Your activity: 6 p.v.

COVID-19: Vaccines

COVID-19: Vaccines
Authors:
Kathryn M Edwards, MD
Walter A Orenstein, MD
Section Editor:
Martin S Hirsch, MD
Deputy Editor:
Allyson Bloom, MD
Literature review current through: Dec 2022. | This topic last updated: Dec 16, 2022.

INTRODUCTION — Vaccines to prevent SARS-CoV-2 infection are considered the most promising approach for curbing the COVID-19 pandemic. Several COVID-19 vaccines are available globally. The World Health Organization (WHO) maintains an updated list of vaccine candidates under evaluation [1].

This topic will cover vaccines for SARS-CoV-2, with a focus on vaccines available in the United States. Other aspects related to prevention of COVID-19 are discussed in detail elsewhere. (See "COVID-19: Epidemiology, virology, and prevention", section on 'Prevention'.)

GENERAL PRINCIPLES

Pace of COVID-19 vaccine development – Although COVID-19 vaccine development has been accelerated, each vaccine that has received emergency use listing by the World Health Organization (WHO; which includes those that have been authorized or approved in the United States) has gone through the standard preclinical and clinical stages of development. Safety criteria have remained stringent; data safety and monitoring committees (DSMCs) composed of independent vaccine experts and study sponsors assess adverse events that are reported in each phase of clinical study and approve advancement to the next phase.

Calculation of vaccine efficacy – Vaccine efficacy in percent is the reduction in disease incidence among those who received vaccine versus those who received the control product and is calculated with the following formula:

([attack rate in the unvaccinated – attack rate in the vaccinated]/attack rate in the unvaccinated) x 100, often abbreviated as ([ARU – ARV]/ARU) x 100

Antigenic target – The major antigenic target for COVID-19 vaccines is the surface spike protein (figure 1). It binds to the angiotensin-converting enzyme 2 (ACE2) receptor on host cells and induces membrane fusion (figure 2) [2]. Antibodies binding to the receptor-binding domain of the SARS-CoV-2 spike protein can prevent attachment to the host cell and neutralize the virus [3].

Vaccine platforms – COVID-19 vaccines have been and are being developed using several different platforms (figure 3) [3]. Some of these are traditional approaches, such as inactivated virus or live attenuated viruses, which have been used for inactivated influenza vaccines and measles vaccine, respectively. Other approaches employ newer platforms, such as recombinant proteins (used for human papillomavirus vaccines) and vectors (used for Ebola vaccines). Some platforms, such as RNA and DNA vaccines, had never been employed in a licensed vaccine. General descriptions of the different platforms used for COVID-19 vaccines are presented in the table (table 1).

Site of delivery and immune response – COVID-19 vaccines have been demonstrated to elicit a sufficient neutralizing response to protect against COVID-19. The site of vaccine delivery may impact the character of the immune response [3]. Natural respiratory infections elicit both mucosal and systemic immune responses. Most respiratory virus vaccines, however, are administered intramuscularly (or intradermally) and elicit primarily a systemic immune response, with less robust protection in the upper respiratory mucosa than after natural infection. Some live attenuated respiratory virus vaccines can be administered intranasally, approximating natural infection, and these may elicit additional mucosal immune responses, although they typically do not induce as high of a systemic antibody response as inactivated vaccines administered systemically do [4,5]. Live attenuated COVID-19 vaccines administered to the respiratory tract are under development.

Vaccine-enhanced disease – Animal studies of certain vaccines for SARS-CoV-1 and MERS-CoV had raised concerns for enhanced disease with vaccination; after challenge with wild-type virus, some previously vaccinated animals developed non-neutralizing antibody and Th2 cell responses that were associated with eosinophilic lung inflammation [6-8]. No enhanced disease was seen in any human studies. Nevertheless, specific immunologic parameters were proposed for animal and human studies to reduce the risk of enhanced disease with COVID-19 vaccines [9]. These include criteria for neutralizing antibody and Th1-polarized cellular immune responses.

APPROACH TO VACCINATION IN THE UNITED STATES

Available vaccines — In the United States, four COVID-19 vaccines are available:

Two mRNA vaccines – Each mRNA vaccine has a monovalent formulation (antigenic target is based on the original SARS-CoV-2 strain) and a bivalent formulation (antigenic target is based on the original SARS-CoV-2 strain and the BA.4/BA.5 Omicron subvariants):

BNT162b2 (Pfizer-BioNTech COVID-19 vaccine) – This is a monovalent vaccine approved by the US Food and Drug Administration (FDA) for persons aged 12 years and older and is available under emergency use authorization (EUA) for children 6 months to 11 years of age [10].

The bivalent formulation (Pfizer-BioNTech COVID-19 vaccine, bivalent [Original and Omicron BA.4/BA.5]) is authorized as a booster dose for individuals aged 12 years or older [11].

mRNA-1273 (Moderna COVID-19 vaccine) – This is a monovalent vaccine approved by the FDA for persons aged 18 years and older and is available under EUA for children 6 months to 17 years of age [12].

The bivalent formulation (Moderna COVID-19 vaccine, bivalent [Original and Omicron BA.4/BA.5]) is authorized as a booster dose for individuals aged 18 years or older [13].

An adjuvanted recombinant protein vaccine (monovalent):

NVX-CoV2373 (Novavax COVID-19 vaccine) – This is available under EUA for individuals aged 12 years or older [14].

An adenoviral vector vaccine (monovalent):

Ad26.COV2.S (Janssen/Johnson & Johnson COVID-19 vaccine) – This is available under EUA for individuals aged 18 years or older who cannot or elect not to use an mRNA vaccine [15].

Indications and vaccine selection — We recommend COVID-19 vaccination for all individuals aged six months and older.

For individuals who are eligible for any COVID-19 vaccine, we suggest an mRNA vaccine (BNT162b2 [Pfizer COVID-19 vaccine] or mRNA-1273 [Moderna COVID-19 vaccine]) or NVX-CoV2373 (Novavax COVID-19 vaccine) rather than Ad26.COV2.S (Janssen/Johnson & Johnson COVID-19 vaccine). Extensive data supporting the use of mRNA vaccines have accumulated since their availability. Less data on safety and efficacy of NVX-CoV2373 are available, but it is also highly effective and may be an attractive option for individuals with concerns about the novelty of the mRNA vaccine platform (table 1). We reserve Ad26.COV2.S for individuals who have contraindications or no access to other COVID-19 vaccines, in agreement with the Centers for Disease Control and Prevention (CDC) and the terms of the EUA for Ad26.COV2.S (see 'Contraindications and precautions (including allergies)' below). Nevertheless, if other vaccines are not options, we recommend vaccination with Ad26.COV2.S rather than forgoing COVID-19 vaccination.

Rationale for vaccine recommendations – Vaccines available in the United States are highly effective, substantially reduce the risk of COVID-19, especially severe/critical disease, and have been associated with substantial reductions in COVID-19-associated hospitalizations and deaths [16-23], even in the context of variants that partially evade vaccine-induced immune responses. Hospitalization and mortality rates for COVID-19 have been consistently higher among unvaccinated compared with vaccinated individuals, with or without booster doses [24]. In addition to direct reductions in COVID-19-associated morbidity and mortality, vaccination has been associated with lower non-COVID-19 mortality rates, supporting evidence that COVID-19 vaccination does not increase the risk of death [25]. Details on the efficacy and safety of the individual vaccines are discussed elsewhere. (See 'Immunogenicity, efficacy, and safety of select vaccines' below and 'Waning effectiveness over time and with variants of concern' below.)

The preference for mRNA vaccines over Ad26.COV2.S is based on a more favorable risk-benefit profile with the mRNA vaccines. Ad26.COV2.S has been associated with thrombosis with thrombocytopenia and possibly Guillain-Barre syndrome, and the mRNA vaccines have been associated with myocarditis. The risks of these events are extremely small, and the benefits of all the vaccines outweigh them. However, cases of vaccine-associated thrombosis with thrombocytopenia and Guillain-Barre syndrome have been more severe with greater morbidity compared with cases of vaccine-associated myocarditis. Potential recipients of any vaccine should be aware of the specific risks, which are discussed in detail elsewhere. (See 'Specific safety concerns' below.)

Additionally, although precise comparative efficacy is uncertain because the different vaccines have not been compared directly in trials, limited evidence suggests that mRNA vaccines may be more effective than Ad26.COV2.S, including against severe infection, and mRNA-1273 may be slightly more effective than BNT162b2 [26-31].

In several observational studies, vaccine effectiveness associated with two doses of the mRNA vaccines is higher than that with one dose of Ad26.COV2.S [16,26,32-34]. As an example, in a case control study of 3689 immunocompetent adults hospitalized for COVID-19, estimated effectiveness against COVID-19-related hospitalization was 93 and 88 percent for mRNA-1273 and BNT162b2, respectively, compared with 71 percent for Ad26.COV2.S. Although the analysis adjusted for age, sex, admission date, geographic region, and race, the contribution of these and other unmeasured confounders, such as variable exposure risk, to the apparent differences in effectiveness is uncertain.

Several observational studies also suggest that mRNA-1273 vaccine effectiveness is slightly higher than that of BNT162b2, although it is unclear whether there is a clinically significant difference [26-31,35]. In a study from the United States that compared over 400,000 veterans who received either mRNA-1273 or BNT162b2, mRNA-1273 was associated with lower rates of documented infection, symptomatic COVID-19, and associated hospitalization over 24 weeks, but the absolute differences were low (differences of 1.23, 0.44, and 0.55 cases per 1000 people, respectively) [28]. Safety of the mRNA vaccines in the same cohort was largely comparable, with only small differences in serious adverse events of uncertain clinical significance [36].

Because it has not been available for as long as the other vaccines, data on NVX-CoV2373 are more limited, although large randomized trials support its efficacy; there may be a risk for myocarditis with this vaccine as well. (See 'NVX-CoV2373 (Novavax COVID-19 vaccine)' below.)

Dose and interval — Vaccine dosing and intervals are also listed in the table (table 2).

Primary vaccine series

Adults and adolescents age 12 years and older — Monovalent vaccines are used for all primary vaccine series. Dosing for individuals ≥12 years of age is as follows:

BNT162b2 (Pfizer-BioNTech COVID-19 vaccine, an mRNA vaccine) – Two intramuscular doses of 30 mcg given three to eight weeks apart (figure 4) [10].

For individuals with certain immunocompromising conditions (table 3), a third dose is given at least 28 days after the second (figure 5). (See 'Immunocompromised individuals' below.)

mRNA-1273 (Moderna COVID-19 vaccine, an mRNA vaccine) Two intramuscular doses of 100 mcg given four to eight weeks apart (figure 4) [12].

For individuals with certain immunocompromising conditions (table 3), a third dose is given at least 28 days after the second (figure 5). (See 'Immunocompromised individuals' below.)

NVX-CoV2373 (Novavax COVID-19 vaccine, an adjuvanted recombinant protein vaccine) – Two intramuscular doses of 5 mcg spike protein/50 mcg adjuvant given three to eight weeks apart (figure 4) [14].

NVX-CoV2373 is not yet authorized to be given as an additional primary series dose for individuals with immunocompromising conditions.

Ad26.COV2.S (Janssen COVID-19 vaccine, also referred to as the Johnson & Johnson vaccine, an adenoviral vector vaccine) This vaccine is only authorized for individuals ≥18 years of age and is reserved for individuals who cannot receive one of the other COVID-19 vaccines. (See 'Indications and vaccine selection' above.)

It is given as one intramuscular dose of 5x1010 viral particles (0.5 mL) (figure 4) [15]. For individuals with certain immunocompromising conditions (table 3), an additional dose with an mRNA monovalent vaccine is recommended at least four weeks later to complete a primary series (figure 5) [12]. (See 'Immunocompromised individuals' below.)

The same vaccine, if available, is generally used to complete the primary series.

Although the mRNA vaccines and NVX-CoV2373 were originally evaluated with a three- to four-week interval between the two primary series doses, extending that interval to eight weeks may be preferable for young, healthy adults who do not need to maximize protection within a shorter period of time. Specifically, an eight-week or longer interval has been associated with a lower risk of vaccine-associated myocarditis than a one month or shorter interval, which is most relevant for males 12 to 39 years old [37] (see 'Myocarditis' below). Additionally, some studies have suggested that increasing the interval between the two doses of the primary series (eg, separating them by 6 to 14 weeks rather than 3 to 4 weeks) is associated with higher titer antibody responses [38,39] and slightly greater vaccine effectiveness [40,41].

Children younger than 12 years of age — Monovalent vaccines are used for all primary vaccine series unless otherwise specified below. Dosing for children <12 years of age is as follows:

BNT162b2 (Pfizer-BioNTech COVID-19 vaccine, an mRNA vaccine)

For children 5 to 11 years of age:

-Primary series – Two intramuscular doses of 10 mcg are given three weeks to eight weeks apart. For individuals in this age group with certain immunocompromising conditions (table 3), the FDA has authorized and the CDC suggests a third primary series dose, given at least 28 days after the second (figure 5) [42]. (See 'Immunocompromised individuals' below.)

For children six months to four years of age:

-Primary series – Three intramuscular doses. The first two doses are each 3 mcg of the monovalent vaccine, given three to eight weeks apart. The third dose is with 3 mcg of the bivalent vaccine (Pfizer-BioNTech COVID-19 vaccine, bivalent), given at least eight weeks after the second [43].

mRNA-1273 (Moderna COVID-19 vaccine, an mRNA vaccine)

For children 6 to 11 years of age:

-Primary series – Two intramuscular doses of 50 mcg given four to eight weeks apart. For individuals in this age group with certain immunocompromising conditions (table 3), the FDA has authorized and the CDC suggests a third primary series dose, given at least four weeks after the second (figure 5) [42]. (See 'Immunocompromised individuals' below.)

For children six months to five years of age:

-Primary series – Two intramuscular doses of 25 mcg given four to eight weeks apart. For individuals in this age group with certain immunocompromising conditions (table 3), the FDA has authorized and the CDC suggests a third primary series dose, given at least four weeks after the second (figure 5) [42]. (See 'Immunocompromised individuals' below.)

The CDC recommends that children should receive the dose and formulation authorized for their age on the day of vaccine receipt, regardless of weight [42]. If they age into the next age group before the next primary series dose is due, they should complete the primary series with the dose and formulation authorized for the older age group. However, COVID-19 vaccine authorization for these age groups allows for such individuals to receive the entire primary series at either the dose authorized for their current age range or the dose authorized for the age they will be.

Additional considerations regarding COVID-19 vaccination in children are discussed elsewhere. (See 'Children' below.)

Booster dose

Individuals age five years and older — For all individuals ≥5 years of age who have completed a primary series of a COVID-19 vaccine (including those who have already received additional booster doses with a monovalent vaccine), the CDC recommends a booster dose with one of the bivalent mRNA vaccines at least two months after the last vaccine dose. Monovalent mRNA vaccines are no longer authorized for booster doses.

Dosing – The dose depends on the patient age:

Pfizer-BioNTech COVID-19 vaccine, bivalent (Original and Omicron BA.4/BA.5)

-For individuals ≥12 years of age: A single intramuscular 30 mcg booster dose [11].

-For individuals 5 to 12 years of age: A single intramuscular 10 mcg booster dose [44].

Moderna COVID-19 vaccine, bivalent (Original and Omicron BA.4/BA.5)

-For individuals ≥12 years of age: A single intramuscular 50 mcg booster dose [13].

-For individuals 6 to 12 years of age: A single intramuscular 25 mcg booster dose [13].

-For individuals 5 years of age: A single intramuscular 10 mcg booster dose.

The recommendation for the bivalent booster dose replaces previous booster recommendations for individuals ≥5 years old, which had included at least one monovalent booster dose for all and an additional booster dose for those who were ≥50 years old or had an immunocompromising condition.

The rationale and evidence supporting the bivalent booster vaccines are discussed elsewhere. (See 'Bivalent mRNA vaccine booster effectiveness and immunogenicity' below.)

Optimal timing – Although individuals are eligible to receive a bivalent booster dose at least two months after their last vaccine dose, the optimal timing after this interval is uncertain. We suggest that individuals who have immunocompromising conditions (table 3) or would otherwise be at high risk of severe disease (eg, because of multiple medical comorbidities) receive the booster as soon as they are eligible. We also suggest that individuals living in a region with high or increasing rates of SARS-CoV-2 transmission receive the booster once eligible. Although it is unknown if waiting longer to receive the booster dose (eg, at least three months after the last vaccine dose) improves immunogenicity or efficacy or reduces the risk of adverse effects, this could be a reasonable option for other individuals, in particular for healthy adolescent and young adult males, the population most at risk for vaccine-associated myocarditis. (See 'Myocarditis' below.)

Timing of vaccination following SARS-CoV-2 infection is discussed elsewhere. (See 'History of SARS-CoV-2 infection' below.)

For individuals 18 years or older who have received a primary vaccine series but no prior booster dose and are unable or unwilling to receive an mRNA vaccine, a single dose of NVX-CoV2373 (Novavax vaccine, 5 mcg spike protein/50 mcg adjuvant) can be used as a booster dose at least six months after the last primary series dose [42,45].

Children six months to four years old — CDC recommendations on booster doses for children 6 months to 4 years old depend on the primary series received [42,43].

For those who received mRNA-1273 (Moderna COVID-19 vaccine) – A single intramuscular 10 mcg booster dose of the bivalent vaccine (Moderna COVID-19 vaccine, bivalent) at least two months after completing the primary series.

For those who received BNT162b2 (Pfizer-BioNTech COVID-19 vaccine) – A booster dose has not been authorized if the three-dose primary series has been completed. However, for those who have not yet completed the vaccine series, a bivalent vaccine (Pfizer-BioNTech COVID-19 vaccine, bivalent) dose has been authorized as the third dose of the primary series. (See 'Children younger than 12 years of age' above.)

Support for bivalent vaccines in young children comes from immunogenicity bridging studies with monovalent vaccines and immunogenicity studies of the bivalent vaccine in other age groups. (See 'Children' below and 'Bivalent mRNA vaccine booster effectiveness and immunogenicity' below.)

Other administration issues

Technique and potential administration errors — In adults and adolescents, intramuscular vaccines are typically injected into the deltoid. Proper injection technique to reduce the risk of shoulder injury involves injection at a 90° angle into the central, thickest part of the deltoid (figure 6). (See "Standard immunizations for nonpregnant adults", section on 'Technique'.)

Additional details on administration can be found on the CDC website. The following table details CDC recommendations on the management of vaccine administration errors (table 4).

Mixing vaccine types

Completing the primary series – For the mRNA and adjuvant recombinant protein vaccines, the CDC suggests that the primary series be completed with the same vaccine, if possible [42]; there are insufficient data to inform the efficacy and safety of mixing vaccines for the primary series. If extenuating circumstances result in needing to complete the series with a different vaccine (ie, because of unavailability, new contraindication), the CDC recommends that the second dose be given at least four weeks after the first. If two different vaccine products are used to complete the series, no additional doses of a COVID-19 vaccine are recommended (table 4). The same principles apply to children aged six months to four years who are started on a three-dose primary series and, presumably, for patients with immunocompromising conditions that warrant a third mRNA vaccine dose.

Providing booster doses – In the United States, bivalent mRNA vaccines are used for booster doses. Those who received an mRNA vaccine for the primary series can use either one of the bivalent mRNA vaccines (if authorized for their age group); they do not have to use the bivalent vaccine from the same manufacturer. (See 'Dose and interval' above.)

For those who received a non-mRNA vaccine for the primary series, studies indicate robust immunogenicity with an mRNA booster dose (ie, a heterologous boost) [46-48] and greater effectiveness than with homologous boosting [49-51]. As an example, in a study of nearly 5 million United States veterans, receipt of an mRNA vaccine boost after a primary Ad26.COV2.S dose was associated with a lower risk of infection than receipt of an Ad26.COV2.S boost (adjusted rate ratio 0.49); for mRNA vaccine recipients, there was not a substantial difference in infection rates in recipients of a homologous mRNA boost versus a heterologous mRNA boost [49]. Immunogenicity studies support these findings. In a randomized trial of individuals who had received a primary Ad26.COV2.S series, an mRNA-1273 (Moderna COVID-19 vaccine) boost resulted in higher binding and neutralizing antibody levels than a BNT162b2 (Pfizer COVID-19 vaccine) boost, and levels with both mRNA vaccine boosts were higher than with another Ad26.COV2.S dose [47]. No safety concerns were identified; the frequency and duration of systemic symptoms (eg, fever, chills, myalgias) may be slightly higher with mRNA-1273 booster doses.

Immunogenicity with a heterologous bivalent vaccine boost is unknown but likely similar to the effect seen with monovalent mRNA vaccine boosting.

Studies from other countries using different vaccines to complete a series (eg, ChAdOx1 nCoV-19/ADZ122 followed by BNT162b2 or mRNA-1273) suggests a more robust, broad, and durable immune response with certain heterologous vaccine combinations, although in some cases there is a higher rate of systemic reactions (fever, fatigue, headaches, myalgias) compared with using the same vaccine for both doses [52-56].

Timing with relation to non-COVID-19 vaccines — The CDC specifies that COVID-19 vaccines can be administered at any time in relation to most other non-COVID-19 vaccines, and if needed, can be administered simultaneously with other vaccines [42].

Coadministration – When coadministered, each vaccine should be injected in different sites separated by at least one inch (and vaccines that are associated with local reactions should ideally be injected in a different limb than COVID-19 vaccines). Limited data suggest that coadministration of COVID-19 vaccines with certain other vaccines is likely safe. In a randomized trial, frequency of adverse effects and immunogenicity were largely similar when a COVID-19 vaccine (BNT162b2 or ChAdOx1) was given concomitantly with either an influenza vaccine or placebo [57].

Additional considerations with orthopoxvirus vaccination – For individuals who have received an orthopoxvirus vaccine to prevent mpox (formerly known as monkeypox), particularly adolescent or young adult males, the CDC suggests that it is reasonable to defer COVID-19 vaccination for four weeks because of the uncertain risk of myocarditis with closely spaced administration [42]. However, recent COVID-19 vaccination should not delay orthopoxvirus vaccination if indicated, given the potential health burden of mpox in the at-risk population.

Limited role for post-vaccination testing — Unless indicated to evaluate for suspected infection, there is no role for routine post-vaccination testing for COVID-19. Specifically, serologic testing following vaccination to confirm an antibody response or to determine whether to give additional doses of vaccine (eg, booster doses) is not indicated. Many serologic tests will not detect the type of antibodies elicited by vaccination. This is discussed elsewhere. (See "COVID-19: Diagnosis", section on 'Testing following COVID-19 vaccination'.)

Some side effects of vaccination overlap with symptoms of COVID-19. Systemic reactions (eg, fever, chills, fatigue, headache) that occur within the first day or two after vaccination and resolve within a day or two are consistent with a reaction to the vaccine. However, respiratory symptoms or systemic symptoms that occur after the first couple days following vaccination or that last several days could be indicative of COVID-19 and warrant testing. (See "COVID-19: Diagnosis", section on 'Choosing an initial diagnostic test'.)

Special populations

History of SARS-CoV-2 infection — We suggest eligible individuals with a history of SARS-CoV-2 infection receive a COVID-19 vaccine; pre-vaccination serologic screening to identify prior infection is not recommended [42]. All recommended primary series and booster doses should be given, even if SARS-CoV-2 infection is diagnosed after vaccination has been initiated.

Timing of vaccination – Individuals with recent, documented SARS-CoV-2 infection (including those who are diagnosed after initiating a vaccine series) should have at least recovered from acute infection and met criteria for discontinuation of isolation precautions before receiving a vaccine dose. Additionally, given the low risk of reinfection soon after prior infection, it is reasonable for individuals with SARS-CoV-2 infection to wait to receive a vaccine dose until three months after infection [42]. This applies to receipt of any primary series or booster dose. Potential reasons not to delay the vaccine dose in this population include high risk for severe infection, high rates of community transmission, and circulating variants associated with a high risk of reinfection. (See 'Other administration issues' above.)

Individuals with a history of MIS – For individuals who had SARS-CoV-2 infection complicated by multisystem inflammatory syndrome (MIS), the decision to vaccinate should be individualized and weigh the risk of exposure, reinfection, and severe disease with infection against the uncertain safety of vaccination in such individuals. Given the hypothesis that MIS is associated with immune dysregulation precipitated by SARS-CoV-2 infection, it is unknown if a SARS-CoV-2 vaccine could trigger a similar dysregulated response. Nevertheless, the benefits of vaccination may outweigh the risk among those with a history of MIS if they have recovered clinically, had MIS ≥90 days previously, and are at increased risk for SARS-CoV-2 exposure, and if the MIS was not associated with COVID-19 vaccination [42].

Vaccination is still beneficial in many patients with a history of SARS-CoV-2 infection. Vaccination appears to further boost antibody levels and cell-mediated responses in those with past infection and might improve the durability and breadth of protection [58-60]. In multiple observational studies of individuals with prior infection, vaccination has been associated with a lower risk of subsequent reinfection and hospitalization [61-68]; it has also been associated with a lower risk of breakthrough infection compared with vaccination in individuals without prior infection [69,70].

Vaccination has been associated with greater protection against hospitalization for COVID-19 compared with prior infection in some studies [71]. However, one study suggested that when Delta variant was prevalent, prior infection was associated with greater protection against COVID-19-related hospitalization than vaccination, although vaccination was still protective; estimates were age adjusted, but this study did not account for other potential confounders that may affect hospitalization risk (eg, comorbidities, exposure risk) [72].

Among individuals who have persistent symptoms following acute COVID-19, vaccination has been associated with a higher likelihood of symptom improvement compared with no vaccination, according to a systematic review by the United Kingdom Health Security Agency; however, for most individuals, symptoms remain unchanged regardless of vaccination [73].

Individuals with a history of SARS-CoV-2 may be more likely to experience local and systemic adverse effects (eg, fevers, chills, myalgias, fatigue) after a first vaccine dose than SARS-CoV-2-naïve individuals [42,74,75]. This is not a contraindication or precaution for subsequent vaccine doses.

Recent SARS-CoV-2 exposure — Individuals with a known SARS-CoV-2 exposure should receive COVID-19 vaccination, as recommended for the general population. However, such individuals who are in the community should wait until they have completed their post-exposure quarantine period to avoid inadvertent exposures to others in the event of infection [42]. Individuals who are exposed to SARS-CoV-2 in a congregate residential setting can receive COVID-19 vaccination without delay.

Given that the time needed to generate a protective immune response following vaccination exceeds the mean incubation period of SARS-CoV-2, post-exposure vaccination would likely not reduce the risk of infection following that specific exposure.

Immunocompromised individuals — We suggest that eligible individuals who have an immunocompromising condition or are taking immunosuppressive agents undergo COVID-19 vaccination. Immunogenicity and effectiveness of COVID-19 vaccines appear lower in such individuals compared with the general population; nevertheless, the potential for severe COVID-19 in this population outweighs the uncertainties. Considerations for immunocompromised patients given the potential for reduced vaccine response include the following:

Additional vaccine dose in the primary series – We agree with recommendations from the Advisory Committee on Immunization Practices (ACIP) that individuals with certain immunocompromising conditions who received a two-dose mRNA vaccine series receive a third dose (if possible, the same vaccine formulation should be used) as part of the primary vaccine series, administered at least 28 days after the second dose (figure 5) [76]; for those who received Ad26.COV2.S, a dose of an mRNA vaccine is recommended at least 28 days later [42]. The vaccine dosage should be the same as that used for all primary series doses (eg, for mRNA-1273 [Moderna COVID-19 vaccine], the 100 mcg dose). (See 'Dose and interval' above.)

Immunocompromising conditions that warrant an additional primary series dose include active use of chemotherapy for cancer, hematologic malignancies, hematopoietic stem cell or solid organ transplant, advanced or untreated HIV infection with CD4 cell count <200 cells/microL, moderate or severe primary immunodeficiency disorder, and use of immunosuppressive medications (eg, mycophenolate mofetil, rituximab, prednisone >20 mg/day for >14 days) (table 3) [77]. This list is not exhaustive; other conditions, such as impaired splenic function [78], may also warrant an additional vaccine dose.

Several other countries, including France, Germany, and Israel, have made similar recommendations [79]. Patients with immunocompromising conditions should be advised to continue other protective measures regardless of the number of vaccine doses received, as immune response may not be optimal even with three doses.

In observational studies of immunocompromised individuals, receipt of three doses of mRNA vaccines is associated with higher vaccine effectiveness than two doses [80,81]. In studies of transplant recipients who received a third dose of mRNA vaccines, seroconversion rates were higher after the additional dose, although approximately 50 to 70 percent who were seronegative after two doses remained seronegative; adverse effects were similar to those reported after prior doses [82-86]. Receipt of an additional dose following three doses of an mRNA vaccine (akin to a booster dose after a three-dose primary series) has also been associated with improved seroconversion rates [87].

Timing immunosuppressive agents and vaccination – Some expert groups recommend holding certain immunosuppressive agents around the time of vaccination or adjusting the timing of vaccination to account for receipt of such agents to try to optimize the vaccine response. As an example, for patients receiving rituximab, the American College of Rheumatology suggests scheduling vaccination so that the series is initiated approximately four weeks prior to the next scheduled rituximab dose and delaying administration of rituximab until two to four weeks after completion of vaccination, if disease activity allows [88]. (See "COVID-19: Care of adult patients with systemic rheumatic disease", section on 'COVID-19 vaccination while on immunosuppressive therapy'.)

Revaccination following certain immunosuppressing therapies – For those who received COVID-19 vaccination prior to hematopoietic cell transplantation (HCT) or chimeric antigen receptor (CAR)-T cell therapy, the CDC recommends repeat vaccination with a full primary series at least three months after the transplant or CAR-T administration [42]. For those who were vaccinated during a limited course of a B-cell-depleting therapy, repeat vaccination is suggested six months following therapy. All these patients meet criteria for receiving a three-dose primary series with the mRNA vaccines. (See "Immunizations in hematopoietic cell transplant candidates and recipients", section on 'COVID-19 vaccine'.)

Continued use of protective measures and potential pre-exposure prophylaxis – We advise immunocompromised patients to maintain personal measures to try to minimize exposure to SARS-CoV-2 (eg, masking, distancing, avoiding crowds when possible) even after they have been vaccinated because of the potential for reduced vaccine effectiveness. Household and other close contacts of immunocompromised patients should be vaccinated.

Immunocompromised patients who are at risk for suboptimal response to vaccination may be eligible for pre-exposure prophylaxis with monoclonal antibodies. This is discussed in detail elsewhere. (See "COVID-19: Epidemiology, virology, and prevention", section on 'Pre-exposure prophylaxis for selected individuals'.)

Limited role for post-vaccination serology – At this time, antibody testing is not recommended to determine response to vaccination; precise immune correlates of protection remain uncertain [42]. Furthermore, heterogeneity in the accuracy of available serologic tests complicates interpretation of results. (See 'Limited role for post-vaccination testing' above.)

Data suggest that COVID-19 vaccines are effective in many immunocompromised patients, even in the context of the Omicron variant, although they are less so than in individuals without compromised immune systems [89-96]. In a cohort study of over 1 million individuals who had received at least one mRNA vaccine in Israel, vaccine effectiveness for symptomatic COVID-19 was 75 percent (95% CI 44-88) among immunocompromised patients compared with 94 percent (95% CI 87-97) overall [89]. Lower vaccine effectiveness against hospitalization for COVID-19 in immunocompromised patients has also been suggested by smaller case-control studies [90]. In studies of individuals hospitalized with COVID-19 despite vaccination, a high proportion (eg, 40 percent) have been immunocompromised [91].

In particular, transplant recipients and individuals taking B-cell-depleting agents have a suboptimal vaccine response [95,97-101]. Among transplant recipients, use of antimetabolites (eg, mycophenolate mofetil, azathioprine) and a shorter time since transplantation have been associated with a higher rate of nonresponse.

Issues related to vaccination of specific immunocompromised populations are discussed in detail elsewhere:

(See "COVID-19: Considerations in patients with cancer", section on 'COVID-19 vaccination'.)

(See "COVID-19: Care of adult patients with systemic rheumatic disease", section on 'COVID-19 vaccination while on immunosuppressive therapy'.)

(See "COVID-19: Issues related to solid organ transplantation", section on 'Vaccination'.)

(See "Immunizations in patients with primary immunodeficiency", section on 'Issues related to SARS-CoV-2 vaccination'.)

Pregnant individuals — Vaccine recommendations for the general population extend to pregnant individuals as well. Data on safety and efficacy of COVID-19 vaccination in individuals who are pregnant or breastfeeding are discussed in detail elsewhere. (See "COVID-19: Overview of pregnancy issues", section on 'Vaccination in people planning pregnancy and pregnant or recently pregnant people'.)

Children — We recommend that eligible children undergo COVID-19 vaccination. Specifically, in the United States, the FDA has authorized and the CDC recommends BNT162b2 (Pfizer COVID-19 vaccine) or mRNA-1273 (Moderna COVID-19 vaccine) for children and adolescents aged six months and older based on evidence that efficacy and immunogenicity are as high as (or higher than) those in older individuals with rare serious adverse effects. Observational studies also indicate that vaccination is associated with reductions in COVID-19-related hospitalization, intensive care unit admission, death in adolescents, and reductions in hospitalization in younger children. For children under five years old, support for vaccine efficacy is primarily extrapolated from studies demonstrating that the neutralizing activity elicited by vaccination is comparable to levels associated with protection in older populations. These data and other data on the efficacy and safety of the specific vaccines are discussed elsewhere (see 'BNT162b2 (Pfizer-BioNTech COVID-19 vaccine)' below and 'mRNA-1273 (Moderna COVID-19 vaccine)' below). Dosing in children is also discussed elsewhere. (See 'Dose and interval' above.)

The individual benefit of COVID-19 vaccination in young children may be somewhat less than in adults because COVID-19 tends to be less severe in children than in adults. Nevertheless, the risk of the multisystem inflammatory syndrome in children (MIS-C) following acute infection, the potential for other sequelae of SARS-CoV-2 infection (eg, “long-COVID-19” and indirect effects on mental health and education), the risk of severe disease in children with underlying medical conditions, and the desire to prevent COVID-19 of any severity in children remain compelling reasons for vaccination of children [102]. Furthermore, even with the lower risk of severe disease among children, the number of COVID-19 deaths among those 6 months to 11 years old during the pandemic exceeds the prevaccination era mortality rates of infections for which childhood vaccines are routinely provided (eg, rotavirus, meningococcal disease, varicella) [103,104].

The association of mRNA COVID-19 vaccines with myocarditis, particularly among male adolescents and young adults, has raised concern about this risk in younger children. However, data suggest that the risk is not higher than baseline [105]. No cases of myocarditis thought related to vaccine were reported in the trials of BNT162b2 or mRNA-1273 in young children [106,107]. In a review of the Vaccine Adverse Event Reporting System (VAERS) following administration of approximately 8.7 million doses of BNT162b2 to children aged 5 to 11 years in the United States, there were 11 verified reports of myocarditis in this age group [108]; no cases were reported following 1.5 million doses among children six months to five years of age [109]. As with other reported cases of mRNA COVID-19 vaccine-associated myocarditis, most cases were mild and of short duration. The benefits of COVID-19 vaccination in children are considered to exceed this risk [103,110]. (See 'Myocarditis' below.)

Given the hypothesis that MIS-C is associated with immune dysregulation precipitated by SARS-CoV-2 infection, similar immune-related side effects following vaccination in children are another concern. Vaccine trials in this age group have not identified a potential signal, although rare case reports of MIS in adults following vaccination highlight the importance of monitoring for this possible adverse effect [111]. Nevertheless, some evidence suggests that vaccination may protect against MIS-C [112,113]. In a study of 102 patients aged 12 to 18 years hospitalized with MIS-C, 95 percent were unvaccinated; of the 5 patients with MIS-C who had previously received primary series of BNT162b2, none required invasive respiratory or cardiovascular support [113]. The decision to vaccinate individuals with a history of MIS-C is discussed elsewhere. (See 'History of SARS-CoV-2 infection' above.)

Most vaccines for children are delivered by private health care providers, although many are purchased using federal or other government funds. The Vaccines for Children (VFC) program is an entitlement program for all ACIP-approved vaccines for eligible children through 18 years of age [114,115]. Eligible children include those on Medicaid, those who are completely uninsured, and American Indian/Alaskan Natives. Approximately 50 percent of children are covered by the VFC. In addition, federal grants to states can be used to purchase vaccines to cover other children. Since COVID-19 vaccines are free to all persons for whom the vaccines are recommended, these funding mechanisms may be used with the COVID-19 vaccines that are licensed in children in addition to other funding sources.

Patient counseling

Expected adverse effects and their management

Common local and systemic reactions – Vaccine recipients should be advised that side effects are common and include local and systemic reactions, including pain at the injection site, ipsilateral axillary lymph node enlargement, fever, fatigue, and headache. Local and systemic side effects may reflect a robust immune response, as some studies suggest that recipients who report symptoms have slightly higher post-vaccination antibody levels than those who did not [116,117]. Nevertheless, almost all immunocompetent recipients develop sufficiently high antibody levels, regardless of side effects. Among mRNA vaccines, BNT162b2 may be associated with slightly lower rates of local and systemic reactions compared with mRNA-1273 [118]. Rates of reactions for the distinct vaccines are discussed in detail elsewhere. (See 'BNT162b2 (Pfizer-BioNTech COVID-19 vaccine)' below and 'mRNA-1273 (Moderna COVID-19 vaccine)' below and 'Ad26.COV2.S (Janssen/Johnson & Johnson COVID-19 vaccine)' below.)

Although analgesics or antipyretics (eg, nonsteroidal anti-inflammatory drugs [NSAIDs] or acetaminophen) can be taken if these reactions develop, prophylactic use of such agents before vaccine receipt is not recommended because of the uncertain impact on the host immune response to vaccination [42]. Although some data with other vaccines suggested a lower antibody response with prophylactic acetaminophen, the antibody responses to these vaccines remained in the protective range [119,120]. Aspirin is not recommended for individuals ≤18 years old because of the risk of Reye syndrome.

Because of the risk of axillary lymph node enlargement following vaccination, some expert societies suggest postponing breast cancer screening mammography for several weeks post-vaccination if it cannot be performed beforehand. (See "COVID-19: Considerations in patients with cancer", section on 'Relative to radiologic imaging'.)

Syncope – Syncope has been reported following receipt of other injectable vaccines, particularly among adolescents and young adults [121]. Monitoring is recommended for 15 to 30 minutes following COVID-19 vaccine receipt, and this may help reduce the risk of syncope-related injury. (See 'Monitoring for immediate reactions to vaccine' below.)

Rare adverse events – Very rare vaccine-associated adverse events include anaphylaxis and myocarditis with the mRNA vaccines (BNT162b2 and mRNA-1273) and unusual types of thrombotic events with thrombocytopenia and Guillain-Barre syndrome with Ad26.COV2.S. These issues are discussed in detail elsewhere. (See 'BNT162b2 (Pfizer-BioNTech COVID-19 vaccine)' below and 'mRNA-1273 (Moderna COVID-19 vaccine)' below and 'Specific safety concerns' below.)

Uncommon skin reactions have also been reported following vaccination. These are also discussed elsewhere. (See "COVID-19: Cutaneous manifestations and issues related to dermatologic care", section on 'Considerations for vaccination to prevent SARS-CoV-2 infection'.)

Other complications (including more common venous thromboembolic events without thrombocytopenia such as deep vein thrombosis or pulmonary embolism, Bell’s palsy, tinnitus) have been reported in vaccine recipients but have not been identified as causally related vaccine-associated adverse events. (See 'Immunogenicity, efficacy, and safety of select vaccines' below.)

Definition of "up to date on vaccination" — In the United States, individuals are considered “up to date” on COVID-19 vaccination if they have completed a primary series and received all recommended booster vaccines based on age and comorbidities [122]. Those who have received a primary series but are not yet eligible for the booster vaccine because of age or timing of the last vaccine dose are still considered up to date.

The CDC had also used the term “fully vaccinated” to refer to individuals who had completed the primary COVID-19 vaccine series at least two weeks prior. However, since the introduction of booster vaccines, public health recommendations have evolved to focus on whether individuals are up to date on vaccination as detailed above. The term “fully vaccinated” is probably no longer useful, as vaccination recommendations vary by age and degree of immunocompromise.

For individuals who were vaccinated in another country, the CDC notes that individuals who received a primary series with a vaccine listed for use by the World Health Organization (WHO) do not need to repeat a primary series with vaccines available in the United States. If they are eligible for an initial booster vaccine and have not yet received one, the CDC recommends administration of a booster dose (or two booster doses, depending on their age and comorbidities), at which point they are considered up to date on vaccination. (See 'Role of booster vaccinations' below.)

Post-vaccine public health precautions — Although SARS-CoV-2 infection might still occur despite vaccination, the risk is substantially lower. Recommendations on public health precautions following vaccination have evolved with new developments in the pandemic (eg, emergence of the highly transmissible Delta and Omicron variants), and the approach should be tailored to the overall rate of transmission in the community. Recommendations on mask-wearing post-exposure management are discussed in detail elsewhere. (See "COVID-19: Epidemiology, virology, and prevention", section on 'Post-exposure management'.)

EUA status of certain vaccines — In addition to standard counseling around vaccine information, vaccine providers are required to inform potential recipients when a vaccine is available under emergency use authorization (EUA). It is not necessary, however, for recipients to sign informed consent documents. (See 'Steps to vaccine availability and delivery' below.)

Contraindications and precautions (including allergies)

Contraindications – The following are the only contraindications to COVID-19 vaccination [42]:

A severe allergic reaction (eg, anaphylaxis) to a previous COVID-19 vaccine dose or to a component of the vaccine or a known (diagnosed) allergy to a component of the vaccine.

Symptoms of immediate reactions are listed on the CDC website. Isolated hives that develop more than four hours after vaccine receipt are unlikely to represent an allergic reaction to the vaccine. (See "COVID-19: Allergic reactions to SARS-CoV-2 vaccines", section on 'Delayed urticarial reactions'.)

The mRNA vaccines, BNT162b2 (Pfizer-BioNTech COVID-19 vaccine) and mRNA-1273 (Moderna COVID-19 vaccine), each contain polyethylene glycol, and NVX-CoV2373 (Novavax COVID-19 vaccine) and Ad26.COV2.S (Janssen COVID-19 vaccine, also known as the Johnson & Johnson vaccine) each contain polysorbate. Allergic reaction to polysorbate is a contraindication to NVX-CoV2373 and Ad26.COV2.S but a precaution to mRNA vaccines.

A history of thrombosis with thrombocytopenia following an Ad26.COV2.S or any other adenoviral vector COVID-19 vaccine is a contraindication to Ad26.COV2.S. Ad26.COV2.S is also not recommended for those with a history of thrombosis or thrombocytopenia that is thought to be immune mediated (including heparin-induced thrombocytopenia) or for those who developed Guillain-Barre syndrome following a prior dose of Ad26.COV2.S.

Precautions – Precautions to a specific COVID-19 vaccine include allergic reactions to other vaccines. Patients with such reactions can generally receive a COVID-19 vaccine but warrant longer post-vaccination monitoring than usual (see 'Monitoring for immediate reactions to vaccine' below):

Immediate allergic reaction to any other (non-COVID-19) vaccine or injectable therapy.

Prior immediate but nonsevere allergic reactions (eg, hives, angioedema that did not affect the airway) to a COVID-19 vaccine is a precaution (not contraindication) to that same vaccine type.

Allergy-related contraindication to one type of COVID-19 vaccine is a precaution to other types of COVID-19 vaccine because of potential cross-reactive hypersensitivity.

Allergy consultation can be helpful to evaluate suspected allergic reactions to a COVID-19 vaccine or its components and assess the risk of future COVID-19 vaccination. This is discussed in detail elsewhere. (See "COVID-19: Allergic reactions to SARS-CoV-2 vaccines", section on 'Possible anaphylaxis'.)

Caution is also warranted for those with a history of myocarditis or pericarditis following a COVID-19 vaccine, MIS, or Guillain-Barre syndrome. These issues are discussed elsewhere. (See 'Myocarditis' below and 'History of SARS-CoV-2 infection' above and 'Guillain-Barre syndrome' below.)

Caution may be warranted prior to administering any vaccine in certain rare but life-threatening conditions, such as acquired thrombotic thrombocytopenia purpura and capillary leak syndrome, exacerbations of which have been reported following COVID-19 vaccination [123,124]. (See "Immune TTP: Management following recovery from an acute episode and during remission", section on 'Vaccinations' and "Idiopathic systemic capillary leak syndrome", section on 'Prodromal symptoms and triggers'.)

History of thromboembolic disease is not a contraindication to vaccination with mRNA vaccines or NVX-CoV2373. Although very rare cases of unusual types of thrombosis associated with thrombocytopenia have been reported following vaccination with both ChadOx1 nCoV-19/AZD1222 (AstraZeneca COVID-19 vaccine) and Ad26.COV2.S, there has not been a concerning signal for this type of thrombotic complication with mRNA COVID-19 vaccines. Furthermore, there is no evidence that classic risk factors for thrombosis (eg, thrombophilic disorders or prior history of venous thromboembolism not associated with thrombocytopenia) increase the risk for this rare adverse event [125], and individuals with these can receive any approved or authorized COVID-19 vaccine. (See 'Thrombosis with thrombocytopenia' below and "COVID-19: Vaccine-induced immune thrombotic thrombocytopenia (VITT)", section on 'Prevention (common questions)'.)

Other reactions or conditions that are neither precautions nor contraindications include:

Late local reactions characterized by a well-demarcated area of erythema appearing at the injection site approximately a week after mRNA COVID-19 vaccination have been reported, with recurrence occurring in some individuals after repeat vaccination [126]. This may occur more frequently with mRNA-1273 than with BNT162b2 [127]. This type of reaction is not a contraindication to vaccination, and individuals who experience this after the initial mRNA vaccine dose can proceed with the second dose as scheduled [42]. (See "COVID-19: Allergic reactions to SARS-CoV-2 vaccines", section on 'Late local reactions'.)

Facial swelling in areas previously injected with cosmetic dermal fillers has also been rarely reported following vaccination with the mRNA COVID-19 vaccines. Dermal fillers are not a contraindication to COVID-19 vaccination, and no specific precautions are recommended [42]. However, it is reasonable to advise individuals with dermal fillers of the possibility of post-vaccination swelling. This is discussed elsewhere. (See "COVID-19: Cutaneous manifestations and issues related to dermatologic care", section on 'Soft tissue fillers'.)

Anticoagulation is not a contraindication to vaccination; excess bleeding is unlikely with intramuscular vaccines in patients taking anticoagulants [128]. Such patients can be instructed to hold pressure over the injection site to reduce the risk of hematoma. (See "Standard immunizations for nonpregnant adults", section on 'Patients on anticoagulation'.)

Monitoring for immediate reactions to vaccine — All individuals should be monitored for immediate vaccine reactions following receipt of any COVID-19 vaccine.

The following warrant monitoring for 30 minutes:

Precautions to the administered vaccine (immediate reaction to any vaccine or injectable therapy; contraindication to the other vaccine type) (see 'Contraindications and precautions (including allergies)' above)

History of anaphylaxis due to any cause

All other individuals are monitored for 15 minutes.

Vaccines should be administered in settings where immediate allergic reactions, should they occur, can be appropriately managed. Recognition and management of anaphylaxis are discussed in detail elsewhere (table 5). (See "Anaphylaxis: Acute diagnosis" and "Anaphylaxis: Emergency treatment".)

Anaphylaxis has been reported following administration of both mRNA COVID-19 vaccines [129]. Following the first several million doses of mRNA COVID-19 vaccines administered in the United States, anaphylaxis was reported at approximate rates of 4.5 events per million doses [130-132]. The vast majority of these events occurred in individuals with a history of allergic reactions and occurred within 30 minutes. The mechanism for the anaphylaxis is under investigation and has not been determined. Some suggest that it is IgE mediated, with polyethylene glycol as the inciting antigen. However, other complement-mediated mechanisms have been suggested in individuals without a previous history of allergy. Evaluation of patients with possible anaphylaxis following COVID-19 vaccination is discussed elsewhere. (See "COVID-19: Allergic reactions to SARS-CoV-2 vaccines", section on 'Possible anaphylaxis'.)

Reporting of adverse events — To facilitate ongoing safety evaluation, vaccine providers are responsible for reporting vaccine administration errors, serious adverse events associated with vaccination, cases of multisystem inflammatory syndrome (MIS), and cases of COVID-19 that result in hospitalization or death through the Vaccine Adverse Event Reporting System (VAERS). (See 'Ongoing safety assessment' below.)

Details on the efficacy and safety of available COVID-19 vaccines are discussed elsewhere. (See 'Immunogenicity, efficacy, and safety of select vaccines' below.)

APPROACH TO VACCINATION IN OTHER COUNTRIES — Various vaccines are available in different countries. A list of vaccines that have been authorized in at least one country can be found at covid19.trackvaccines.org/vaccines. Data on some of these vaccines can be found elsewhere. (See 'Immunogenicity, efficacy, and safety of select vaccines' below.)

Dosing schedules vary by vaccine. Additionally, different countries may have specific recommendations for vaccine use. Clinicians should refer to local guidelines for vaccine recommendations in their location. (See 'Society guideline links' below.)

Several countries had paused use of ChAdOx1 nCoV-19/AZD1222 to investigate scattered reports of thromboembolic events; many have since resumed use, in some cases with age restrictions. This is discussed in detail elsewhere. (See 'Thrombosis with thrombocytopenia' below.)

IMMUNOGENICITY, EFFICACY, AND SAFETY OF SELECT VACCINES — Selected vaccines that are available for use in different countries are described here (table 2). They represent different vaccine approaches, including RNA vaccines, replication-incompetent vector vaccines, recombinant protein vaccines, and inactivated vaccines; the general features of these different platforms are described elsewhere. (See 'General principles' above.)

Immunogenicity, efficacy, and safety of specific vaccines are discussed below. General issues related to breakthrough infections, impact on transmission, effectiveness against variants of concern, and duration of effect are discussed elsewhere. (See 'Ongoing safety assessment' below.)

BNT162b2 (Pfizer-BioNTech COVID-19 vaccine) — This mRNA vaccine is delivered in a lipid nanoparticle to express a full-length spike protein (table 2). Clinical use of the vaccine is discussed elsewhere. (See 'Approach to vaccination in the United States' above.)

Efficacy and immunogenicity – Randomized trials in children and adults demonstrate a substantially reduced risk of symptomatic and severe COVID-19 in the first several months after BNT162b2 vaccination. In large placebo-controlled trials, vaccine efficacy of the two-dose primary series in preventing symptomatic COVID-19 at a median of two-month follow-up was 95 percent (95% CI 90.3-97.6) for individuals aged 16 years or older [133,134], 100 percent (95% CI 75.3-100) for individuals aged 12 to 15 years [135], and 91 percent for individuals aged 5 to 11 years [136]. Among adults ≥65 years of age who had other medical comorbidities or obesity, vaccine efficacy was 91.7 percent (95% CI 44.2-99.8). On longer follow-up, vaccine efficacy remained high but slightly decreased to 90 percent at two to four months post-vaccination and 84 percent at four to six months [137]. Of 30 severe infections (ie, with hypoxia, organ dysfunction, or critical illness) among nearly 50,000 trial participants over six months, only 1 occurred in a vaccinated individual. In a small trial of children aged six months to four years, vaccine efficacy was 80.4 percent (95% CI 14.1-96.7), although the estimate was uncertain because of the low number of cases overall [107]. Two participants were seen in the emergency department or hospitalized with COVID-19; both were vaccine recipients who also had coinfection with other respiratory viruses.

Observational data from various countries following their national roll-outs of BNT162b2 support the trial findings in adults and adolescents [16-19,138-149]. Specifically, BNT162b2 has been associated with approximately 90 percent or higher vaccine effectiveness in preventing COVID-19-related hospitalization, intensive care unit admissions, and death among adolescents and adults. Some, but not all observational data suggest that vaccine effectiveness in children aged 5 through 11 years may be lower than that among older adolescents, but it is unclear how much of that difference is related to reduced vaccine effectiveness in general against the Omicron variant, which dominated soon after introduction of vaccine for the younger children; vaccination still substantially reduces COVID-19-associated hospitalizations in this age group, even in the context of Omicron prevalence [150-159].

Vaccine effectiveness wanes over time and may be decreased in protecting against infection with certain SARS-CoV-2 variants, although protection against severe disease due to variants remains substantial. These issues are discussed in detail elsewhere. (See 'Waning effectiveness over time and with variants of concern' below.)

These efficacy data are consistent with evidence from immunogenicity studies that demonstrated robust binding and neutralizing antibody responses with BNT162b2, with some variability by age [135,136,160]. Responses in participants ≥65 years old were generally lower than in younger subjects, but still comparable or higher than titers in convalescent plasma. For children under five years old, three doses (of a lower vaccine dose) were necessary to elicit neutralizing antibody levels comparable to those in older individuals after two doses [107].

Neutralizing antibody titers decline with time following BNT162b2 vaccination in adults; in one study, steeper declines in neutralizing titers over six months were observed among males, individuals ≥65 years old, and immunocompromised individuals [161]. Neutralizing activity is lower against the Delta variant (B.1.617.2) [162-164] and substantially lower against the Omicron variant (B.1.1.529) compared with activity against previously circulating strains.

Preliminary data presented in a press release from the manufacturer indicated that in children aged six months to five years, three doses of a low-dose BNT162b2 formulation resulted in antibody responses similar to those seen in young adults following two standard doses [165]. Additional details are pending.

Common side effects – Local and systemic adverse effects are relatively common, particularly after the second dose; most are of mild or moderate severity (ie, do not prevent daily activities) and are limited to the first two days after vaccination [118,132,133]. Injection site reactions (mainly pain, but also redness, swelling, and pruritus) occur in approximately 65 percent; fatigue, headache, and myalgias in approximately 40 to 50 percent; and fevers, chills, and joint pain in approximately 20 percent [118]. Rates are slightly higher among adolescents aged 12 through 15 years and slightly lower among children aged 5 through 11 years [135,136]. Among young children, irritability, crying, drowsiness, and loss of appetite were common; febrile seizures were rare [107]. Local and systemic reactions occur less frequently among recipients aged 65 years or older but are still relatively common.

Serious adverse effects – Myocarditis and pericarditis, mainly in male adolescents and young adults, have been reported more frequently than expected following receipt of BNT162b2. This is discussed in detail elsewhere. (See 'Myocarditis' below.)

Anaphylaxis following vaccination has been reported at an approximate rate of 5 events per 1 million doses; 80 percent of anaphylaxis cases have occurred in individuals with a history of allergic reactions and 90 percent occurred within 30 minutes [130]. Other reported allergic reactions included pruritus, rash, scratchy sensations in the throat, and mild respiratory symptoms [166]. (See "COVID-19: Allergic reactions to SARS-CoV-2 vaccines", section on 'mRNA vaccines'.)

Other major adverse events have not been consistently associated with BNT162b2 receipt [167]. Rare cases of Bell's palsy were noted in the phase III trial in adults (four in vaccine and zero in placebo recipients) [133]; however, the rate did not exceed background rates found in the general population (15 to 30 cases per 100,000 people per year), and post-vaccine monitoring has not identified an association between vaccination and Bell’s palsy [130]. In a large cohort study from Israel, BNT162b2 receipt was most strongly associated with myocarditis, lymphadenopathy, appendicitis, and herpes zoster [168].

mRNA-1273 (Moderna COVID-19 vaccine) — This messenger RNA (mRNA) vaccine was one of the first vaccines for SARS-CoV-2 to be produced; it was developed and administered to humans within two months of publication of the SARS-CoV-2 genomic sequence. The vaccine utilizes mRNA delivered in a lipid nanoparticle to express a full-length spike protein (table 2). Clinical use of the vaccine is discussed elsewhere. (See 'Approach to vaccination in the United States' above.)

Efficacy and immunogenicity Randomized trials in adults demonstrate a substantially reduced risk of symptomatic and severe COVID-19 in the first several months after mRNA-1273 vaccination. In a large placebo-controlled trial, vaccine efficacy of the two-dose primary series in preventing symptomatic COVID-19 at a median of two-month follow-up was 94.1 percent (95% CI 89.3-96.8) among adults 18 years or older [169]. Among adults ≥65 years of age, vaccine efficacy was 86.4 percent (95% CI 61.4-95.5). After a median follow-up of 5.2 months, vaccine efficacy was 93.2 percent for symptomatic infection (9.6 versus 136.6 cases per 100 person-years with placebo) and 98.2 percent for severe disease (ie, with hypoxia, organ dysfunction, or critical illness; 2 versus 106 cases with placebo) [170]. Limited trial data also suggest that vaccine efficacy against symptomatic COVID-19 is high in children aged 12 to 17 years (100 percent, 95% CI 28.9-nonevaluable) and aged 6 to 11 years (69 percent, 95% CI -131.4-95.8), although the estimates of effect were uncertain because of the low number of cases [106]. Efficacy was lower (41.5 percent, 95% CI 23.8-55) in a trial of younger children that was performed when Omicron variant was circulating, although this estimate is largely consistent with observational data on vaccine effectiveness against Omicron in adults. There were no severe COVID-19 cases in any of the trials of children.

Observational data evaluating vaccine effectiveness also support the trial findings in adults [16,146,147,149,171-173]. Specifically, mRNA-1273 has been associated with approximately 90 percent or higher vaccine effectiveness in preventing COVID-19-related emergency visits, hospitalization, intensive care unit admission, and death.

Vaccine effectiveness wanes over time and may be decreased in protecting against infection with certain SARS-CoV-2 variants, although protection against severe disease due to variants remains substantial. These issues are discussed in detail elsewhere. (See 'Waning effectiveness over time and with variants of concern' below.)

These efficacy data are consistent with evidence from immunogenicity studies that demonstrated robust binding and neutralizing antibody responses with mRNA-1273 in adults of all ages [174,175]. Immunogenicity in children or adolescents aged 6 months to 5 years (with a quarter-dose), aged 6 to 11 years (with a half-dose), and aged 12 to 17 years (with a standard dose) is comparable to or higher than that seen in young adults [176-178]. Over six months, antibody titers decline slightly but remain high and neutralizing activity persists [179]. Vaccination with mRNA-1273 is associated with higher antibody titers after the second dose compared with BNT162b2 [180,181]. Neutralizing activity is lower against Delta (B.1.617.2) [163] and substantially lower against Omicron (B.1.1.529) compared with activity against previously circulating strains.

Common side effects – Local and systemic adverse effects are relatively common, particularly after the second dose; most are of mild or moderate severity (ie, do not prevent daily activities or require pain relievers) and are limited to the first two days after vaccination [118,132,182]. Injection site reactions (mainly pain, but also redness, swelling, and pruritus) occur in approximately 75 to 80 percent; fatigue, headache, and myalgias in approximately 50 to 60 percent; and fevers, chills, and joint pain in approximately 30 to 40 percent [118]. Local and systemic reactions occur less frequently among recipients 65 years or older but were still relatively common. Among young children, irritability, crying, sleepiness, and loss of appetite were common; febrile seizures were rare [106].

Severe adverse effects – Myocarditis and pericarditis, mainly in male adolescents and young adults, have been reported more frequently than expected following receipt of mRNA-1273. This is discussed in detail elsewhere. (See 'Myocarditis' below.)

Anaphylaxis following vaccination has been reported at an approximate rate of 2.8 events per 1 million doses; 86 percent of anaphylaxis cases have occurred in individuals with a history of allergic reactions, and 90 percent occurred within 30 minutes [130,166]. (See "COVID-19: Allergic reactions to SARS-CoV-2 vaccines", section on 'mRNA vaccines'.)

In trials among adults, there were rare cases of Bell's palsy that were considered potentially related to vaccination (three in the vaccine and one in the placebo group). However, the rate did not exceed the background rate in the general population (15 to 30 cases per 100,000 people per year), and post-vaccine monitoring has not identified an association between vaccination and Bell's palsy [130].

No other major vaccine-associated adverse events have been identified in post-vaccine surveillance [167].

NVX-CoV2373 (Novavax COVID-19 vaccine) — This is a recombinant protein subunit vaccine composed of trimeric spike glycoproteins and a potent Matrix-M1 adjuvant (table 2). Clinical use of the vaccine is discussed elsewhere. (See 'Approach to vaccination in the United States' above.)

Efficacy and immunogenicity – In a phase III efficacy trial in the United States and Mexico, NVX-CoV2373 had 90.4 percent (95% CI 82.9-94.6) efficacy in preventing symptomatic COVID-19 in seronegative individuals aged 18 to 84 years [183]. The four severe cases occurred in the placebo group. Among participants 65 year of age or older, the estimate of vaccine efficacy was lower, but was also less certain because of the small number of cases in this subgroup (78.6 percent, 95% CI -16.64 to 96). Similar vaccine efficacy (89.7 percent, 95% CI 80.2-94.6) was reported in a phase III trial in the United Kingdom [184].

Safety and side effects – Local and systemic adverse effects are relatively common, particularly after the second dose, with a median time to onset of two days and median durations of two to three days; most are of mild or moderate severity (ie, do not prevent undertaking daily activities or require pain relievers). The most common systemic reactions were fatigue/malaise, headache, and myalgias. Systemic reactions were more common among recipients under 65 years of age.

In the phase III efficacy trial, serious adverse event rates in the vaccine and placebo group were similar [183,185]. However, there were six cases of myocarditis/pericarditis in the vaccine group, five of which occurred within two weeks of vaccine receipt, and four of which occurred in young males, consistent with the pattern of myocarditis associated with mRNA vaccines (see 'Myocarditis' below). There were also small numerical excesses of certain rare adverse events in the vaccine groups (including uveitis, thromboembolic events, cholecystitis, and cardiomyopathy), but the limited data cannot establish or rule out a causal relationship.

Ad26.COV2.S (Janssen/Johnson & Johnson COVID-19 vaccine) — This vaccine is based on a replication-incompetent adenovirus 26 vector that encodes a stabilized spike protein (table 2). Clinical use of the vaccine is discussed elsewhere. (See 'Approach to vaccination in the United States' above.)

Efficacy and immunogenicity – Randomized trials in adults demonstrate a substantially reduced risk of symptomatic and severe COVID-19 in the first several months after Ad26.COV2.S vaccination. In a large placebo-controlled trial, vaccine efficacy of the one-dose primary series in preventing moderate to severe/critical COVID-19 (which included patients with pneumonia, dyspnea, tachypnea, or at least two symptoms of COVID-19) at a median of two-month follow-up was 66.9 percent efficacy (95% CI 59.0-73.4) in adults age 18 years or older [186]. Vaccine efficacy against severe/critical infection (ie, with hypoxia, organ dysfunction, or critical illness) trended higher at 78 and 85 percent after 14 and 28 days post-vaccination. Efficacy estimates after a median of four months follow-up were 56.3 percent (95% CI 51.3-60.8) for at least moderate COVID-19 and 74.6 percent (95% CI 64.1-82.1) for severe/critical COVID-19 [187].

Observational data evaluating vaccine effectiveness largely support the trial findings; a single dose of Ad26.COV2.S has been associated with vaccine effectiveness of 67 to 75 percent against COVID-19-related emergency care and hospitalization and 83 percent against COVID-19-related death [16,188,189]. These efficacy data are consistent with evidence from immunogenicity studies that demonstrated post-vaccination binding and neutralizing antibody responses that overlapped with but were slightly lower than those in convalescent plasma [190,191]. These neutralizing responses are largely stable over eight months with both one- and two-dose regimens [192], in contrast to the neutralizing antibody levels following mRNA vaccination, which wane over time (although remain higher than after Ad26.COV2.S) [193]. Neutralizing activity is also retained against the Delta (B.1.617.2) variant at only a slightly lower level than against previously circulating strains but is substantially reduced against Omicron (B.1.1.529).

Waning vaccine effectiveness and impact of SARS-CoV-2 variants are discussed in detail elsewhere. (See 'Waning effectiveness over time and with variants of concern' below.)

Safety and side effects – Local and systemic adverse effects are relatively common; most are of mild or moderate severity (ie, do not prevent undertaking daily activities or require pain relievers) and most commonly occur the first day after vaccination [194]. Among over 330,000 vaccine recipients in the United States who responded to post-vaccination surveys, 76 percent reported at least one systemic reaction and 61 percent at least one injection site reaction in the first week. The most common systemic reactions were fatigue, pain, and headache. Anxiety-related events, including tachycardia, hyperventilation, light-headedness, and syncope, have also been reported following Ad26.COV2.S administration [195].

In the phase III efficacy trial, serious adverse event rates in the vaccine and placebo group were similar [186]. There were more cases of thromboembolic events (11 versus 3), tinnitus (6 versus 0), and seizures (4 versus 1) among vaccine compared with placebo recipients, but the numbers of events were too few to determine whether there is a causal association with vaccination. In a report of over 400,000 health care workers who received Ad26.COV2.S in South Africa, there were two cases of thrombosis with thrombocytopenia and four cases of Guillain-Barre syndrome following vaccination; the incidence of these did not exceed the expected incidence in the general population [196]. However, other studies suggest that the vaccine is associated with a specific syndrome of thrombosis with thrombocytopenia and is possibly associated with Guillain-Barre syndrome; these are discussed in detail elsewhere. (See 'Thrombosis with thrombocytopenia' below and 'Guillain-Barre syndrome' below.)

ChAdOx1 nCoV-19/AZD1222 (University of Oxford, AstraZeneca, and the Serum Institute of India) — This vaccine is based on a replication-incompetent chimpanzee adenovirus vector that expresses the spike protein. It is given intramuscularly as two doses. The World Health Organization (WHO) recommends that the two doses be given 8 to 12 weeks apart [197].

Efficacy and immunogenicity – Randomized trials in adults demonstrate a substantially reduced risk of symptomatic COVID-19 in the first several months after vaccination. In large placebo-controlled trials, vaccine efficacy of a two-dose primary series in preventing symptomatic COVID-19 at a median of two-month follow-up was 70 to 76 percent (95% CI 54.8-80.6) at or after 14 days following the second dose [198,199]. Additional analysis of this trial suggested that receipt of the second dose at 12 weeks or later was associated with higher vaccine efficacy than receipt at <6 weeks (81 versus 55 percent) [200]. These findings lend support to extending the time interval between the first and second dose to 12 weeks.

Observational data from various countries following their national roll-outs of ChAdOx1 nCoV-19/AZD1222 also support the trial findings [18,201], although they suggest that effectiveness, even against severe infection, wanes over time [202]. Waning vaccine effectiveness and impact of SARS-CoV-2 variants are discussed in detail elsewhere. (See 'Waning effectiveness over time and with variants of concern' below.)

These efficacy data are consistent with evidence from immunogenicity studies that demonstrated robust binding and neutralizing antibody responses in vaccine recipients [203-205]. The Delta (B.1.617.2) and Omicron (B.1.1.529) variants evade immune responses in some vaccinated individuals [164,206].

Safety and side effects – In earlier-phase trials, fatigue, headache, and fever were relatively common after vaccine receipt and were severe in up to 8 percent of recipients [203]. In the phase III trial, there were two cases of transverse myelitis in ChAdOx1 nCoV-19 vaccine recipients [198]. One was thought to be possibly related to vaccination and was described as an idiopathic, short-segment spinal cord demyelination; the other was in a participant with previously unrecognized multiple sclerosis and thought to be unrelated to the vaccine. The vaccine also may be associated with an extremely small risk of thrombotic events associated with thrombocytopenia, which is discussed in detail elsewhere. (See 'Thrombosis with thrombocytopenia' below.)

The general thromboembolic risk with ChadOx1 nCoV-19/AZD1222 is uncertain. Some analyses have suggested that the total rate of thromboembolic events following vaccination is lower than that expected based on the background rate in the general population [207,208]. However, separate analyses from Europe suggest a slightly higher total rate of thromboembolic events (including pulmonary embolism) following ChadOx1 nCoV-19/AZD1222 than expected [209,210].

Other vaccines — Details on select vaccines that are available internationally are presented below. A list of vaccines that have been authorized in at least one country can be found at covid19.trackvaccines.org/vaccines.

Convidencia (CanSino Biologics) – This vaccine is based on a replication-incompetent adenovirus 5 vector that expresses the spike protein. It is given as a single intramuscular dose. In early clinical trials, both pre-existing immunity to adenovirus 5 and older age were associated with lower titers of binding and neutralizing antibodies following vaccination; this may limit its utility in locations where pre-existing immunity is prevalent [211]. In a randomized phase III trial, vaccine efficacy was 57.5 percent (95% CI 39.7-70.0) for symptomatic infection and 91.7 percent (95% CI 36.1-99.0) for severe disease [212]. This vaccine is available in China and some other countries, including Mexico and Pakistan.

Gam-COVID-Vac/Sputnik V (Gamaleya Institute) – This is a vaccine developed in Russia that uses two replication-incompetent adenovirus vectors that express a full-length spike glycoprotein (table 2). The vaccine is given intramuscularly as an initial adenovirus 26 vector dose followed by an adenovirus 5 vector boosting dose 21 days to 3 months later [213]. This vaccine is available in Russia and several other countries, including Mexico. According to interim analysis of a phase III trial, this vaccine had 91.6 percent (95% CI 85.6-95.2) efficacy in preventing symptomatic COVID-19 at the time of the second dose [214]. All 20 cases of severe COVID-19 that occurred 21 days after the first dose were in the placebo group. Local and systemic flu-like reactions were more common in the vaccine group, at rates of 15 and 5 percent, respectively. No serious adverse events were deemed related to vaccine.

Covilo/BBIBP-CorV (Sinopharm) – This is an inactivated, whole-virus vaccine with an aluminum hydroxide adjuvant. It is given intramuscularly in two doses 28 days apart. In a phase III efficacy trial, vaccine efficacy was estimated as 78 percent (95% CI 65-86) compared with an alum-only placebo [215]. Only two severe cases occurred, both in the placebo group. Systemic and injection site reactions occurred at similar frequencies with vaccine or placebo (eg, pain in 20 to 27 percent, headache in 13 percent, fatigue in 11 percent). This vaccine is available in China and some other countries, including the United Arab Emirates and Hungary.

CoronaVac (Sinovac) – This inactivated COVID-19 vaccine was developed in China; it has an aluminum hydroxide adjuvant. The vaccine is given intramuscularly in two doses 28 days apart. According to interim results of a phase III trial in Turkey, vaccine efficacy was 83.5 percent (95% CI 65.4–92.1) [216]; however, lower efficacy rates have been reported in small trials from different countries [217,218]. In an observational study that included over 10 million individuals in Chile, estimated vaccine effectiveness was 70 percent for preventing COVID-19 and 86 to 88 percent for preventing hospital admission or death [219]; a subsequent study in Brazil reported lower vaccine effectiveness among adults older than 70 years in the context of prevalent Gamma variant (47, 56, and 61 percent against COVID-19, hospitalization, and death, respectively) [220]. This vaccine is available in China and some other countries, including Brazil, Chile, Indonesia, Mexico, and Turkey.

Covaxin (Bharat Biotech/Indian Council of Medical Research) – This inactivated COVID-19 vaccine (also called BBV152) was developed and is being used in India; it has an aluminum hydroxide and a toll-like receptor agonist adjuvant. It is given intramuscularly in two doses 29 days apart. In a randomized trial, vaccine efficacy against symptomatic COVID-19 was 78 percent (95% CI 65-86); there was 1 case of severe COVID-19 in the vaccine group and 15 in the placebo group [221]. Serious adverse events were not deemed related to vaccine except for one possibly related case of immune thrombocytopenic purpura.

ZyCoV-D (Zydus Cadila) – This is the first DNA COVID-19 vaccine made available, first authorized in India in August 2021 [222]. A needleless device delivers the vaccine subcutaneously with a high-pressure stream. In a trial of 28,000 participants aged 12 years or older, vaccine efficacy against symptomatic COVID-19 was 67 percent (95% CI 47.6-80.7) following three doses, each given 28 days apart; only one severe case of COVID-19, in the placebo group, occurred [223].

GENERAL EFFICACY ISSUES

Role of booster vaccinations

Waning effectiveness over time and with variants of concern — Although the initial clinical trials reported extremely high efficacy rates of COVID-19 vaccines (in particular with mRNA vaccines) in preventing laboratory-confirmed symptomatic infection, their observed effectiveness against infection has decreased over time because of waning immunity and immune evasion by certain circulating SARS-CoV-2 variants. Decreases in observed vaccine effectiveness may also be related to overall decreases in the risk of severe infection because of a higher prevalence of prior infection (which also provides protection against severe infection) as well as variants associated with milder infection. Although vaccine effectiveness against severe disease has also decreased over time and in the setting of Omicron subvariants, COVID-19 vaccines continue to provide good levels of protection against severe disease; the risk of hospitalization and death associated with COVID-19 has consistently remained higher among unvaccinated individuals [23,24].

Waning effectiveness – Multiple observational studies have suggested that vaccine protection against SARS-CoV-2 infection wanes over time (after both primary series and booster vaccinations) in children and adults [33,224-233]. Protection against hospitalization and death also wanes somewhat, but less than protection against infection [234-241].

As an example, in a study of statewide data in North Carolina that included over 10 million adults, adjusted mRNA vaccine effectiveness at seven months following the primary series was 54 to 70 percent against infection, 86 to 90 percent against hospitalization, and 90 to 93 percent against death [239]. At 12 months, the same measures were 38 to 47 percent, 60 to 65 percent, and 70 to 75 percent. Similarly, vaccine effectiveness of an mRNA booster dose compared with the primary series peaked at about one month and waned over four to six months, but the booster dose continued to be associated with protection against hospitalization and death. Another study suggested that a primary series and booster dose were associated with a 60 percent reduction in risk of hospitalization at five months [241].

Attenuated effectiveness against certain variants of concern – Several SARS-CoV-2 variants that are concerning for their potential for immune escape have emerged over the course of the pandemic (table 6). For the Omicron variant and its sublineages (BA.1, BA.2, BA.2.12.1, BA.4, and BA.5), COVID-19 vaccines remain effective in preventing severe disease, but effectiveness in preventing symptomatic infection is attenuated.

Observational studies consistently suggest that vaccination has substantially reduced effectiveness against symptomatic infection with Omicron subvariants compared with other variants and wanes after several months. Effectiveness against severe disease (as reflected by hospitalization) has remained relatively high, particularly among those who received a booster dose. However, effectiveness against severe infection is lower with Omicron than with other variants and also wanes [242-251]. As an example, in an observational study from the United States, vaccine effectiveness against hospitalization within five months of receiving the last of three mRNA COVID-19 vaccine doses was 79 and 60 percent during the BA.1/BA.2 and BA.4/BA.5 periods, respectively, but decreased to 41 and 29 percent more than five months after vaccination [251].

Consistent with the observed attenuated vaccine effectiveness against infection with the Omicron subvariant, neutralizing activity of sera from vaccinated individuals is reduced against Omicron compared with the original Wuhan strain virus and the Delta variant; the majority of infection-naïve individuals who received a primary vaccine series have no detectable neutralizing activity against Omicron [252-261]. However, previously infected individuals who received a primary series and individuals who receive booster vaccination retain adequate neutralizing titers against Omicron sublineages BA.1 and BA.2; neutralizing titers from such individuals are usually lower for BA.4 and BA.5 but in most cases still above baseline [262-265]. The discrepancy between the lack of neutralizing activity against Omicron in sera from vaccinated individuals and the persistence of protection against severe disease with vaccination may be in part because neutralizing activity is not the only immune measure of vaccine protection. Vaccine- or infection-induced cellular immunity appears robust against Omicron [266-269]. (See "COVID-19: Epidemiology, virology, and prevention", section on 'Omicron (B.1.1.529) and its sublineages'.)

Monovalent booster effectiveness — Because of waning immunity and decreased vaccine efficacy against circulating variants, public health authorities in many countries have recommended booster vaccines after the primary series [270]. In the United States, monovalent mRNA vaccine booster doses are no longer recommended. The bivalent mRNA vaccines are recommended for booster doses. This is discussed elsewhere. (See 'Bivalent mRNA vaccine booster effectiveness and immunogenicity' below.)

Effectiveness of a single monovalent booster dose – A booster vaccine improves vaccine effectiveness in the short term, including in the context of Omicron prevalence [271-279]. In a placebo-controlled, randomized trial of 10,000 individuals who had received two primary series doses of BNT162b2, vaccine efficacy of a third booster dose (given a median of 11 months after the prior dose) against symptomatic COVID-19 was 95.3 percent (95% CI 89.5-98.3) in the first two months [271]. Only two severe COVID-19 cases occurred, both in the placebo group, and neither patient was hospitalized. Similarly, in an observational study from Israel of over four million individuals aged 16 years or older who had received two doses of BNT162b2 at least five months previously, receipt of a booster dose was associated with a 10-times lower rate of infection in all age groups compared with those who did not receive a booster (adjusted rate difference of 57 to 90 infections per 100,000 days, depending on age group) and among individuals 60 years or older, an 18-times lower rate of severe illness (absolute difference 5.4 cases per 100,000 days) [272]. Observational data suggest that a booster dose is associated with enhanced short-term vaccine effectiveness in individuals 12 to 16 years of age [154]. Data on booster doses in children aged 5 to 11 years are limited to an unpublished trial demonstrating a sharp increase in neutralizing activity following a third BNT162b2 dose [280].

Data informing the use of heterologous booster vaccines are discussed elsewhere. (See 'Mixing vaccine types' above.)

Effectiveness of a second monovalent booster dose An additional booster dose appears to improve relative vaccine effectiveness against both infection and severe disease, at least in the short term [281-286]. In a retrospective study from Israel that included over 1 million individuals ≥60 years old who had received a primary series and an initial booster dose with BNT162b2, receipt of a second booster dose at least four months after the last was associated with twofold lower risk of confirmed infection and a 3.5-fold lower risk of severe infection [281]. The risk reduction for confirmed infection waned by eight weeks after the second booster dose, whereas the risk reduction for severe infection remained stable over the study period. Another study from Israel similarly suggested that a second booster dose was associated with a reduction in COVID-19-related death (adjusted hazard ratio 0.22) [282]. However, in both studies, the overall risk of severe disease or death was very low regardless of second booster, the follow-up period was very short (only 40 to 60 days), and the study design could not exclude the possibility that other differences between the two groups (eg, behavior, health status) could have contributed to the findings.

Limited evidence suggests that the added protection of a second booster dose against infection wanes after several months [287].

Safety of booster doses – The rate and severity of adverse reactions following booster doses are similar to those reported following a primary series. For mRNA vaccines, local and systemic reactions are reported slightly less frequently after the booster dose than the second dose and less frequently after the second booster dose than the first [288,289]. The risk of myocarditis also appears lower after a booster dose than the second dose of an mRNA vaccine [288,290,291]. Following 82.6 million booster doses administered to adults in the United States, there were 37 reports to the Centers for Disease Control and Prevention (CDC) that met criteria for myocarditis [288]. The highest rate was among males aged 18 to 24 years following an mRNA-1273 boost (8.7 per 1 million doses), considerably lower than that reported after the primary series. Similarly, following 2.8 million BNT162b2 booster doses administered to adolescents in the United States, the estimated rate of myocarditis among males aged 12 to 17 years was 11.4 per 1 million booster doses [292]. In a preliminary study from Israel, there were no cases of myocarditis in 12- to 15-year-olds after >6000 booster doses administered [290]. (See 'Myocarditis' below.)

Bivalent mRNA vaccine booster effectiveness and immunogenicity — To address issues of both waning efficacy since the last vaccine dose and the attenuated efficacy of COVID-19 vaccines against variants that escape the immune response directed against spike proteins targeted by the original vaccines, some countries have introduced bivalent mRNA COVID-19 vaccine boosters that encode spike protein from the original SARS-CoV-2 strain and from the Omicron variants. In some countries, the Omicron spike protein encoded in the bivalent mRNA COVID-19 vaccines is based on the BA.1 subvariant; in the United States, it is based on the BA.4 and BA.5 subvariants (they have identical spike proteins). Recommendations on bivalent vaccine booster doses in the United States are discussed elsewhere. (See 'Booster dose' above.)

Evidence for the effectiveness of bivalent booster vaccines is limited but suggests modest to moderate protection against infection depending on time since the last vaccine dose. In a study in the United States of over 250,000 symptomatic individuals who were tested for SARS-CoV-2 and had received two to four doses of monovalent vaccines, estimated vaccine effectiveness of an additional bivalent booster dose was 28 to 31 percent among those who had last been vaccinated two to three months previously and 43 to 56 percent among those last vaccinated more than eight months previously [293]. Data from immunogenicity studies evaluating bivalent vaccines are mixed and some have not yet been peer reviewed. Some studies, including those performed by the vaccine manufacturers, suggest that bivalent boosters that include the BA.4/5 spike protein induce higher antibody levels against BA.4/5 virus compared with pre-booster levels and compared with monovalent boosters [294-296]. Some of these studies also suggest that the antibody response elicited by the bivalent boosters sufficiently neutralizes other Omicron subvariants, such as BQ.1.1 (table 6). However, in other studies, the antibody response to the bivalent booster was similar to that with the monovalent booster and had minimal neutralizing activity against other Omicron subvariants [297,298]. The reasons for these discrepancies are uncertain but may be related to the specific assay or techniques used. Ultimately, more clinical evidence is needed to better assess the effect of the bivalent vaccines.

Nevertheless, indirect evidence from monovalent booster studies support the use of bivalent vaccines and supports the concept that their effectiveness would be expected to be greater than the monovalent product (see 'Monovalent booster effectiveness' above). Using a bivalent versus monovalent vaccine is analogous to updating the seasonal influenza vaccine; new vaccines are produced each year so that the antigenic targets match circulating virus, and these vaccines are made available globally prior to repeated clinical evaluation because of extensive experience with prior versions.

Booster doses following a primary vaccine series are a distinct issue from administering an additional dose in the primary series (eg, three doses of a primary mRNA vaccine series) for certain immunocompromised patients. Booster doses are also recommended in immunocompromised individuals who received an additional vaccine dose in the primary series [42]. This is discussed in detail elsewhere. (See 'Immunocompromised individuals' above.)

Breakthrough infections after vaccination — The risk of breakthrough infection is higher with certain variants, such as Delta and Omicron. Nevertheless, the individual risk of severe breakthrough infection with Omicron, particularly among those who received a booster vaccine dose, remains low. (See 'Waning effectiveness over time and with variants of concern' above.)

Breakthrough infection after vaccination is substantially less likely to cause severe disease than infection in unvaccinated individuals [299-302]. In a study of over 1 million vaccinated members of a large health system in the United States, the rates of severe disease and death due to breakthrough COVID-19 were 1.5 and 0.3 per 10,000, respectively [303]. Risk factors for poor outcomes are similar to those for unvaccinated individuals: older age (>65 years) and multiple comorbidities [303,304].

Other observational data suggest that breakthrough infection is associated with a lower number of symptoms, shorter duration of symptoms, lower likelihood of persistent symptoms for >28 days, and a higher likelihood of asymptomatic infection compared with infection in unvaccinated individuals [73,305-307].

Impact on transmission risk — Widespread vaccination reduces the overall transmission risk, since vaccinated individuals are less likely to get infection. Previous data had also suggested that individuals who developed infection despite vaccination may be less likely to transmit to others, thereby further decreasing transmission risk. However, the risk of transmission with a breakthrough Omicron infection is uncertain.

In a study of over 100,000 patients with SARS-CoV-2 and their contacts, a history of mRNA vaccine receipt was associated with lower secondary attack rates with Delta compared with no vaccination (adjusted relative risk 0.5), although the effect was not as great as with Alpha (adjusted relative risk 0.32) and further decreased by 12 weeks after vaccination [308]. Other studies performed during the period of Delta variant predominance suggested that viral RNA levels at the time of diagnosis were similar in unvaccinated and vaccinated individuals; however, they declined more rapidly or were associated with lower levels of infectious virus in the upper respiratory tract among vaccinated individuals [309-313]. These findings support the concept that vaccinated individuals were less likely to transmit certain SARS-CoV-2 variants, including Delta, if infected. Additional data are needed to evaluate the impact of vaccination on the viral kinetics of Omicron breakthrough infection and better understand their clinical implications.

Immune correlates of protection — Although data remain limited, analyses of vaccine trials support the concept that binding and neutralizing antibody levels against the spike protein and its receptor-binding domain are the primary immune predictors of protection against symptomatic infection, with increasing levels associated with progressively higher vaccine efficacy [314,315]. Data from these studies can help assess likely efficacy of new vaccines or regimens in different patient populations when large efficacy trials cannot be performed. However, the application to clinical care is uncertain; it is unknown how well results from the various commercially available serologic tests correspond to the measurement of antibody levels in these studies. Furthermore, the immune correlates of protection against severe infection have not been fully elucidated.

SPECIFIC SAFETY CONCERNS — COVID-19 vaccines are exceedingly safe. The primary safety concerns are a very rare risk of myocarditis with mRