-CoV-2 antigens mount sufficient protective immunity. Evidence suggests that both neutralizing antibodies, B-cell (i.e., immune cells that produce virus targeting antibodies) and memory T-cell (i.e., immune cells that guide the cell mediated adaptive immune responses) activity specific to SARS-CoV-2 are currently the best indicators of protective immunity. However, the variation and interplay of antibodies, B and T-cell responses to infection and/or vaccination, as well as the variety of detection techniques complicates the assessment of long-term immunity.
This rapid review summarizes the evidence from recent studies on breakthrough infection post-vaccination, persistence of antibodies and other immune markers for ≥6 months following vaccination published before October 22, 2021. Due to the abundance of human data, animal models of disease and in vitro studies were not included.
Key points
There were 42 studies on breakthrough infection and 36 on the kinetics and durability of antibodies and other immunity markers at ≥6 months post-vaccination. The review is divided into two sections and two populations including breakthrough infections in people that were vaccinated with (n=3) and without (n=41) history of prior SARS-CoV-2 infection. As well as studies that capture immune response markers ≥6 months post full vaccination in people with (n=10) and without (n=26) history of prior infection. No studies captured evidence on immune correlates ≥6 months after a breakthrough infection following vaccination.
Breakthrough infections post primary series COVID-19 vaccination
Forty-two studies including randomized controlled trials, prospective cohorts and case control studies were summarized to estimate total number of breakthrough infections (i.e., asymptomatic and symptomatic) following a full primary vaccine series, Table 1:
- Breakthrough infections confirmed by RT-PCR among people with no history of COVID-19 occurred at variable rates (0.2-6.6%) across prospective studies conducted December 2020-August 2021 following full vaccination with the Comirnaty (Pfizer), Spikevax (Moderna), or Vaxzevria (AstraZeneca) vaccinesFootnote 3Footnote 4Footnote 5Footnote 6Footnote 7Footnote 8. Studies that included infections determined via antigen detection reported higher breakthrough rates (8.6-26%)Footnote 9Footnote 10.
- Vaccine effectiveness (VE) against overall infection were similar for original variants and Alpha (B.1.1.7)Footnote 11Footnote 12Footnote 13Footnote 14Footnote 15Footnote 16Footnote 17 and Beta (B.1.351)Footnote 12Footnote 14Footnote 17Footnote 18. Recent data has suggested that there are more breakthrough infections due to Delta (B.1.617.2) compared to other variants of concern (VOC) and the original variantFootnote 9Footnote 15Footnote 16Footnote 19Footnote 20Footnote 21Footnote 22Footnote 23Footnote 24. VE estimates against Delta infections were lower compared to Alpha and higher for mRNA vaccines (Comirnaty/Spikevax: 66-79% Delta vs. 90-92% Alpha) than viral-vector based vaccines (Vaxzevria/Janssen: 51-67% Delta vs. 72-79% Alpha)Footnote 5Footnote 16Footnote 25Footnote 26. For more information on VE estimates against VOCs please refer to the living review maintained by COVID-ENDFootnote 27.
- Vaccinated individuals with a history of SARS-CoV-2 infection prior to vaccination had higher levels of protection against breakthrough infection compared to vaccinated individuals without a prior infectionFootnote 11Footnote 25Footnote 28.
- There was a lot of variability in the risk of breakthrough infection between studies which are likely due to a combination of factors, such as what SARS-CoV-2 variants are circulating, stage of the epidemic within the study area, the level of immunity in the study population, vaccine effectiveness, type of vaccine, administration protocol (e.g., interval between doses) and length of time since vaccination.
Immune response markers ≥6 months post full vaccination in individuals not previously infected
Twenty-six studies reported on circulating antibodies (n=26) or cellular immune activity (n=4) at 6-8 months post vaccination in participants that did not contract COVID-19 prior to vaccination, Table 2.
Four studies on Comirnaty (Pfizer) and one study on Spikevax (Moderna) reported memory B-cell or T-cell responses at 6 months following a completed primary series of COVID-19 vaccination:
- Elevated T-cell (n= 3 studies) or B-cell (n= 2 studies) responses were detected in the majority of individuals at 6 months (42%-90% for T-cells)Footnote 29Footnote 30. Longitudinal sampling in some studies showed responses were still increasing and diversifying from earlier time pointsFootnote 29Footnote 30Footnote 31 or were reported as being maintained at 6 monthsFootnote 29.
- Two studies indicated decreasing CD8+ T-cells and increasing CD4+ T-cells at 6 months, which indicates development of immune memory B-cellsFootnote 29Footnote 30.
- One out of two studies that measured correlations at 6 months between T-cell levels and presence of antibodies showed a correlationFootnote 29, while the other did not find any correlationsFootnote 30.
Twenty-six studies reported on circulating antibodies 6-9 months post primary vaccine series in people who had not had COVID-19 infection. The majority of studies reported that antibody levels had declined from peak, but were detectable for Spike (S) IgG and Receptor Binding Domain (RBD) IgG and neutralizing antibodies (NAb). There is some evidence that antibody titers are correlated with protective immunity, but the exact level of protection is uncertainFootnote 32Footnote 33:
- Studies comparing Comirnaty and Spikevax find that Spikevax recipients had higher initial antibody titers with slower declines up to 6 month post vaccination samplesFootnote 34Footnote 35.
- NAb titers were maintained in the majority of individuals in the 6-9 month post vaccination samples (57%-100%)Footnote 29Footnote 36Footnote 37Footnote 38 and S-IgG or RBD-IgG was positively correlated with NAbsFootnote 29Footnote 39Footnote 40.
- NAb titers among nursing home residents who received Comirnaty were notably lower (30%) compared to NAb titres among 84% of the healthcare workers (HCWs) working in the nursing home who also received ComirnatyFootnote 38.
- Immunocompromised individuals had slightly lower seropositivity or titers than non-immunocompromised controls (79-90.2% vs 84-100% respectively) 6 months post a two dose primary series of vaccination with ComirnatyFootnote 40Footnote 41Footnote 42Footnote 43. These populations included patients with cancer, dialysis, and multiple sclerosis with anti-S IgG seropositivity 6 months after vaccination at 90.2% for chronic lymphocytic leukemia (CLL) /small lymphocytic lymphoma (SLL) vaccinated with ComirnatyFootnote 42, 79% for patients with cancer receiving ComirnatyFootnote 43, and 56.1%-85.9% for patients on dialysis receiving either Comirnaty, Spikevax, or JanssenFootnote 35.
- Individual factors associated with seropositivity or NAb titers included a negative correlation of antibody titers with increasing age, but in most cases antibodies were still detectable at 6 months in individuals vaccinated with Comirnaty (n=4 studies), Spikevax (n= 2), and Jansen (n=1)Footnote 36Footnote 37Footnote 38Footnote 40Footnote 44Footnote 45Footnote 46Footnote 47. One study reported no difference (less than 10% difference in S-IgG levels) between those over 60 and under 60 years of age who received ComirnatyFootnote 48.
Immune response markers in previously infected individuals ≥6 months post primary series of COVID-19 vaccinations)
Ten studies reported on immune response markers in previously infected individuals greater or equal to six month post primary series of COVID-19 vaccinations, Table 3. Cellular immune markers in vaccinated individuals who did not have COVID-19 infection history compared to vaccinated individuals who were previously infected showed the latter maintained their T-cell levels better in two studies and the frequency of B-cell populations or decay rates for T-cells was found to be the same in another studyFootnote 29Footnote 30Footnote 39.
Antibody S-IgG and RBD-IgG titers) and NAb titers were generally higher in vaccinated individuals who were previously infected compared to individuals without history of prior infectionFootnote 38Footnote 39Footnote 47Footnote 49:
- NAb titers were similar in vaccinated people with (81-100%) and without history of prior infection (57-100%)Footnote 29Footnote 36Footnote 37Footnote 38.
- In three studies, at 6-7 months post vaccination, previously infected vaccinated individuals had a smaller decline in spike specific antigens and NAbs than those vaccinated who did not have a history of prior infectionFootnote 39Footnote 47Footnote 50, while four studies reported no difference or higher antibody decay rates among those previously infected then vaccinatedFootnote 29Footnote 34Footnote 38Footnote 46.
The specific relationship between the correlates of immunity and protection against SARS-CoV-2 infection is not fully understood and additional data is needed to fill knowledge gaps.
Overview of the evidence
Breakthrough infection studies: Only data in which breakthrough infections occurred ≥14 days after completing the primary series of a COVID-19 vaccine were included in this review (as per the CDC definition of breakthrough infection). This review focuses on the highest level of evidence: randomized controlled trials, prospective cohort studies and case control studies. Double-blind placebo-controlled trials are the gold standard for measuring the impact of an intervention, but do not necessarily provide an accurate estimate of how effective the vaccination will be in the real-world, nor are they as likely to occur during an ongoing pandemic scenario. Observational studies provide a real-world assessment of an intervention, but may also be at risk of more biases. These include the retrospective nature of case control studies and reliance on self-reported symptoms in many cohort studies. In cohort studies, people who get vaccinated may differ in health seeking behaviour (i.e., getting tested for SARS-CoV-2) than people who do not get vaccinated. Using a test-negative case control design can help minimize this type of bias, as both groups are seeking testing. Prospective cohort design in which participants are tested on a longitudinal basis also helps to minimize bias. Retrospective cohorts of medical record data or routinely collected surveillance data on COVID-19 were excluded from this review so the review could focus on studies with a lower risk of bias.
Long-term immunity studies mainly include longitudinal evidence from observational studies, particularly of prospective cohort, large case series and cross-sectional design, which are at moderate to high risk of selection biases and confounding factors. For example, most studies reported clinical infection severity among study participants, but many did not analyse or control for risk factors such as age, that may explain some of the heterogeneity in correlates of immunity. Differences in study participant demographics, baseline immune status, length of time from infection to vaccination, clinical severity of infections, investigated immune outcomes, follow-up time and measurement methods likely contributed to some of the observed heterogeneity. Variability may have come from the application of different antibody and immune cell detection methods with different test sensitivity and specificity parameters. All of these factors make it difficult to compare results across studiesFootnote 51.
Knowledge gaps:
- Lack of understanding of how strong the correlation is between markers of immunity (e.g., neutralizing antibody titers) and protection from infection or severe disease and how vaccine mediated protection may be different than protection post infection.
- The role of specific antibodies, B-cells and T-cells in the preventing or clearing infection have not been definitively identified in humans.
- The majority of evidence on protective immunity against VOCs is on Alpha. Prospective studies are needed on the other emerging VOCs, particularly Delta and Omicron.
- Further evidence is required on waning immunity over time for all combinations of vaccinated and/or previously infected individuals, as well as what demographic variables and other risk factors may impact waning immunity.
Breakthrough infections post full vaccination
Although COVID-19 vaccines have been shown to be very effective at preventing severe disease, some vaccine breakthrough cases are expected. Evidence on the extent of vaccine protection against infection or severe illness from various VOCs is rapidly emerging and our understanding of vaccine immunity is evolving. The definition for breakthrough infection used in this review was based on the CDC case definition for breakthrough infectionFootnote 2: a person who has SARS-CoV-2 RNA or antigen detected on a respiratory specimen collected ≥14 days after completing the primary series of a COVID-19 vaccine (e.g., two weeks post second dose of a two-dose vaccine or two weeks post second dose of a one-dose vaccine). Studies that only included estimates of symptomatic or severe infection following vaccination, rather than the total number of breakthrough infections (i.e., asymptomatic and symptomatic) were excluded. High level points are listed below and detailed outcomes for each study are located in Table 1.
Vaccines used in the included studies on breakthrough infections included Comirnaty (Pfizer-BioNTech/ BNT162b2), Spikevax (Moderna/mRNA-1273), Vaxzevria (AstraZeneca/Covishield/ChAdOx1-S/AZD1222), Janssen (Johnson & Johnson/Ad26.COV2.S), and Bharat Biotech (Covaxin /BBV152)).
Five studies detailing findings from randomized controlled trials of vaccine efficacy reported that breakthrough infection within 0.5-6 months following two doses of vaccine is relatively low. The primary endpoints for many RCTs were symptomatic COVID-19 cases at least one week after 2nd dose; thus, did not meet our inclusion criteria:
- Spikevax (Moderna): In a US RCT with 5 months follow-up, infections were recorded in 2% of vaccinated individuals compared to 9.5% of unvaccinated (placebo) individualsFootnote 52. Efficacy of the Spikevax vaccine against infection was 82.0% (95%CI 79.5-84.2) and did not wane over a 5 month period following the second doseFootnote 52.
- Vaxzevria (Astra Zeneca): In a UK RCT with up to 6 months follow-up, infections were recorded in 4% of vaccinated individuals compared to 8% of controls (who were vaccinated with the meningococcal vaccine MenACWY)Footnote 53Footnote 54Footnote 55. The vaccine efficacy ranged from 73.5% (95%CI 55.5-84.2) in an early analysis of the trial data (May-Nov 2020) to 50.9% (95%CI 41.0-59.0) in a later analysis of the trial data (Oct 2020-Jan 2021)Footnote 53Footnote 54Footnote 55.
- BBV152 (Covaxin): In a trial from India with a 2-month follow-up period, infections occurred in 0.2% of the vaccine group and 0.9% or the placebo groupFootnote 56. Efficacy against asymptomatic infection, symptomatic infection, and Delta infection was 63.6% (95%CI 29.0–82.4), 77.8% (95%CI 65.2–86.4), and 65.2% (95%CI 33.1–83.0), respectivelyFootnote 56.
Thirty-seven observational studies of real-world vaccine effectiveness (VE) also demonstrate that breakthrough infection from original variants and the Alpha variant following two doses of vaccine is low during short term follow-up (0.5 -7 months). However, some VOCs are showing more concerning trends:
- Overall, unvaccinated subjects were at a significantly higher risk of developing infection as compared to fully vaccinated subjects (Vaxzevria RR=2.5; Vaxzevria and Comirnaty OR=2.7-10.9; Comirnaty HR=9.1)Footnote 3Footnote 8Footnote 9Footnote 19Footnote 26Footnote 57Footnote 58.
- Breakthrough infections were low following vaccination with the Comirnaty, Spikevax, and Vaxzevria vaccines.
- Five prospective cohort studies of HCWs, with follow-up ranging from 2-6 months post vaccination with Comirnaty or Spikevax, reported that the incidence of RT-PCR confirmed infection was lower in the vaccinated HCWs (0.2-6.3% across studies) compared to the unvaccinated HCWs (2.2-7.5% across studies)Footnote 3Footnote 4Footnote 5Footnote 6Footnote 7. For Vaxzevria in India, the incidence was 6.6% (vaccinated HCWs) vs. 43.8% (unvaccinated HCWs)Footnote 8.
- Estimates of breakthrough infection were higher in studies that identified infections by the detection of SARS-CoV-2 antigen in a respiratory specimen. One prospective cohort study of HCWs in the US conducted between Mar-Aug 2021 found a high proportion of Comirnaty breakthrough infections (detected by SARS-CoV-2 nucleocapsid seroconversion) at 26.0% (59/227), similar to the unvaccinated HCWs at 23.5% (4/17)Footnote 10. Another study (conducted between Jan-May 2021 in India), which characterized vaccine breakthroughs by either the detection of SARS-CoV-2 RNA or antigen, reported infection in 8.6% of fully vaccinated HCWs (Vaxzevria) vs. 21.5% of unvaccinated HCWsFootnote 9.
- Among HCWs in a prospective cohort, post infection immunity (85%) and post vaccination immunity (90%, Comirnaty or Vaxzevria 15-42 days post second dose) offered similar protection compared to the incidence of infections in the unvaccinated and not previously infected groupFootnote 11. Another cohort of HCWs found that infection after vaccination was rare (1.1% of HCWs, 20/1818) and significantly less frequent compared to reinfection after initial infection (9.5%, 8/84)Footnote 59. In a UK general population study up to August 2021 there was no evidence that effectiveness of Vaxzevria was different than protection afforded by previous infection without vaccination (p=0.33), but protection from vaccination with Comirnaty was greater than protection from infection (p=0.04), and those previously infected and then vaccinated had the highest protection against new infection (p<0.006)Footnote 25.
- Comirnaty, Spikevax, and Vaxzevria VE against infection in the US, UK, and Israel were ≥80% between Dec 2020-Apr 2021Footnote 3Footnote 5Footnote 6Footnote 11Footnote 15Footnote 25Footnote 60Footnote 61Footnote 62. However, lower vaccine effectiveness of these vaccines has been reported in more recent studies: Comirnaty and Spikevax in the US was 66-74% in July/August 2021, Janssen in the US was 51% in July/August 2021, Comirnaty and Vazevria VE in the UK was 67-80% in May 2021, Spikevax and Vaxzevria VE in Spain was 66% Jan-Apr 2021, Comirnaty and Spikevax VE in Qatar was 54-87% Dec 2020-Jul 2021, Vaxzevria and Covaxin VE in India was 54-83% Jan-Jun 2021, and Coronavac/Biotec VE in China was 59% between May-Jun 2021Footnote 5Footnote 19Footnote 23Footnote 25Footnote 26Footnote 58Footnote 63Footnote 64Footnote 65Footnote 66. This variability in VE may be related to multiple factors, such as stage of epidemic within the study area, level of immunity in the study population, administration protocols (e.g., interval between doses), length of time since vaccination, and an increase/change in VOCs.
- Preliminary evidence indicates that VE against infection is similar for original variants and AlphaFootnote 11Footnote 12Footnote 13Footnote 14Footnote 15Footnote 16Footnote 17. VE against Beta may be slightly less compared to Alpha, but the difference was not statistically significantFootnote 12Footnote 14Footnote 17Footnote 18. A study from the US (Feb-Apr 2021) found that VOCs were overrepresented in mRNA breakthrough cases compared to cases in the unvaccinated populationFootnote 67. The frequency of all VOCs in breakthrough cases increased by 1.47-fold compared with that of SARS-CoV-2 sequences taken from a general sample of infections in the same areaFootnote 67.
- Genomic analysis has revealed a rise of Delta variant in breakthrough infections since Apr 2021Footnote 9Footnote 15Footnote 19Footnote 20Footnote 21Footnote 22 and studies have shown reduced vaccine effectiveness against the Delta variant compared to other variants; however, these results may be affected by increasing Delta cases, easing of public health restrictions, and increased time since primary series of vaccines during the analysis periods.
- There was a higher risk of Delta breakthrough infection compared to Alpha in a study from Portugal (aOR=1.96, 95%CI 1.2-3.1)Footnote 21. VE estimates against Delta infection were lower compared to Alpha in studies from Scotland (Comirnaty: 79% vs. 92%; Vaxzevria: 60% vs. 72%)Footnote 16 and the US (Comirnaty/Spikevax: 66%% vs. ~90%)Footnote 5. One longitudinal household study from the UK, found that VE when Delta was dominant was not significantly different than when Alpha was dominant (Comirnaty: 80% vs. 78%; Vaxzevria: 67% vs. 79%)Footnote 25.
- An increase in breakthrough infections has been particularly evident in QatarFootnote 23Footnote 24. The percentage of all daily diagnosed SARS-CoV-2 infections that were vaccine breakthroughs (Comirnaty and Spikevax) increased over time and reached 36.4% in September 2021Footnote 24. VE against infection declined to 20% 5-7 months after second dose of Comirnaty or SpikevaxFootnote 24. The dominant variant during the entire study period was Beta, but a similar pattern of waning of protection was also demonstrated for the recently dominant Delta variant.
- Evidence of waning immunity was modest in the US, with Delta VE decreasing from 94.1% (90.5-96.3%) 14-60 days after vaccination to 80% (70.2-86.6%) 151-180 days after vaccinationFootnote 15.
Previously infected individuals that received a full primary series of vaccines have high levels of protection (n=3 studies):
- Vaccinating previously infected HCWs (96% reduction in incidence compared to the naïve group) was not significantly more protective than those fully vaccinated (90%) 15-42 days post vaccination or previously infected (85%)Footnote 11.
- A case control study of the general population in Kentucky found that previously infected individuals who were not vaccinated had 2.34 times the odds of reinfection compared with those who were fully vaccinated with Janssen, Comirnaty, or SpikevaxFootnote 28.
- In a large longitudinal study of households in the UK, protection against infection was significantly higher for vaccinated individuals with prior infection than vaccinated individuals without prior infection (Comirnaty p=0.006 / Vaxzevria p<0.0001)Footnote 25.
- While not included in-depth in this review as they did not meet the specified inclusion criteria, multiple studies have also found that vaccinated individuals with a history of SARS-CoV-2 infection show high levels of protection from breakthrough infectionFootnote 68Footnote 69Footnote 70.
One study was identified that investigated breakthrough infections following a booster dose after a primary series (n=1):
- Booster doses were reported in a study from Israel investigating third shots of ComirnatyFootnote 71. Breakthrough infections in August occurred in 5.5% of individuals that had received 2-doses in January vs. 3.6% of individuals that received the booster in AugustFootnote 71. Across a test-negative and matched case control analysis within the same study, they estimated a 70-84% reduction in the odds of testing positive 14-20 days after the booster compared to protection from the two dose primary series administered 6 months agoFootnote 71.
Immune response markers
This section summarizes 26 studies reporting on immune response markers longitudinally measured up to 8 months following vaccination in individuals with (n=10) and without (n=26) history of previous SARS-CoV-2 infection. The included studies were limited to studies that reported on >30 participants ≥6 months after vaccination with no prior infection (Table 2) or in those vaccinated after recovery from COVID-19 (Table 3). Twenty-six studies looked at circulating serum antibody levels after vaccination, and four studies reported on multiple cellular and humoral immune markers (i.e., B-cells and/or T-cells and antibodies) in the same sample of naïve – vaccinated individuals. Ten studies also reported immune markers in previously infected and vaccinated individuals.
Currently approved vaccines in Canada, Comirnaty (Pfizer-BioNTech/ BNT162b2), Spikevax (Moderna/mRNA-1273), Vaxzevria (AstraZeneca/Covishield/ChAdOx1-S/AZD1222), Janssen (Johnson & Johnson/Ad26.COV2.S), and Bharat Biotech (Covaxin /BBV152)), have been developed to target Spike protein including the RBD of SARS-CoV-2 and thus studies focus on positivity for these markers of immunity rather than nucleocapsid, membrane or envelope proteinsFootnote 72Footnote 73.
The majority of included studies were prospective cohorts or randomized controlled trials that took samples from vaccinated individuals over time. High-level points are listed below and detailed outcomes for each study are located in Table 2 and Table 3. Overall, there was considerable variability across outcomes and studies due to differences in study participants, frequency and duration of follow-up, investigated immune outcomes and measurement methods, which limit the synthesis of results across studies. Studies of infection prior to vaccination frequently did not report the interval from recovery to vaccination or infection severity and post vaccination immune correlates. Furthermore, the evidence is limited for associations between measured long-term immune markers and protection from infection in specific populations, such as children, the elderly, the immunocompromised (e.g., individuals with HIV) and immunosuppressed populations (undergoing cancer treatment or taking immunosuppressant treatments) from both the wild-type and emerging VOCs.
Outcomes reported included both cellular and humoral markers of immunity and a brief background to these markers is provided:
- Cellular immune markers include memory B-cells (i.e., immune cells that produce virus targeting antibodies) and memory T-cells (i.e., immune cells that guide the cell mediated adaptive immune responses) are considered to be indicators of long-term immunityFootnote 1Footnote 74Footnote 75. T-cells are immune cells classified by surface receptors CD4+ or CD8+. The primary role of T-cells can be separated into the production of antibodies via B-cell activation (CD4+ T-cells) or the destruction of infected cells presenting certain antigens (CD8+ T-cells)Footnote 76. Memory B-cells are a type of B lymphocyte that forms part of the adaptive immune system. The included studies isolated peripheral blood mononuclear cells (PBMCs) from serum samples then measured T-cell or B-cell numbers, phenotypes or activity after simulation with various SARS-CoV-2 peptide sequence pools (i.e., amino acids that make up viral proteins)Footnote 77Footnote 78. The variability and/or the lack of detail on peptide sequences used in stimulation studies limit the comparability of study results. Studies also report Interferon-γ (IFNγ), interleukin-2 (IL-2), and/or Tumor Necrosis Factor α (TNFα) from commercial kits to measure T-cell activity against antigens based on secreted cytokinesFootnote 77.
- Humoral immunity, also called antibody-mediated immunity, generally refers to circulating antibodies that are directed at viral antigensFootnote 1Footnote 74. Among included studies, circulating antibodies in serum samples were measured by antibody affinity assays, pseudovirus neutralization assays, flow cytometry, and other molecular biology-based techniques. Variation between assays was noted in several studies with large disagreement between results in some analyses; this is a source of between study heterogeneityFootnote 79Footnote 80Footnote 81Footnote 82. An example of this was a diagnostic test accuracy study that reported the Euroimmun assay had missed 40% of positives in 8 month samples compared to Roche assaysFootnote 81. The range of reported antibody outcomes included total antibodies, neutralizing antibodies (NAbs), antibody class (i.e., IgG, IgM, IgA) which were occasionally described by subclass (i.e., IgG1, IgG3), and/or binding affinity to SARS-CoV-2 viral antigens. Many studies often specified the viral antigen targets of the measured Ig antibodies, which mainly included viral structural proteins: spike (S) protein, S1 or S2 subunit of the S protein and receptor binding domain (RBD) proteins for vaccinated individuals as opposed to other targets, such as nucleocapsid (N), envelope (E), or membrane (M) proteins which are not present in many vaccinesFootnote 73.
Immune response markers ≥6 months post primary series of vaccination in individuals with no history of prior COVID-19
Preliminary data on long-term markers of immunity ≥6 months post vaccination in individuals not previously infected, included 18 studies post Comirnaty vaccination, eight post Spikevax , two post Janssen, one Coronavac (Sinopharm), and two inactivated SARS-CoV-2 (not approved for use) vaccine studies. Most studies measured immune response markers up to 6 months. There were only 3 studies with data to 7 months and 1 studies with data to 8-9 months. Key results from included studies are listed below and detailed outcomes for each study are located in the Table 2.
Key outcomes from B-cell and T-cell immune responses at 6 to 7 months post vaccination (n=4) with Spikevax and Comirnaty demonstrate detectable and durable cellular immune responsesFootnote 29Footnote 30Footnote 31Footnote 39:
- Two studies (Comirnaty n=2 and Spiekvax n=1) indicated stable or increasing antigen specific memory B-cell populations, which is a significant indication of an effective immune responseFootnote 29Footnote 31. One US based study of a general population noted class switching between IgA+ and IgM+ B-cells 1 to 3 months after Comirnaty vaccination towards an increasing number and variety of antigen specific IgG+ B-cellsFootnote 29.
- Reactivation of B-cells collected at 6 months showed strong S-IgG production, which was correlated to Spike+ memory B-cells detected by flow cytometryFootnote 29. Additionally, humoral immunity (Spike and RBD-IgG) was correlated with cellular immune markers CD4+ and Th1 at 6 monthsFootnote 29. Another study reported no correlation between antibodies and cellular immune markersFootnote 30.
- At six months after Comirnaty vaccination effector T-cell and TH1 levels were stable indicating durable memory T-cell responses which may have a limited change in magnitude or composition in response to booster vaccinesFootnote 29:
- Two studies of Comirnaty vaccinated individuals indicated decreases in IFN-γ CD8+ T-cells with increases in IFN-γ CD4+ T-cells (memory B-cells) at 6 months which is a good indication of an effective immune response to vaccinationFootnote 29Footnote 30.
Key outcomes from humoral immune responses at 6 months post full vaccination (n=26 studies) reported the following:
- After 2 doses of Spikevax, the spike IgG antibodies and NAbs remained detectable and stable up to 6 months (n=8)Footnote 29Footnote 35Footnote 36Footnote 49Footnote 83Footnote 84Footnote 85.
- Two publications from the same Spikevax (Moderna) trial report that antibodies remained detectable up to 6 months after the second dose and NAbs were present for almost all participants, however, lower levels of neutralization activity were observed in people over 55 yearsFootnote 36. The other publication reported on neutralization of VOCs which ranged from 96% with Alpha and Delta to 54% with BetaFootnote 44.
- A randomized control trial found Spikevax NAbs neutralized the original variant at 6 months, however 30%-45% of samples did not neutralize Beta, Gamma and DeltaFootnote 83.
- Among American patients on dialysis in a retrospective cohort, 14.1% of Spikevax recipients had seroreverted based on S-IgG levels at 6 monthsFootnote 35.
- After 2 doses of Comirnaty, Spike IgG, RBD IgG antibodies and NAbs decreased from peak at 1 to 3 months after the second dose but remained detectable at 3-7 months (n=18)Footnote 30Footnote 31Footnote 35Footnote 38Footnote 39Footnote 40Footnote 42Footnote 43Footnote 45Footnote 46Footnote 47Footnote 48Footnote 49Footnote 86.
- At 6 months 83.9-99% had detectable RBD-IgG antibodiesFootnote 45Footnote 47.
- One study of 17 individuals with Comirnaty vaccinated and not previously infected indicated all individuals had detectable antibodies at 6 monthsFootnote 50.
- NAb titers were highly maintained for at least 6 months in healthy populations (57%-100%)Footnote 29Footnote 36Footnote 37Footnote 38.
- High risk populations that completed the primary series of Comirnaty with ≥6 months follow-up were reported in seven studies:
- NAbs in nursing home residents at 6 months post vaccination were detectable in 30% of residents that did not have a history of COVID-19 infectionFootnote 38. There was also an 84% reduction in NAb, S-IgG and RBD-IgG titers at 6 months post vaccinationFootnote 38. A second study in nursing home residents found 100% of nursing home residents who had detectable antibodies (RBD IgG/IgM) at baseline had detectable antibodies at 6 monthsFootnote 30.
- A US retrospective cohort that received Comirnaty reported 43.9% of patients on dialysis had seroreverted based on S-IgG at 6 months post vaccinationFootnote 35.
- A prospective cohort of patients with solid tumor had lower positive serology at 6 months compared with healthy controls (79% vs 84%) and 15% of patients with cancer had serorevertedFootnote 43.
- An Israeli prospective cohort study of chronic lymphocytic leukemia (CLL) or small lymphocytic lymphoma (SLL) patients exhibited high level of response to vaccine with 90.2% anti-S IgG positive antibodies compared to 100% of healthy controls, 9.8% of CLL/SLL patients had serorevertedFootnote 42.
- A cross-sectional study of 414 multiple sclerosis (MS) patients receiving treatment had lower S-IgG titers than MS patients not receiving treatment or heathy controlsFootnote 41.
- In one Israeli prospective cohort study found the probability of having low NAb titers (NAb titer <16) 6 months after being fully vaccinated was highest among immunosuppressed men ≥65 compared to healthy adultsFootnote 40.
- Janssen:
- In a randomized control trial of the Janssen vaccine 7 to 9 months after first dose resulted in higher levels of detectable NAbs for adults aged 18 to 55 (95%-100%) compared to older adults >65 (68-69%)Footnote 37. 93% of adults and 86% of older adults had detectable S-IgG at 6 monthsFootnote 37.
- A second US retrospective cohort study indicated that dialysis patients reached the lower limit of S-IgG detection at 6 month follow-upFootnote 35.
- Other vaccines:
- 6 months after 2 doses of inactivated SARS-CoV-2 vaccine NAbs and S protein antibodies decline to 35.6%-51.7% and 52.1%-52.7% depending on the dosing schedule, respectivelyFootnote 87. At 8 months another study reported NAbs were present in 48% of those vaccinatedFootnote 88.
- A single CoronaVac (Sinopharm) study found that NAbs declined from 100% to 12-22% at 6 monthsFootnote 89.
- Studies looking to find associations between immunity markers and demographic variables reported inconclusive results. Two studies indicated no difference in responses due to age, sex, or dose scheduleFootnote 43Footnote 88 while seven indicated lower antibody responses for older vaccine recipients at 6 months post second dose of vaccineFootnote 36Footnote 37Footnote 38Footnote 40Footnote 44Footnote 45Footnote 48.
Immune response markers in previously infected individuals ≥6 months post primary series of COVID-19 vaccination
Preliminary data on long-term markers of immunity ≥6 months post vaccination in individuals with evidence of a previous infection was included in 10 studies post Comirnaty vaccination, three post Spikevax, and one post Janssen. High level points are listed below and detailed outcomes for each study are located in the Table 3:
- Two studies of the Comirnaty vaccine indicate T-cell titers were higher among previously infected vaccinees and they were more likely to have detectable T-cells at month 6 to 7 compared to those that had not had COVID-19 prior to vaccinationFootnote 30Footnote 39. One US prospective cohort study of Spikevax and Comirnaty vaccine found T-cell responses were not significantly elevated in previously infected vaccinees compared to those that had not had COVID-19 prior to vaccination at 6 monthsFootnote 29.
- One Swedish prospective cohort study of 66 previously infected Comirnaty vaccinated healthcare workers indicated vaccinated individuals with a history of COVID-19 had at least 2-fold higher neutralization titers for Alpha, Beta, Gamma, and Delta than vaccinated individuals with no history of COVID-19Footnote 39.
- NAb titers were not dissimilar in previously infected vaccinees (81% - 100%)Footnote 29Footnote 38 compared vaccines with no history of infection (57%-100%)Footnote 29Footnote 36Footnote 37Footnote 38. Larger differences in NAbs were seen among nursing home residents (65% in previously infected vaccinated individuals vs 30% in vaccinated individuals with no history of infection)Footnote 38.
- Two studies (Comirnaty and Spikevax) found no difference in either the decay rate post vaccination of S or RBD-IgG in previously infected and uninfected individualsFootnote 29Footnote 34. One prospective cohort study conducted in Italy of Comirnaty vaccinated healthcare workers reported antibody decay was faster among previously infected compared to uninfected individualsFootnote 46 and two prospective cohort studies of the Comirnaty vaccine reported slower declines in RBD-IgG, IgA, and IgM over time towards 6 months in the vaccinate and previously infected group compared to those with no history of infectionFootnote 47Footnote 50.
Review literature
Three relevant rapid and systematic reviews include COVID-19 research from up to July 2021 on correlates of immunity from vaccinated individuals (Table 4). These are included as resources for research on time points for immune markers earlier than 6 months and analyses of factors that correlate with a strong immune response to vaccination. There are also systematic reviews reinfection data including summaries of confirmed reinfections typically reported as case reports which are not included in this review.
Methods
A daily scan of the literature (published and pre-published) is conducted by the Emerging Science Group, PHAC. The scan has compiled COVID-19 literature since the beginning of the outbreak and is updated daily. Searches to retrieve relevant COVID-19 literature are conducted in Pubmed, Scopus, BioRxiv, MedRxiv, ArXiv, SSRN, Research Square and cross-referenced with the COVID-19 information centers run by Lancet, BMJ, Elsevier, Nature and Wiley. The daily summary and full scan results are maintained in a Refworks database and an excel list that can be searched. Targeted keyword searching was conducted within these databases to identify relevant citations on COVID-19 and SARS-COV-2. Three separate searches were conducted to identify citations relevant to reinfection, breakthrough infections and immunity. Search terms used included: breakthrough terms (efficacy or effective* or breakthrough) across studies with the vaccine tag immunity terms (month* or longitudinal) across studies with the immunology tag.
This review contains research published up to October 22, 2021.
Each potentially relevant reference was examined to confirm it had relevant data and relevant data was extracted into the review.
Acknowledgments
Prepared by: Kaitlin Young, Austyn Baumeister, Lisa Waddell, National Microbiology Laboratory Emerging Science Group, Public Health Agency of Canada.
Editorial review, science to policy review, peer-review by a subject matter expert and knowledge mobilization of this document was coordinated by the Office of the Chief Science Officer: ocsoevidence-bcscdonneesprobantes@phac-aspc.gc.ca
Evidence tables
Table 1 . Randomized controlled trials, prospective cohort and case control studies evaluating vaccine breakthrough infections (n=42)
Study | Method | Key Outcomes |
---|---|---|
Trials (n=5) | ||
Randomized controlled trial US |
Phase 3, observer-blinded, placebo-controlled clinical trial of Spikevax. Included adult volunteers who were at high risk for Covid-19 or its complications and randomly assigned in a 1:1 ratio two intramuscular injections of Spikevax or placebo, 28 days apart, at 99 centers across the United States. The primary end point was prevention of Covid-19 illness with onset at least 14 days after the second dose in participants who had not previously been infected. Asymptomatic infections were identified by seroconversion (antibody specific to nucleocapsid protein) as scheduled visits (months 1 and 2). Efficacy was estimated with a stratified Cox proportional-hazards model. Incidence rates and vaccine efficacy were estimated by 1 minus the hazard ratio (Spikevax vs. placebo), and the corresponding 95% confidence interval was based on the total number of cases adjusted according to total person-time. The duration of follow-up from the second dose was ~5 months. |
|
Feng (2021)Footnote 53 RCT UK |
Data from a randomized efficacy trial of the Vaxzevria vaccine in the United Kingdom (COV002) was analyzed to determine the antibody levels associated with protection against SARS-CoV-2. Using data from this efficacy trial, the authors assessed the correlation between immune markers at 28 days post the second dose of Vaxzevria vaccination and symptomatic and asymptomatic infections. Participants with symptoms were assessed in clinic with a nose and throat swab taken for nucleic acid amplification testing. Additionally, participants were asked to complete a nose and throat swab at home each week, which was used to detect asymptomatic infections. |
|
Emary (2021)Footnote 54 UK |
Volunteers (aged ≥18 years) who were enrolled in phase 2/3 vaccine efficacy studies in the UK (COV002), and who were randomly assigned (1:1) to receive Vaxzevria or a meningococcal conjugate control (MenACWY) vaccine, provided upper airway swabs on a weekly basis and also if they developed symptoms of COVID-19 disease were included. |
|
Voysey (2021)Footnote 55 RCT UK |
This study presents data from three single-blind randomised controlled trials—one phase 1/2 study in the UK (COV001), one phase 2/3 study in the UK (COV002), and a phase 3 study in Brazil (COV003)—and one double-blind phase 1/2 study in South Africa (COV005). Only the UK (COV002) study included symptomatic and asymptomatic cases, thus only these findings are summarized herein. This trial included individuals working in professions with high possible exposure to SARS-CoV-2, such as health and social care settings and elderly (>59 years). MenACWY vaccine was used as the control. Those who met symptomatic criteria had a clinical assessment and a swab taken for a nucleic acid amplification test. To test for asymptomatic infections, participants provided a weekly self-administered nose and throat swab for NAAT testing. Efficacy against SARS-CoV-2 more than 14 days after a second dose of Vaxzevria vaccine was calculated from a Poisson model. |
|
Ella (2021)Footnote 56 Randomized controlled trial India |
Phase 3 clinical trial in 25 Indian hospitals to evaluate the efficacy of the BBV152 COVID-19 vaccine. Healthy adults received two intramuscular doses of vaccine (n=12,221) or placebo (n= 12,198) administered four weeks apart. The primary outcome was laboratory-confirmed symptomatic COVID-19, occurring at least 14 days after the second dose. However, in addition to symptomatic follow-up, a series of post-dose 2 nasopharyngeal swabs were collected on-site for detection of asymptomatic COVID-19 infection at monthly intervals (n=8,721) |
|
Observational studies (n=37) |
||
Moncunill (2021)Footnote 4 Prospective cohort Spain |
This cohort included randomly selected HCWs at baseline. Participants were recruited at the peak of the first wave of the pandemic in Spain and attended several follow-up visits to assess antibody kinetics and information on infection. At month 12, most of the participants had received two doses of either mRNA vaccine (Comirnaty or Spikevax). They collected information on new SARS-CoV-2 infection episodes in this cohort until 6 months after vaccination (M18) through the Occupational Health department at the hospital. |
|
Fowlkes (2021)Footnote 5 Prospective cohort US |
Data from the HEROES-RECOVER Cohorts, a network of prospective cohorts among frontline workers, is reported. Workers were tested weekly for SARS-CoV-2 infection by reverse transcription–polymerase chain reaction (RT-PCR) and upon the onset of any COVID-19–like illness. Reports vaccine effectiveness (VE) estimates for Comirnaty and Spikevax, and examines whether VE differs for adults with increasing time since completion of all recommended vaccine doses. Cox proportional hazards models were used to calculate ratios of unvaccinated to fully vaccinated (≥14 days after receipt of all recommended COVID-19 vaccine doses) infection rates, adjusted for occupation, site, and local viral circulation, and weighted for inverse probability of vaccination using sociodemographic characteristics, health information, frequency of close social contact, and mask use. |
During the total 35 week study period:
During time period when Delta was dominant:
|
Laing (2021)Footnote 10 Prospective cohort US |
HCWs who had no history of COVID-19 were enrolled and followed in the Prospective Assessment of SARS-CoV-2 Seroconversion (PASS) study. Participants were asked to obtain nasopharyngeal SARS-CoV-2 PCR testing upon experiencing symptoms. Asymptomatic infections were determined by nucleocapsid protein (NP) seroconversions (antigen testing) assessed monthly over 6 months post full Comirnaty vaccination. Excluding individuals infected prior to January 31 of 2021, the study followed 227 participants fully vaccinated with BNT162b2 and 17 unvaccinated participants. |
|
Novazzi (2021)Footnote 7 Prospective cohort Italy |
Fully vaccinated HCWs (all considered fully vaccinated in February 2021) who worked in 8 wards deemed to be at “high risk” had a mandatory RT-PCR test every 2 weeks (n=789), and those in 8 “moderate-risk” wards were tested every 4 weeks (n=1,387). If SARS-CoV-2 RNA was detected, the HCW was tested daily until 2 consecutive swabs were negative for SARS-CoV-2. |
|
Shamier (2021)Footnote 20 Prospective cohort Netherlands |
Healthcare workers were followed for primary and breakthrough infections (Comirnaty, Spikevax, Vaxzevria, or Janssen). Compared virological characteristics of first RT-PCR positive samples collected from HCWs with breakthrough infections (occurring between Apr-Jul 2021) to first RT-PCR positive samples from the same cohort of HCWs prior to the onset of vaccination (Apr-Dec 2020). Infections were classified as breakthrough infections if the date of the first positive SARS-CoV-2 RT-PCR was more than 14 days after completion of all recommended vaccine doses. |
|
Issac (2021)Footnote 8 Prospective cohort India |
Prospectively evaluated 324 employees working in hospital throughout the study period of 170 days (January 27, 2021 - July 15, 2021). Of this population, 243 (75%) completed the full primary cycle of vaccination, i.e., completed 14 days post-vaccination with 2 doses of Vaxzevria vaccine, and 80 (25%) were not vaccinated. All the employees were under surveillance after vaccination to quantify the breakthrough infections (the symptomatic suspected cases, as well as the high-risk contacts of infected cases, were confirmed using RT-PCR screening). The cohorts were compared using a binary logistic regression and a time-dependent cox proportionality hazard model. The event time observed was the number of days from the second dose until a COVID-19 infection was confirmed. Covariates tested were age, sex, and contact exposure status. |
|
Ronchini (2021)Footnote 59 Prospective cohort Italy |
Healthcare workers at hospital sites in Milan were followed. Antibody testing was done every 4 weeks. PCR test was done after a positive serological test, in case of symptom, after holidays and every 2 weeks for medical doctors. Vaccinations started in January 2021 (follow-up time max for breakthrough infection was 5 months). |
|
Pouwels (2021)Footnote 25 Prospective cohort UK |
Investigated the effectiveness of Comirnaty and Vaxzevria vaccines in a large, community-based follow-up study of randomly selected households (n=221909 Alpha Dec 1, 2020- May 16, 2021, n= 358983 Delta May 17– Aug 1, 2021). RT–PCR tests were performed after a pre-determined schedule, irrespective of symptoms every week for the first month and then monthly for 12 months from enrollment. At each visit, enrolled household members provided a nose and throat self-swab following instructions from the study worker. Outcomes investigated: variation in vaccine effectiveness by time from second vaccination, long-term health conditions, age and prior infection and assessed viral burden in new PCR-positive cases occurring ≥14 d after second vaccination using Ct values. Adjusted analysis for risk factors that also affect vaccination: patient-facing healthcare work, long-term health conditions, background ‘force of infection’, infection rates varying by age, calendar time and geographical region. Follow-up post second vaccination was median (IQR) Vaxzevria 41 days (27–57), Comirnaty 59 d (35-86). |
|
Kale (2021)Footnote 9 Prospective cohort India |
The study was conducted on HCWs (n=1858) receiving two doses of Vaxzevria vaccine. Serial blood samples were collected to measure SARS-CoV-2 IgG and neutralizing antibodies. A vaccine breakthrough infection was defined as the detection of SARS-CoV-2 RNA or antigen in a respiratory specimen collected from an individual who had received either one or two doses of vaccine. Fully vaccinated was defined as HCWs who received two doses of vaccine and developed infection after 14 days of the second dose. 46 RT-PCR positive samples from breakthrough infections were subjected to whole genome sequencing (WGS). |
|
Wickert (2021)Footnote 57 Prospective cohort US |
Vaccine effectiveness at preventing infection was estimated by comparing infection risk as a function of time since vaccination in a cadet population (n=4200). Weekly surveillance testing using the Sofia SARS Antigen Fluorescent Immunoassay (FIA) provided infection point prevalence estimates. Asymptomatic individuals identified during surveillance testing and symptomatic individuals received RT-PCR based tests. Infection risk as a function of vaccination time status was determined by comparing the total person-days within the observation period and the total infection count, per 10,000 person-days for each vaccination group. At the end of the study period, 36% of cadets were fully vaccinated (defined as 14 days after the 2nd dose of Comirnaty). |
|
Muhsen (2021)Footnote 3 Prospective cohort Israel |
Long-term care facility (LTCF) HCWs underwent weekly nasopharyngeal SARS-CoV-2 RT-PCR testing. Fully vaccinated (14+ days after second dose of Comirnaty; n=6960) and unvaccinated HCWs (n=2202) were followed until SARS-CoV-2 acquisition, or end of follow-up. Hazard ratios were calculated via Cox proportional hazards regression models, adjusting for socio-demographics and residential-area COVID-19 incidence. |
|
Thompson (2021)Footnote 61 Prospective cohort US |
Prospective cohorts of healthcare personnel, first responders, and other essential and frontline workers (n=3,975) completed weekly SARS-CoV-2 testing. Self-collected mid-turbinate nasal swabs were tested by qualitative and quantitative RT-PCR. HCWs were considered fully vaccinated 14 days after second dose of either Comirnaty or Spikevax mRNA vaccines. Hazard ratios were estimated by the Andersen-Gill extension of the Cox proportional hazards model, which accounted for time-varying vaccination status. |
|
Martínez-Baz (2021)Footnote 63 Prospective cohort Spain |
This study followed all individuals aged ≥ 18 years covered by the Navarre Health Service, who had been close contacts of laboratory-confirmed COVID-19 cases from January to April 2021. Close contacts were tested by RT-PCR for SARS-CoV-2 initially and 10 days after the last contact. Cox regression provided estimates of the crude and adjusted relative risks. Adjusted models included age groups, sex, major chronic condition, contact setting (household or other) and month. Contacts were considered fully vaccinated 14 days after second dose of Comirnaty or Spikevax. |
|
Thompson 2021Footnote 60 Prospective cohort US |
Prospective cohorts of health care personnel, first responders, and other essential and frontline workers in eight U.S. locations were included. Active surveillance for symptoms consistent with COVID-19–associated illness occurred through weekly text messages, e-mails, and direct participant or medical record reports. Participants self-collected a midturbinate nasal swab weekly, regardless of COVID-19–associated illness symptom status and collected an additional nasal swab and saliva specimen at the onset of COVID-19–associated illness. Vaccine effectiveness was analyzed in participants with full immunization of two doses of mRNA COVID-19 vaccines (Comirnaty and Spikevax. |
|
Katz (2021)Footnote 6 Prospective cohort Israel |
Followed HCWs in 6 hospitals to estimate the effectiveness of the Comirnaty COVID-19 vaccine in preventing SARS-CoV-2 infection. Participants filled out weekly symptom questionnaires and provided weekly nasal specimens. VE against PCR-confirmed SARS-CoV-2 infection was calculated using the Cox Proportional Hazards model. Estimated VE >14 days after receipt of the second vaccine dose. Only included participants who were seronegative at enrollment and did not have PCR-confirmed SARS-CoV-2 infection at or prior to enrollment. |
|
Lumley (2021)Footnote 11 Prospective cohort UK |
Healthcare workers (HCWs) (n=13,109) were followed to investigate and compare the protection from SARS-CoV-2 infection conferred by 2 doses of vaccine (by either Comirnaty vaccine or Vaxzevria vaccine) with onset at least 14 days after the second injection. Protection from prior infection was also examined (Results in Table 1). Staff remained at risk of infection until the earliest of the study end, or a positive PCR test. To assess the impact of the Alpha variant on (re)infection risk, they analysed PCR-positive results with and without S-gene target failure (SGTF), and those confirmed as Alpha on genome sequencing. Protection was calculated as 100*(1-IRR). |
|
Patalon (2021)Footnote 71 Test negative case control Israel |
The study population consisted of Maccabi Healthcare Services (MHS) members, aged 40+, who received either two or three doses of the Comirnaty vaccine. Second doses were typically administered in January and booster doses in August, end of follow up was August 21st, 2021. Participants were excluded if they tested positive for SARS-CoV-2 before the start of the follow-up period. A test-negative case control analysis sought to estimate the reduction in the odds of a positive test at different time intervals following receipt of the booster (third) dose (0-6 days, 7-13 days, 14-20 days) compared to two-dose only vaccinees. Covariates included the 10-year age category, biological sex, time since receipt of the 2nd dose, and comorbidities. A matched case control design was also conducted. Cases were defined as individuals with a positive PCR test occurring after August 1, 2021, among those 40 years of age or older who did not have a previous positive test recorded and who received at least two doses of the vaccine. Up to 20 controls per case were drawn from the entire population. |
|
Chemaitelly (2021)Footnote 24 Test negative case control Qatar |
Estimate vaccine effectiveness against any SARS-CoV-2 infection for Comirnaty. Authors define breakthrough as any infection following any dose no matter the time frame but present study findings separately for post 1st and 2nd dose and beyond. Comirnaty effectiveness was assessed against Alpha, Beta, and Delta infections separately to investigate whether declining effectiveness could have been confounded by exposure to different variants over time. Case participants (PCR-positive persons) and controls (PCR-negative persons) were matched one to one according to sex, 10-year age group, nationality, reason for SARS-CoV-2 PCR testing, and calendar week of PCR test. |
|
Bruxvoort (2021)Footnote 15 Test negative case control US |
Whole genome sequencing was conducted for SARS-CoV-2 positive specimens to determine Spikevax effectiveness against Delta, Mu and other VOCs. Test-positive cases (n=8,163) were matched 1:5 to test-negative controls on age, sex, race/ethnicity, and specimen collection date. Conditional logistic regression was used to compare odds of vaccination among cases versus controls, adjusting for confounders. Analyses of VE by time since receipt of second dose of Spikevax (14-60 days, 61-90 days, 91- 120 days, 121-150 days, 151-180 days, and >180 days) were conducted for Delta (overall and by age), non-Delta variants, and unidentified variants. |
|
Tang (2021)Footnote 23 Test negative case control Qatar |
Between December 21, 2020 and July 21, 2021, 877,354 individuals completed the two-dose regimen of Comirnaty and 409,041 completed the two-dose regimen of Spikevax. Vaccine effectiveness was estimated against documented infection (defined as a PCR-positive swab regardless of the reason for PCR testing or presence of symptoms) with the Delta variant. Cases and controls were matched one-to-one by sex, 10-year age group, nationality, reason for SARS-CoV-2 polymerase chain reaction (PCR) testing, and calendar week of PCR test. |
|
Barlow (2021)Footnote 26 Test negative case control US |
Estimate the effectiveness of vaccination against SARS-CoV-2 infection during July 2021 (when Delta was dominant). 500 case control pairs were matched (n=1000). Cases included a random sample of individuals that tested positive for SARS-CoV-2 in July 2021 and were reported by electronic laboratory report, were >15 years of age, and had no prior known SARS-CoV-2 infections. Controls were age and postal code matched individuals that tested negative for SARS-CoV-2 during July 2021. Vaccinations were considered invalid if they were administered <14 days prior to the case's positive test collection date. |
|
Singh (2021)Footnote 58 Test negative case control India |
Vaccine effectiveness of vaccination was investigated. The vaccines administered at the time were Vaxzevria and Covaxin. This case control study was conducted among people aged ≥45 years. The cases were the COVID-19 patients who were admitted or visited the All India Institute of Medical Sciences (AIIMS) flu clinic. The controls were the individuals tested negative for severe acute respiratory syndrome coronavirus-2 (SARS CoV-2) at the Virology laboratory. This was an unmatched case control study, but logistic regression analysis was adjusted for age, sex, occupation, COVID-inappropriate behaviour score, chronic co-morbidity, H/O hospitalisation, ILI, prior COVID-19 and high-risk contact with a case or suspect. |
|
Li (2021)Footnote 66 Test negative case control China May-Jun 2021 |
Estimate vaccine effectiveness of two SARS-CoV-2 inactivated vaccines (China National Biotec Group SARS-CoV-2 vaccine and the CoronaVac vaccine) against infection or pneumonia associated with the Delta variant. Defined the second-dose vaccination (fully vaccinated) as having elapsed for more than 14 days after the second dose upon the clinical diagnosis (for cases) or the last contact with the cases (for contacts). |
|
Sheikh (2021)Footnote 16 Test negative case control Scotland |
Estimate vaccine effectiveness (Comirnaty and Vaxzevria) against risk of SARS-CoV-2 infection. S gene-positive detection was used as a proxy for Delta identification and S-gene negative detection was representative of Alpha. Vaccine effectiveness estimates were obtained from a generalised additive logistic model adjusting for age, temporal trend when the swab was taken, and number of previous tests using splines plus sex and deprivation. |
|
Butt (2021)Footnote 64 Test negative case control Qatar |
Determined the vaccine effectiveness of mRNA vaccines (Comirnaty and Spikevax) in preventing confirmed SARS-CoV-2 infection in pregnant women at a national referral hospital ≥14 days after the second dose of the vaccine. For each woman who tested positive, they identified up to 3 RT-PCR negative controls matched on age and reason for testing. Authors did not match the cases and controls for the time of testing. |
|
Chemaitelly (2021)Footnote 12 Test negative case control Qatar |
With essentially only Beta and Alpha cases identified in the viral genome sequencing and the multiplex RT–qPCR variant screening conducted on cases between 8 March and 10 May 2021, an Alpha infection was proxied as an S-gene target failure case and a Beta infection as an S-gene target positive case. Spikevax COVID-19 Vaccine effectiveness analyses against Alpha were performed using independent samples of n=21,305 PCR-positive cases and n=21,305 PCR-negative controls while VE analyses against Beta were performed using n=44,737 PCR-positive cases and n=44,737 PCR-negative controls. |
|
Pramod (2021)Footnote 65 Test negative case control India |
Information on vaccination status (Covishield) of cases with COVID-19 among healthcare workers and an equal number of matched controls, (i.e., positive and negative for SARS-CoV-2 by RT-PCR), was obtained. The cases and controls were matched for age and date of testing (n=360 case control pairs). The groups were compared using multivariable conditional logistic regression to calculate odds ratios (OR), adjusted for gender, occupational role, presence of symptoms and presence of a comorbidity condition. |
|
Abu-Raddad (2021)Footnote 17 Test negative case control Qatar |
Data for SARS-CoV-2 were extracted from Qatar’s nationwide digital-health information platform. Cases and controls were matched one-to-one by age, sex, nationality and reason for PCR testing. Effectiveness was estimated against documented infection with the Alpha or Beta variants. |
|
Thiruvengadam (2021)Footnote 19 Test negative case control India |
Determined the vaccine effectiveness of Vaxzevria vaccine in preventing confirmed infection during a surge of Delta infections in India. Cases were RT-PCR positive for SARS-CoV-2 infection (n=2766). The controls (n=2377) were selected randomly from the individuals who tested negative and were matched in numbers for each calendar week of testing during the study period. Defined complete vaccination as when the participant had completed at least 14 days after the second dose of the vaccine. Adjusted odds ratio (aOR) was estimated by a multivariable logistic regression model which included confounders. |
|
Andrejko (2021)Footnote 62 Test negative case control US |
Enrolled cases (testing positive, n=525) and controls (testing negative, n=498) from among the population whose SARS-CoV-2 molecular diagnostic test results were reported to the California Department of Public Health. Participants were matched on age, sex, and geographic region. Assessed participants’ self-reported history of mRNA-based COVID-19 vaccine receipt (Comirnaty and Spikevax). Participants were considered fully vaccinated two weeks after second dose receipt. |
|
Kislaya (2021)Footnote 21 Case-case Portugal |
Utilized RT-PCR positive cases notified to the National Surveillance System and electronic vaccination register to calculate the odds of vaccine breakthrough in Delta cases compared to Alpha SARS-CoV-2 cases. This was estimated by conditional logistic regression adjusted for age group, sex, and matched by the week of diagnosis. Whole-genome sequencing (WGS) or spike (S) gene target failure (SGTF) data were used to classify Delta and Alpha. Participants were considered fully vaccinated two weeks after second dose receipt of mRNA vaccine (Comirnaty or Moderna). |
|
Cavanaugh (2021)Footnote 28 Case control US |
Among Kentucky residents infected with SARS-CoV-2 in 2020, vaccination status of those reinfected during May–June 2021 was compared with that of residents who were not reinfected. A case-patient was defined as a Kentucky resident with laboratory-confirmed SARS-CoV-2 infection in 2020 and a subsequent positive NAAT or antigen test result during May 1–June 30, 2021. Control participants were Kentucky residents with laboratory-confirmed SARS-CoV-2 infection in 2020 who were not reinfected through June 30, 2021. Case-patients and controls were matched on a 1:2 ratio based on sex, age, and date of initial positive SARS-CoV-2 test. Case-patients were considered fully vaccinated if a single dose of Janssen (Johnson & Johnson) or a second dose of an mRNA vaccine (Comirnaty or Spikevax) was received ≥14 days before the reinfection date. |
|
Chau (2021)Footnote 22 Case control Vietnam |
Studied breakthrough infections during an outbreak among healthcare workers of a major infectious diseases hospital. Used available data on neutralizing antibodies from a vaccine study for case control analyses. Matched cases with the controls by age and gender. |
|
Duerr (2021)Footnote 13 Case control US |
Compared the SARS-CoV-2 genomes of 76 breakthrough cases after full vaccination with Comirnaty, Spikevax, or Janssen to unvaccinated controls (n=1,046) in metropolitan New York, including their phylogenetic relationship, distribution of variants, and full spike mutation profiles. Unmatched and matched statistical analyses considering age, sex, vaccine type, and study month as covariates were conducted. Breakthrough infection was defined as infection occurring >14 days after inoculation with the second dose of the mRNA vaccines, or with the single dose COVID-19 Janssen vaccine. |
|
McEwen (2021)Footnote 67 Case control US |
Examined SARS-CoV-2 genomes isolated from individuals identified as vaccine breakthrough cases (n=20) and compared them with the background of SARS-CoV-2 sequences from Washington over the same time interval (n=5174). Vaccine breakthrough was defined as testing positive via RT-PCR >2 weeks post second dose of Comirnaty or Spikevax vaccines). |
|
Mor (2021)Footnote 18 Case control Israel |
Used logistic regression, with variant type as the dependent variable, vaccination status (Comirnaty) as the main explanatory variable, controlling for age, sex, subpopulation, place of residence and time of sample, to estimate the odds ratio for a vaccinated case to have the Beta versus the Alpha variant, within vaccinated and unvaccinated persons who tested positive. Information (including sequencing results) on confirmed COVID-19 cases in the country was retrieved from the Israeli Ministry of Health’s databases. |
|
Kustin (2021)Footnote 14 Case control study Israel |
Examined the distribution of SARS-CoV-2 variants observed in infections of vaccinated individuals and matched infections of unvaccinated individuals. The authors conducted the analysis with breakthrough infections defined as individuals who had a positive PCR test that was performed at least one week after the second vaccine dose (denoted as full effectiveness, FE). However, they also provide case numbers that were infected post 14 days of 2nd dose. Conducted PCR and viral genome sequencing on 149 paired “fully vaccinated” individuals and 247 pairs of “partially vaccinated” individuals (only 1 dose). |
|
Table 2 . Immune responses ≥6 months after the primary series of COVID-19 vaccination in individuals with no history of prior COVID-19 (n=26)
Study | Method | Key outcomes |
---|---|---|
Circulating antibody, B-cell and T-cell immune responses (n=4) | ||
7 months | ||
Haverall (2021)Footnote 39 Prospective cohort Sweden |
Healthcare worker binding antibodies (IgG), T-cell responses, and neutralizing antibodies against wild-type and Delta were assessed using longitudinally collected blood samples from the COMMUNITY (COVID-19 Immunity) study for up to 7 months. At the last time measurement data was available for 246 naïve individuals (66 previously infected) who received the Comirnaty vaccine. S-IgG binding antibodies were determined by multiplex antigen bead array, IFN-γ T-cells through IGRA assay as well as T-SPOT® Discovery SARS-CoV-2 kit, and neutralizing antibodies were through pseudotyped virus assays, and for a subset of 17 naïve vaccinated live virus micro-neutralization. Outcomes related to infected then vaccinated individuals is located in Table 3. |
T-cells at 7 months:
Antibodies at 7 months:
VOC at 7 months:
|
6 months | ||
Giménez (2021)Footnote 30 LTE Prospective cohort Spain |
Forty-six (10 previously infected) nursing home residents from a prior study that captured both B and T cell responses after Comirnaty vaccination were reassessed between 179 to 195 days for total RBD and N antibodies (IgG and IgM) (n=45) using the Roche Elecsys® electrochemiluminescence assay as well as IFNγ‐producing‐CD8+ and CD4+ T cells measured through flow cytometry. Outcomes related to infected then vaccinated individuals are in Table 3. |
T-cell at 6 months:
Antibodies at 6 months:
|
Goel (2021)Footnote 29 Prospective cohort US |
Longitudinal antibody (1, 3, 6 months after second dose) and memory B and T cell responses including against VOCs (Alpha, Beta, Delta) after mRNA vaccination (Comirnaty or Spikevax) were measured in 45 naïve and 16 recovered individuals. RBD and Spike IgG were measured by ELISA, NAbs were determined against pseudo typed variants, T-cell were detected with activation induced marker assay (AIM) and B-cells through using biotinylated proteins in combination with different streptavidin (SA)-fluorophore conjugates from peripheral blood mononuclear cell (PBMC) samples. Outcomes related to infected then vaccinated individuals is located in Table 3. |
B-cells at 6 months:
T-cells at 6 months:
Antibodies at 6 months:
NAbs at 6 months:
|
Ciabattini (2021)Footnote 31 Prospective cohort Italy |
Spike-specific memory B cells and humoral responses up to 6 months after vaccination with Comirnaty vaccine (2 doses- 3 weeks apart) was investigated in 145 HCWs aged 24-75 without a laboratory confirmed history of SARS-CoV-2 infection. Plasma samples were collected after first dose 21 days, after second dose 7 days, 21, 28, 90 and 160-180. Surrogate neutralization assays were used to assess whether the antibodies block the RBD-ACE2 interaction. ELISpot assays were used to measure spike-specific antibody secreting cells in restimulation experiments. |
Spike specific IgG antibodies:
Spike specific memory B cells
|
Circulating antibody immune responses (n=22) | ||
Canaday (2021)Footnote 38 Prospective cohort US |
Circulating antibodies and NAbs were measured in 120 nursing home residents and 92 HCWs 2 weeks and 6 months after a full primary series of Comirnaty vaccination. S and RBD-IgG were measured through ELISA and NAbs through pseudovirus neutralization assay. Outcomes related to previously infected then vaccinated individuals are in Table 3. |
At 6 months:
|
Eyran (2021)Footnote 50 Prospective cohort Israel |
A subset of 20 COVID-19 recovered patients and 17 COVID-19 naïve individuals who received the Cominarty vaccine and were followed for samples 8, 35, 91, and 182 days (6 months) after the second dose to measure RBD Ig levels. Outcomes related to previously infected then vaccinated individuals are in Table 3. |
Antibodies at 6 months:
|
Hsu (2021)Footnote 35 Retrospective cohort US |
Dialysis patients (1567 with no prior history of COVID-19 and 299 with prior COVID-19) who attended a maintenance dialysis center and had both vaccine doses (441 Comirnaty/779 Spikevax/347 Janssen*) were analyzed for their long term Spike (S) -IgG responses. SARS-CoV-2 spike antigen was measured using the chemiluminescent assay ADVIA Centaur® XP/XPT COV2G. Outcomes related to previously infected then vaccinated individuals are in Table 3. |
Vaccinated naïve dialysis patients:
|
Kontopoulou (2021)Footnote 47 Prospective cohort Greece |
RBD IgG responses after Comrinaty were investigated longitudinally from 2 weeks up to 6 months in a cohort of 252 HCWs (35 prior infection/217 no prior infection). IgG antibodies were assessed SARS-CoV-2 IgG II Quant assay. Outcomes related to previously infected then vaccinated individuals are in Table 3. |
|
Remy (2021)Footnote 34 Prospective cohort US |
A convenience sample of medical research company employees and household members (n=261) was used. Voluntary self-collected blood samples were measured for Spike IgG once per month up to 13 months. Persons who completed their primary series of vaccination (n= 21 Janssen, n= 78 Moderna, n=152 Pfizer) and 9 were unvaccinated. Forty-three participants reported prior positive PCR before vaccination, 9 reported a breakthrough infection and 24 reported a booster vaccination. Outcomes related to previously infected then vaccinated individuals are in Table 3. |
6 months post vaccination:
|
Salvagno (2021)Footnote 46 Prospective cohort Italy |
787 HCWs who received the Comirnaty vaccine 3 weeks apart, had blood samples drawn before the first and second dose as well as 1, 3, and 6 months after the second dose to follow the kinetics of total antibodies as measured through the Roche Elecsys Anti-SARS-CoV-2 S chemiluminescent Immunoassay. Outcomes related to previously infected then vaccinated individuals are in Table 3. |
Antibodies at 6 months:
|
Zhong (2021)Footnote 49 Prospective cohort US |
1960 HCWs, 1887 HCWs with no prior infection (1530 Comirnaty/357 Spikevax), before vaccination, and 73 with prior infection (62 Comirnaty/11 Spikevax) were analyzed to determine IgG responses 1, 3, 6 months after vaccination other vaccines not specified). Anti spike (S) IgG was measured through the Euroimmun ELISA assay. Linear regression adjusted median IgG for time since vaccination, prior infection, vaccine, age and sex. Outcomes related to previously infected then vaccinated individuals are in Table 3. |
At 6 months:
|
Doria-Rose (2021)Footnote 36 Randomized controlled trial US |
Vaccine immunity was evaluated in 33 healthy adult participants in an ongoing phase 1 trial reported on antibodies 180 days post second dose of mRNA-1273 (Moderna). Vaccine schedule was two doses 28 days apart. Half-life was estimated using an exponential decay model and power-law model. |
At 6 months antibody activity was high in all participants and mean binding antibodies were lower with increasing age geometric mean end-point titers (GMTs):
Nearly all participants had detectable activity in a pseudoneutralization assay with 50% inhibitory dilution (ID50) GMTs:
All participants had activity on the live-virus focus-reduction neutralization nNeonGreen test ID50 GMTs with lower levels at increasing age:
Estimated means for half-life for binding antibodies after day 43 was 52-109 days, nAbs 69-173 days and live-virus neutralization 68 -202 days, which is consistent with data from previously infected patients up to 8 months. |
Pegu (2021)Footnote 44 Prospective cohort US |
The impact of SARS-CoV-2 variants on binding, neutralizing, and ACE2-competing antibodies elicited by the vaccine mRNA-1273 (Moderna) over seven months was evaluated. Vaccine schedule was two doses 28 days apart. Sera from a random sample of 8 volunteers in each of three age groups (18-55, 55-70, and 71+) was tested at four time points: 4 weeks after the first dose, and 2 weeks, 3 months, and 6 months after the second dose (days 29, 43, 119, and 209 after the first dose, respectively). Three functional assays and two binding assays were used to assess the humoral immune response to the SARS-CoV-2 spike protein. |
|
Liao (2021)Footnote 87 Prospective cohort China |
Serum samples of 158 adults aged 18-59 were evaluated for immune persistence 180 days after a 2nd dose of inactivated SARS-CoV-2 Vaccine (clinical trial NCT04412538) |
|
Li (2021)Footnote 89 Randomized controlled trail China |
In phase 1 of this trial, 68 healthy adults aged 60+ were randomly allocated (1:1:1) to either a 3 μg, 6 μg, or placebo group. Neutralizing antibody titers were evaluated at 6 months or more after the second dose of CoronaVac in all participants. The impact of a 3rd dose of CoronaVac on immune responses was evaluated up to 28 days post booster given at 8 months after the 2nd dose. 303 participants were randomly assigned (2:2:2:1) to either a 1.5 μg, 3 μg, 6 μg, or placebo group. |
|
Herishanu (2021)Footnote 42 Prospective cohort Israel |
This report follows up with a cohort of naïve adult patients with chronic lymphocytic leukemia (CLL) or small lymphocytic lymphoma (SLL) who were immunized with two doses of Comirnaty. This report covered antibody responses for 61 CLL/SLL patients and 39 healthy controls 6 months (IQR: 168.0 -178.5 days) measured the Elecsys® Anti-SARS-CoV-2 S assay. |
|
Levin (2021)Footnote 40 Prospective cohort Israel |
HCWs from the Sheba Medical Center were recruited before administration of the Comirnaty vaccine then sampled monthly after the second dose for up to 6 months. Of the 12603 elibible HCWs, 4868 were recruited for study participation. At 6 month follow-up 1370 HCWs were tested for antibodies and 517 for NAbs. RBD-IgG was assessed with the commercially available Beckman Coulter SARS-CoV-2 IgG assay in addition to pseudovirus neutralization assays. |
Antibodies at 6 months:
NAb:
|
Kertes (2021)Footnote 48 Retrospective cohort Israel |
This study extracted data from a large health centre database which carried out serology testing (Abbot Quant II IgG anti-Spike CoV2-SARS kit) for employees and geriatric residents of medical and retirement facilities. All health center members that received both Comirnaty vaccine doses and had a subsequent IgG test were included. Serology results were available for 1,820 at the 6-month mark (>150 days post vaccination). |
At 6 months:
|
Yue (2021)Footnote 88 Prospective cohort China |
Three hundred and fifty-five volunteers involved in the development of inactivated vaccines received two doses of vaccine (0 and 14 days or 0 and 28 days). Neutralizing antibody titers were then measured at 1 and 8 months after the second dose. |
|
Choi (2021)Footnote 83 Randomized controlled trial US |
In this ongoing phase 2a Spikevax trial, where participants where the 100-µg dose for primary vaccine series group received a booster dose of Spikevax or variant-modified mRNA vaccine including multivalent mRNA-1273.211 (mixed Spikevax and mRNA-1273.351) or mRNA-1273.351 on its own. Only data collected at 6 months post primary series and before booster doses met the inclusion criteria of this review, n= 59 participants. |
6 months after primary series:
|
Waldhorn (2021)Footnote 43 Prospective cohort Israel |
One hundred and fifty four patients with two doses of Comirnaty undergoing cancer treatment during the whole study were recruited then matched to HCWs of the same age, both had serology done at the same time points. Serum antibodies were measured using the Liaison; DiaSorin SARS-CoV-2 anti-spike (S) S1/S2 IgG assay after the first vaccination, 14 days after the second dose and at 6 months. |
At 6 months:
|
Sadoff (2021)Footnote 37 Randomized control trial US, Belgium |
Long-term follow-up of participants in a Phase 1/2a trial for Janssen single dose (5x1010 virus particles (vp)). |
Post 1 dose Janssen:
Booster dose at 6 months:
|
Israel (2021)Footnote 45 Retrospective cohort Israel |
IgG antibody kinetics were investigated in a cohort of 2,653 fully vaccinated with Comrinaty (summarized) and 4,361 non-vaccinated previously infected patients (not summarized) drawn from the Leumit Health Services (LHS), a large nation-wide health maintenance organization (HMO). Serology results (Abbot Alinity™ i system) target the spike protein IgG antibodies and demographics were extracted from health records. |
Vaccinated individuals up to 6 months:
|
Achiron (2021)Footnote 41 Cross-sectional study Israel |
Multiple sclerosis (MS) patients (n=414) and healthy controls (n=89) had blood drawn at least 28 days (2.3 to 6.3 months) after the administration of Comirnaty vaccine. Samples were assessed for levels of S-IgG (Euroimmun) and for a subset were assessed for B and T-cell responses* 2 to 4 months after vaccination (Mabtech RBD ELISpotPlus and IFNg and IL2 FluoroSpot assays). |
Antibodies:
|
Chu (2021)Footnote 84 Randomized controlled trial US |
In this ongoing Phase 2 trial 600 individuals were randomized to placebo, 50 µg Spikevax or 100 µg Spikevax boosters. Pseudo virus neutralizing antibody titers were assessed before the booster was administered 6 to 8 months after the primary series. |
|
Pan (2021)Footnote 85 Surveillance program NR |
As part of a surveillance testing program using the NIDS® COVID-19 Neutralizing Antibody (NAb) Rapid Test, 93 Spikevax and 122 Cominarty vaccinated individuals had neutralizing antibody titers tested up to 7 months after full vaccination. |
5 and 7 months after the full vaccine series:
|
LTE= letter to the editor, est= estimated date based on publication submission, RBD= Receptor binding domain, S = Spike, HCW = Healthcare worker |
Table 3 . Immune responses in ≥6 months after primary series of COVID-19 vaccination in individuals with a history of COVID-19 (n=10)
Study | Method | Key outcomes |
---|---|---|
Circulating antibody, B-cell and T-cell immune responses (n=3) | ||
Goel (2021)Footnote 29 new Prospective cohort US |
Longitudinal antibody (1, 3, 6 months after second dose) and memory B and T cell responses including against VOCs (Alpha, Beta, Delta) after mRNA vaccination (Comirnaty or Spikevax) were measured in 45 naïve and 16 recovered individuals. RBD and Spike IgG were measured by ELISA, NAbs were determined against pseudo typed variants, T-cell were detected with activation induced marker assay (AIM) and B-cells through using biotinylated proteins in combination with different streptavidin (SA)-fluorophore conjugates from peripheral blood mononuclear cell (PBMC) samples. |
B-cells at 6 months:
Antibodies at 6 months:
NAbs at 6 months:
|
Haverall (2021)Footnote 39 Prospective cohort Sweden |
HCWs binding antibodies (IgG) and neutralizing antibodies against wild-type and VOC were assessed using longitudinally collected blood samples from the COMMUNITY (COVID-19 Immunity) study for up to 7 months. At the last time measurement data was available for 246 naïve individuals (66 previously infected) who received the Comirnaty vaccine. S-IgG binding antibodies were determined by multiplex antigen bead array, IFN-γ T-cells through IGRA assay as well as T-SPOT® Discovery SARS-CoV-2 kit, and neutralizing antibodies were through pseudotyped virus assays and for a subset of 17 naïve vaccinated live virus micro-neutralization. |
T-cells at 7 months:
Antibodies at 7 months:
VOC at 7 months:
|
Giménez (2021)Footnote 30 Prospective cohort Spain |
Forty-six (10 previously infected) nursing home residents from a prior study that captured both B and T cell responses after Comirnaty vaccination were reassessed between 179 to 195 days for total RBD and N antibodies (IgG and IgM ) (n=45) using the Roche Elecsys® electrochemiluminescence assay as well as IFNγ‐producing‐CD8+ and CD4+ T cells measured through flow cytometry. |
Antibodies:
T-cell:
|
Circulating antibody immune responses (n=7) | ||
Salvagno (2021)Footnote 46 Prospective cohort Italy |
787 HCWs who received the Comirnaty vaccine 3 weeks apart, had blood samples drawn before the first and second dose as well as 1, 3, and 6 months after the second dose to follow the kinetics of total antibodies as measured through the Roche Elecsys Anti-SARS-CoV-2 S chemiluminescent Immunoassay. |
|
Eyran (2021)Footnote 50 Prospective cohort Israel |
A subset of 20 COVID-19 recovered patients and 17 COVID-19 naïve individuals who received the Comirnaty vaccine and were followed for samples 8, 35, 91, and 182 days (6 months) after the second dose to measure RBD Ig levels. |
Recovered individuals received a single dose:
|
Kontopoulou (2021)Footnote 47 Prospective cohort Greece |
RBD IgG responses after Comirnaty were investigated longitudinally from 2 weeks up to 6 months in a cohort of 252 HCWs (217 no prior infection). IgG antibodies were assessed SARS-CoV-2 IgG II Quant assay. |
|
Zhong (2021)Footnote 49 Prospective cohort US |
1960 HCWs, 1887 HCWs with no prior infection (1530 Comirnaty/357 Spikevax), before vaccination, and 73 with prior infection (62 Comirnaty/11 Spikevax) were analyzed to determine IgG responses 1, 3, 6 months after vaccination other vaccines not specified). Anti spike (S) IgG was measured through the Euroimmun ELISA assay. Linear regression adjusted median IgG for time since vaccination, prior infection, vaccine, age and sex. |
At 6 months:
|
Remy (2021)Footnote 34 Preprint new Prospective cohort US |
A convenience sample of medical research company employees and household members (n=261) was used. Voluntary self-collected blood samples were measured for Spike IgG once per month up to 13 months. Persons who completed their primary series of vaccination (n= 21 Janssen, n= 78 Moderna, n=152 Pfizer) and 9 were unvaccinated. Forty-three participants reported prior positive PCR before vaccination, 9 reported a breakthrough infection and 24 reported a booster vaccination. |
|
Canaday (2021)Footnote 38 Prospective cohort US |
Circulating antibodies and NAbs were measured in 120 nursing home residents and 92 healthcare workers (HCWs) 2 weeks and 6 months after a full primary series of Comirnaty vaccination. S and RBD-IgG were measured through ELISA and NAbs through pseudovirus neutralization assay. |
At 6 months:
|
Hsu (2021)Footnote 35 Retrospective cohort US |
Dialysis patients (1567 with no prior history of COVID-19 and 299 prior COVID-19) who attended a maintenance dialysis center and had both vaccine doses (441 Comirnaty/779 Spikevax/347 Janssen*) were analyzed for their long term Spike (S) -IgG responses. SARS-CoV-2 spike antigen was measured using the chemiluminescent assay ADVIA Centaur® XP/XPT COV2G. |
Infected then vaccinated dialysis patients:
|
LTE= letter to the editor, est= estimated date based on publication submission, RBD= Receptor binding domain S = Spike HCW = Healthcare worker |
Table 4 . Systematic and rapid reviews relevant to vaccinated immunity (n=3)
Study | Method | Key outcomes |
---|---|---|
Chen (2021)Footnote 90 Systematic review NA |
A systematic review of 6 databases was conducted with a search date of July 8, 2021. PROSPERO registration no. CRD42021256932. |
|
Notarte (2021)Footnote 91 Systematic Review NA |
A systematic literature search was conducted to identify studies reporting the factors affecting humoral response of individuals who received the mRNA vaccines. Search date end of July 2021. |
|
Carr (2021)Footnote 92 Systematic Review NA |
A systematic review of immunity after vaccination in chronic kidney disease (CKD) cases including those on dialysis and transplant patients. |
|
est= Search date or publication date when search date was not available was used. |
References
- Footnote 1
-
Poland GA, Ovsyannikova IG, Kennedy RB. SARS-CoV-2 immunity: Review and applications to phase 3 vaccine candidates. Lancet. 2020 Oct 13 DOI:10.1016/s0140-6736(20)32137-1.
- Footnote 2
-
Centers for Disease Control and Prevention. COVID-19 Breakthrough Case Investigations and Reporting. Page Update Date: 2021.Accessed:April 21 .URL: https://www.cdc.gov/vaccines/covid-19/health-departments/breakthrough-cases.html
- Footnote 3
-
Muhsen, Khitam, Maimon, Nimrod, Mizrahi, Ami, Bodenneimer, Omri, Cohen, Dani, Maimon, Michal, Grotto, Itamar,Dagan, Ron. Effectiveness of BNT162b2 mRNA COVID-19 vaccine against acquisitions of SARS-CoV-2 among health care workers in long-term care facilities: A prospective cohort study. SSRN- Lancet prepublication. 2021
- Footnote 4
-
Moncunill G, Aguilar R, Ribes M, et al. Determinants of early antibody responses to COVID-19 mRNA vaccines in exposed and naive healthcare workers. medRxiv. 2021:2021.09.08.21263232. DOI:10.1101/2021.09.08.21263232.
- Footnote 5
-
Fowlkes A, Gaglani M, Groover K, et al. Effectiveness of COVID-19 vaccines in preventing SARS-CoV-2 infection among frontline workers before and during B.1.617.2 (delta) variant predominance - eight U.S. locations, December 2020-August 2021. MMWR Morb Mortal Wkly Rep. 2021 Aug 27;70(34):1167-9. DOI:10.15585/mmwr.mm7034e4.
- Footnote 6
-
Katz MA, Bron Harlev E, Chazan B, et al. COVID-19 vaccine effectiveness in healthcare personnel in six Israeli hospitals (CoVEHPI). medRxiv. 2021:2021.08.30.21262465. DOI:10.1101/2021.08.30.21262465.
- Footnote 7
-
Novazzi F, Taborelli S, Baj A, et al. Asymptomatic SARS-CoV-2 vaccine breakthrough infections in health care workers identified through routine universal surveillance testing. Ann Intern Med. 2021 Oct 19 DOI:10.7326/m21-3486.
- Footnote 8
-
Issac A, Kochuparambil JJ, Elizabeth L. SARS-CoV-2 breakthrough infections among the healthcare workers post-vaccination with ChAdOx1 nCoV-19 vaccine in the South Indian state of Kerala. medRxiv. 2021:2021.08.07.21261587. DOI:10.1101/2021.08.07.21261587.
- Footnote 9
-
Kale P, Gupta E, Bihari C, et al. Clinicogenomic analysis of breakthrough infections by SARS CoV2 variants after ChAdOx1 nCoV- 19 vaccination in healthcare workers. medRxiv. 2021:2021.06.28.21259546. DOI:10.1101/2021.06.28.21259546.
- Footnote 10
-
Laing ED, Weiss CD, Samuels EC, et al. Durability of antibody responses and frequency of clinical and subclinical SARS-CoV-2 infection six months after BNT162b2 COVID-19 vaccination in healthcare workers. medRxiv. 2021:2021.10.16.21265087. DOI:10.1101/2021.10.16.21265087.
- Footnote 11
-
Lumley SF, Rodger G, Constantinides B, et al. An observational cohort study on the incidence of SARS-CoV-2 infection and B.1.1.7 variant infection in healthcare workers by antibody and vaccination status. Clin Infect Dis. 2021 Jul 3 DOI:10.1093/cid/ciab608.
- Footnote 12
-
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 Jul 9 DOI:10.1038/s41591-021-01446-y.
- Footnote 13
-
Duerr R, Dimartino D, Marier C, et al. Dominance of alpha and iota variants in SARS-CoV-2 vaccine breakthrough infections in New York City. J Clin Invest. 2021 Aug 10 DOI:10.1172/jci152702.
- Footnote 14
-
Kustin T, Harel N, Finkel U, et al. Evidence for increased breakthrough rates of SARS-CoV-2 variants of concern in BNT162b2-mRNA-vaccinated individuals. Nat Med. 2021 Jun 14 DOI:10.1038/s41591-021-01413-7.
- Footnote 15
-
Bruxvoort K, Sy LS, Qian L, et al. Effectiveness of mRNA-1273 against delta, mu, and other emerging variants. medRxiv. 2021:2021.09.29.21264199. DOI:10.1101/2021.09.29.21264199.
- Footnote 16
-
Sheikh A, McMenamin J, Taylor B, et al. SARS-CoV-2 delta VOC in Scotland: Demographics, risk of hospital admission, and vaccine effectiveness. The Lancet. 2021 2021/06 DOI:10.1016/S0140-6736(21)01358-1.
- Footnote 17
-
Abu-Raddad L, 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 May 28 DOI:10.1093/jtm/taab083.
- Footnote 18
-
Mor, Orna, Zuckerman, Neta S., Hazan, Itay, Fluss, Ronen, Ash, Nachman, Ginish, Netanel, Mendelson, Ella, Alroy-Preis, Sharon, Freedman, Laurence,Huppert, Amit. BNT162b2 vaccination efficacy is marginally affected by the SARS-CoV-2 B.1.351 variant in fully vaccinated individuals. SSRN- Lancet prepublication. 2021
- Footnote 19
-
Thiruvengadam R, Awasthi A, Medigeshi G, et al. Cellular immune responses are preserved and may contribute to Chadox1 ChAdOx1 nCoV-19 vaccine effectiveness against infection due to SARS-CoV-2 B·1·617·2 delta variant despite reduced virus neutralisation. SSRN- Lancet prepublication. 2021
- Footnote 20
-
Shamier MC, Tostmann A, Bogers S, et al. Virological characteristics of SARS-CoV-2 vaccine breakthrough infections in health care workers. medRxiv. 2021:2021.08.20.21262158. DOI:10.1101/2021.08.20.21262158.
- Footnote 21
-
Kislaya I, Rodrigues EF, Borges V, et al. Delta variant and mRNA covid-19 vaccines effectiveness: Higher odds of vaccine infection breakthroughs. medRxiv. 2021:2021.08.14.21262020. DOI:10.1101/2021.08.14.21262020.
- Footnote 22
-
Chau NVV, Ngoc NM, Nguyet LA, et al. PMC8481205; an observational study of breakthrough SARS-CoV-2 delta variant infections among vaccinated healthcare workers in Vietnam. EClinicalMedicine. 2021 Nov;41:101143. DOI:10.1016/j.eclinm.2021.101143.
- Footnote 23
-
Tang P, Hasan MR, Chemaitelly H, et al. BNT162b2 and mRNA-1273 COVID-19 vaccine effectiveness against the delta (B.1.617.2) variant in Qatar. medRxiv. 2021:2021.08.11.21261885. DOI:10.1101/2021.08.11.21261885.
- Footnote 24
-
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 Oct 6 DOI:10.1056/NEJMoa2114114.
- Footnote 25
-
Pouwels KB, Pritchard E, Matthews PC, et al. Effect of delta variant on viral burden and vaccine effectiveness against new SARS-CoV-2 infections in the UK. Nat Med. 2021 Oct 14 DOI:10.1038/s41591-021-01548-7.
- Footnote 26
-
Barlow RS, Jian K, Larson L. Effectiveness of COVID-19 vaccines against SARS-CoV-2 infection during a delta variant epidemic surge in Multnomah County, Oregon, July 2021. medRxiv. 2021:2021.08.30.21262446. DOI:10.1101/2021.08.30.21262446.
- Footnote 27
-
COVID-19 Evidence Network to Support Decision Making - in Canada. COVID-19 Living Evidence Synthesis #6: What is the efficacy and effectiveness of available COVID-19 vaccines for variants of concern? Page Update Date: 2021.Accessed:10/22 .URL: https://www.mcmasterforum.org/find-evidence/products/project/covid-19-living-evidence-synthesis-6-what-is-the-efficacy-and-effectiveness-of-available-covid-19-vaccines-for-variants-of-concern
- Footnote 28
-
Cavanaugh AM, Spicer KB, Thoroughman D, et al. Reduced risk of reinfection with SARS-CoV-2 after COVID-19 vaccination - Kentucky, May-June 2021. MMWR Morb Mortal Wkly Rep. 2021 Aug 13;70(32):1081-3. DOI:10.15585/mmwr.mm7032e1.
- Footnote 29
-
Goel RR, Painter MM, Apostolidis SA, et al. mRNA vaccines induce durable immune memory to SARS-CoV-2 and variants of concern. Science. 2021 Oct 14:eabm0829. DOI:10.1126/science.abm0829.
- Footnote 30
-
Gimenez E, Alberola J, Torres I, et al. Evolution of SARS-CoV-2 immune responses in nursing home residents following full dose of the Comirnaty(R) COVID-19 vaccine. J Infect. 2021 Nov 2 DOI:S0163-4453(21)00542-9 [pii].
- Footnote 31
-
Ciabattini A, Pastore G, Fiorino F, et al. Evidence of SARS-CoV-2-specific memory B cells six months after vaccination with the BNT162b2 mRNA vaccine. Frontiers in Immunology. 2021;12:3751.
- Footnote 32
-
Khoury DS, Cromer D, Reynaldi A, et al. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med. 2021 May 17 DOI:10.1038/s41591-021-01377-8.
- Footnote 33
-
Earle KA, Ambrosino DM, Fiore-Gartland A, et al. Evidence for antibody as a protective correlate for COVID-19 vaccines. Vaccine. 2021 DOI:10.1016/j.vaccine.2021.05.063.
- Footnote 34
-
Remy L, Tomomori-Sato C, Conkright-Fincham J, et al. Comparison of antibody levels in response to SARS-CoV-2 infection and vaccination type in a midwestern cohort. medRxiv. 2021:2021.08.16.21262036. DOI:10.1101/2021.08.16.21262036.
- Footnote 35
-
Hsu CM, Weiner DE, Manley HJ, et al. Seroresponse to SARS-CoV-2 vaccines among maintenance dialysis patients over six months. medRxiv. 2021:2021.09.13.21263535. DOI:10.1101/2021.09.13.21263535.
- Footnote 36
-
Doria-Rose N, Suthar MS, Makowski M, et al. Antibody persistence through 6 months after the second dose of mRNA-1273 vaccine for COVID-19. N Engl J Med. 2021 Apr 6 DOI:10.1056/NEJMc2103916.
- Footnote 37
-
Sadoff J, Le Gars M, Cardenas V, et al. Durability of antibody responses elicited by a single dose of Ad26.COV2.S and substantial increase following late boosting. medRxiv. 2021:2021.08.25.21262569. DOI:10.1101/2021.08.25.21262569.
- Footnote 38
-
Canaday DH, Oyebanji O, Keresztesy D, et al. Significant reduction in humoral immunity among healthcare workers and nursing home residents 6 months after COVID-19 BNT162b2 mRNA vaccination. medRxiv. 2021:2021.08.15.21262067. DOI:10.1101/2021.08.15.21262067.
- Footnote 39
-
Havervall S, Marking U, Greilert-Norin N, et al. Impact of SARS-CoV-2 infection on longitudinal vaccine immune responses. medRxiv. 2021:2021.10.16.21264948. DOI:10.1101/2021.10.16.21264948.
- Footnote 40
-
Levin EG, Lustig Y, Cohen C, et al. Waning immune humoral response to BNT162b2 COVID-19 vaccine over 6 months. N Engl J Med. 2021 Oct 6 DOI:10.1056/NEJMoa2114583.
- Footnote 41
-
Achiron A, Mandel M, Dreyer-Alster S, et al. Humoral immune response in multiple sclerosis patients following PfizerBNT162b2 COVID19 vaccination: Up to 6 months cross-sectional study. J Neuroimmunol. 2021;361 DOI:10.1016/j.jneuroim.2021.577746.
- Footnote 42
-
Herishanu Y, Avivi I, Levi S, et al. Six months antibody persistence after BNT162b2 mRNA COVID-19 vaccination in patients with chronic lymphocytic leukemia. Blood Adv. 2021 Oct 6 DOI:10.1182/bloodadvances.2021005998.
- Footnote 43
-
Waldhorn I, Holland R, Goshen-Lago T, et al. Six month efficacy and toxicity profile of BNT162b2 vaccine in cancer patients with solid tumors. Cancer Discov. 2021 Sep 2 DOI:10.1158/2159-8290.Cd-21-1072.
- Footnote 44
-
Pegu A, O'Connell S, Schmidt SD, et al. Durability of mRNA-1273 vaccine-induced antibodies against SARS-CoV-2 variants. Science. 2021 Aug 12 DOI:10.1126/science.abj4176.
- Footnote 45
-
Israel A, Shenhar Y, Green I, et al. Large-scale study of antibody titer decay following BNT162b2 mRNA vaccine or SARS-CoV-2 infection. medRxiv. 2021:2021.08.19.21262111. DOI:10.1101/2021.08.19.21262111.
- Footnote 46
-
Salvagno GL, Henry B, Pighi L, et al. Total anti-SARS-CoV-2 antibodies measured 6 months after Pfizer-BioNTech COVID-19 vaccination in healthcare workers. SSRN- Lancet prepublication. 2021
- Footnote 47
-
Kontopoulou K, Nakas C, Ainatzoglou A, et al. Evolution of antibody titers up to 6 months post-immunization with the BNT162b2 Pfizer/BioNTech vaccine in Greece. SSRN- Lancet prepublication. 2021
- Footnote 48
-
Kertes J, Baruch Gez S, Saciuk Y, et al. Effectiveness of the mRNA BNT162b2 vaccine six months after vaccination: Findings from a large Israeli HMO. medRxiv. 2021:2021.09.01.21262957. DOI:10.1101/2021.09.01.21262957.
- Footnote 49
-
Zhong D, Xiao S, Debes AK, et al. Impact of prior SARS-CoV-2 infection on post-vaccination SARS-CoV-2 spike IgG antibodies in a longitudinal cohort of healthcare workers. medRxiv. 2021:2021.09.16.21263576. DOI:10.1101/2021.09.16.21263576.
- Footnote 50
-
Eyran T, Vaisman-Mentesh A, Dror Y, et al. The longitudinal kinetics of antibodies in COVID-19 recovered patients over 14 months. medRxiv. 2021:2021.09.16.21263693. DOI:10.1101/2021.09.16.21263693.
- Footnote 51
-
Choe PG, Kim KH, Kang CK, et al. Antibody responses 8 months after asymptomatic or mild SARS-CoV-2 infection. Emerging Infect Dis. 2020 Dec 22;27(3) DOI:10.3201/eid2703.204543.
- Footnote 52
-
El Sahly HM, Baden LR, Essink B, et al. Efficacy of the mRNA-1273 SARS-CoV-2 vaccine at completion of blinded phase. N Engl J Med. 2021 Sep 22 DOI:10.1056/NEJMoa2113017.
- Footnote 53
-
Feng S, Phillips DJ, White T, et al. Correlates of protection against symptomatic and asymptomatic SARS-CoV-2 infection. Nat Med. 2021 Sep 29 DOI:10.1038/s41591-021-01540-1.
- Footnote 54
-
Emary KRW, Golubchik T, Aley PK, et al. Efficacy of ChAdOx1 nCoV-19 (AZD1222) vaccine against SARS-CoV-2 variant of concern 202012/01 (B.1.1.7): An exploratory analysis of a randomised controlled trial. The Lancet. 2021 2021/03 DOI:10.1016/S0140-6736(21)00628-0.
- Footnote 55
-
Voysey M, Costa Clemens SA, Madhi SA, et al. Single-dose administration and the influence of the timing of the booster dose on immunogenicity and efficacy of ChAdOx1 nCoV-19 (AZD1222) vaccine: A pooled analysis of four randomised trials. Lancet. 2021 Feb 19 DOI:10.1016/s0140-6736(21)00432-3.
- Footnote 56
-
Ella R, Reddy S, Blackwelder W, et al. Efficacy, safety, and lot to lot immunogenicity of an inactivated SARS-CoV-2 vaccine (BBV152): A, double-blind, randomised, controlled phase 3 trial. medRxiv. 2021:2021.06.30.21259439. DOI:10.1101/2021.06.30.21259439.
- Footnote 57
-
Wickert DP, Almand EA, Baldovich KJ, et al. Estimates of single dose and full dose BNT162b2 vaccine effectiveness among USF academy cadets, 1 Mar - 1 May 2021. medRxiv. 2021:2021.07.28.21261138. DOI:10.1101/2021.07.28.21261138.
- Footnote 58
-
Singh C, Naik BN, Pandey S, et al. Effectiveness of COVID-19 vaccine in preventing infection and disease severity: A case control study from an eastern state of India. Epidemiol Infect. 2021 Oct 11:1-20. DOI:10.1017/s0950268821002247.
- Footnote 59
-
Ronchini C, Gandini S, Pasqualato S, et al. Lower probability and shorter duration of infections after COVID-19 vaccine correlate with anti-SARS-CoV-2 circulating IgGs. medRxiv. 2021:2021.09.24.21263978. DOI:10.1101/2021.09.24.21263978.
- Footnote 60
-
Thompson MG, Burgess JL, Naleway AL, et al. Interim estimates of vaccine effectiveness of BNT162b2 and mRNA-1273 COVID-19 vaccines in preventing SARS-CoV-2 infection among health care personnel, first responders, and other essential and frontline workers - eight U.S. locations, december 2020-march 2021. MMWR Morb Mortal Wkly Rep. 2021 Apr 2;70(13):495-500. DOI:10.15585/mmwr.mm7013e3.
- Footnote 61
-
Thompson MG, Burgess JL, Naleway A, et al. Prevention and attenuation of COVID-19 by BNT162b2 and mRNA-1273 vaccines. medRxiv. 2021:2021.06.01.21257987. DOI:10.1101/2021.06.01.21257987.
- Footnote 62
-
Andrejko KL, Pry J, Myers JF, et al. Prevention of COVID-19 by mRNA-based vaccines within the general population of California. Clin Infect Dis. 2021 Jul 20 DOI:10.1093/cid/ciab640.
- Footnote 63
-
Martínez-Baz I, Miqueleiz A, Casado I, et al. Effectiveness of COVID-19 vaccines in preventing SARS-CoV-2 infection and hospitalisation, Navarre, Spain, January to April 2021. Euro surveillance : bulletin Europeen sur les maladies transmissibles = European communicable disease bulletin. 2021;26(21) DOI:10.2807/1560-7917.ES.2021.26.21.2100438.
- Footnote 64
-
Butt AA, Chemaitelly H, Al Khal A, et al. SARS-CoV-2 vaccine effectiveness in preventing confirmed infection in pregnant women. J Clin Invest. 2021 Oct 6 DOI:10.1172/jci153662.
- Footnote 65
-
Pramod S, Govindan D, Ramasubramani P, et al. Effectiveness of Covishield vaccine in preventing COVID–19 — A test–negative case–control study. medRxiv. 2021:2021.07.19.21260693. DOI:10.1101/2021.07.19.21260693.
- Footnote 66
-
Li XN, Huang Y, Wang W, et al. Efficacy of inactivated SARS-CoV-2 vaccines against the delta variant infection in Guangzhou: A test-negative case-control real-world study. Emerg Microbes Infect. 2021 Aug 14:1-32. DOI:10.1080/22221751.2021.1969291.
- Footnote 67
-
McEwen AE, Cohen S, Bryson-Cahn C, et al. Variants of concern are overrepresented among post-vaccination breakthrough infections of SARS-CoV-2 in Washington State. Clin Infect Dis. 2021 Jun 24 DOI:10.1093/cid/ciab581.
- Footnote 68
-
Rovida F, Cassaniti I, Paolucci S, et al. SARS-CoV-2 vaccine breakthrough infections with the alpha variant are asymptomatic or mildly symptomatic among health care workers. Nat Commun. 2021 Oct 15;12(1):6032. DOI:10.1038/s41467-021-26154-6.
- Footnote 69
-
Blain H, Tuaillon E, Pisoni A, et al. Prior covid-19 and high RBD-IgG levels correlate with protection against VOC-delta SARS-CoV-2 infection in vaccinated nursing home residents. medRxiv. 2021:2021.09.21.21263880. DOI:10.1101/2021.09.21.21263880.
- Footnote 70
-
Gazit S, Shlezinger R, Perez G, et al. Comparing SARS-CoV-2 natural immunity to vaccine-induced immunity: Reinfections versus breakthrough infections. medRxiv. 2021:2021.08.24.21262415. DOI:10.1101/2021.08.24.21262415.
- Footnote 71
-
Patalon T, Gazit S, Pitzer VE, et al. Short term reduction in the odds of testing positive for SARS-CoV-2; a comparison between two doses and three doses of the BNT162b2 vaccine. medRxiv. 2021:2021.08.29.21262792. DOI:10.1101/2021.08.29.21262792.
- Footnote 72
-
Teijaro JR, Farber DL. PMC7934118; COVID-19 vaccines: Modes of immune activation and future challenges. Nat Rev Immunol. 2021 Mar 5:1-3. DOI:10.1038/s41577-021-00526-x.
- Footnote 73
-
Dai L, Gao GF. Viral targets for vaccines against COVID-19. Nat Rev Immunol. 2020 Dec 18:1-10. DOI:10.1038/s41577-020-00480-0.
- Footnote 74
-
Post N, Eddy D, Huntley C, et al. Antibody response to SARS-CoV-2 infection in humans: A systematic review. PLoS One. 2020;15(12):e0244126. DOI:10.1371/journal.pone.0244126.
- Footnote 75
-
Wang Z, Muecksch F, Schaefer-Babajew D, et al. Naturally enhanced neutralizing breadth against SARS-CoV-2 one year after infection. Nature. 2021 Jun 14 DOI:10.1038/s41586-021-03696-9.
- Footnote 76
-
Lu Z, Laing ED, Pena DaMata J, et al. Durability of SARS-CoV-2-specific T cell responses at 12-months post-infection. J Infect Dis. 2021 Oct 21 DOI:10.1093/infdis/jiab543.
- Footnote 77
-
Rank A, Tzortzini A, Kling E, et al. One year after mild COVID-19: The majority of patients maintain specific immunity, but one in four still suffer from long-term symptoms. Journal of Clinical Medicine. 2021;10(15) DOI:10.3390/jcm10153305.
- Footnote 78
-
Feng C, Shi J, Fan Q, et al. Protective humoral and cellular immune responses to SARS-CoV-2 persist up to 1 year after recovery. Nat Commun. 2021 Aug 17;12(1):4984. DOI:10.1038/s41467-021-25312-0.
- Footnote 79
-
Sonnleitner ST, Prelog M, Jansen B, et al. Maintenance of neutralizing antibodies over ten months in convalescent SARS-CoV-2 afflicted patients. Transbound Emerg Dis. 2021 May 7 DOI:10.1111/tbed.14130.
- Footnote 80
-
Kahre E, Galow L, Unrath M, et al. Kinetics and seroprevalence of SARS-CoV-2 antibodies: A comparison of 3 different assays. Sci Rep. 2021 Jul 21;11(1):14893. DOI:10.1038/s41598-021-94453-5.
- Footnote 81
-
Perez-Saez J, Zaballa ME, Yerly S, et al. PMC8261139; persistence of anti-sars-cov-2 antibodies: Immunoassay heterogeneity and implications for serosurveillance. Clin Microbiol Infect. 2021 Jul 7 DOI:10.1016/j.cmi.2021.06.040.
- Footnote 82
-
Vengesai A, Midzi H, Kasambala M, et al. A systematic and meta-analysis review on the diagnostic accuracy of antibodies in the serological diagnosis of COVID-19. Syst Rev. 2021 May 26;10(1):155. DOI:10.1186/s13643-021-01689-3.
- Footnote 83
-
Choi A, Koch M, Wu K, et al. Safety and immunogenicity of SARS-CoV-2 variant mRNA vaccine boosters in healthy adults: An interim analysis. Nat Med. 2021 DOI:10.1038/s41591-021-01527-y.
- Footnote 84
-
Chu L, Montefiori D, Huang W, et al. Immune memory response after a booster injection of mRNA-1273 for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). medRxiv. 2021:2021.09.29.21264089. DOI:10.1101/2021.09.29.21264089.
- Footnote 85
-
Pan J, Li Z, Wang L, et al. COVID-19 neutralizing antibody surveillance testing for fully vaccinated individuals during delta variant spread. medRxiv. 2021:2021.10.01.21264371. DOI:10.1101/2021.10.01.21264371.
- Footnote 86
-
Goel RR, Painter MM, Apostolidis SA, et al. mRNA vaccination induces durable immune memory to SARS-CoV-2 with continued evolution to variants of concern. bioRxiv. 2021:2021.08.23.457229. DOI:10.1101/2021.08.23.457229.
- Footnote 87
-
Liao Y, Zhang Y, Zhao H, et al. Intensified antibody response elicited by boost suggests immune memory in individuals administered two doses of SARS-CoV-2 inactivated vaccine. Emerg Microbes Infect. 2021 May 29:1-10. DOI:10.1080/22221751.2021.1937328.
- Footnote 88
-
Yue L, Xie T, Yang T, et al. A third booster dose may be necessary to mitigate neutralizing antibody fading after inoculation with two doses of an inactivated SARS-CoV-2 vaccine. J Med Virol. 2021 Sep 13 DOI:10.1002/jmv.27334.
- Footnote 89
-
Li M, Yang J, Wang L, et al. A booster dose is immunogenic and will be needed for older adults who have completed two doses vaccination with CoronaVac: A randomised, double-blind, placebo-controlled, phase 1/2 clinical trial. medRxiv. 2021:2021.08.03.21261544. DOI:10.1101/2021.08.03.21261544.
- Footnote 90
-
Chen X, Chen Z, Azman AS, et al. Neutralizing antibodies against SARS-CoV-2 variants induced by natural infection or vaccination: A systematic review and pooled meta-analysis. Clin Infect Dis. 2021 Jul 24 DOI:10.1093/cid/ciab646.
- Footnote 91
-
Notarte KI, Ver AT, Velasco JV, et al. Effects of age, sex, serostatus and underlying comorbidities on humoral response post-SARS-CoV-2 Pfizer-BioNTech vaccination: A systematic review. medRxiv. 2021:2021.10.10.21264825. DOI:10.1101/2021.10.10.21264825.
- Footnote 92
-
Carr EJ, Kronbichler A, Graham-Brown M, et al. PMC8257418; systematic review of early immune response to SARS-CoV-2 vaccination among patients with chronic kidney disease. Kidney Int Rep. 2021 Jul 6 DOI:10.1016/j.ekir.2021.06.027.
Page details
- Date modified: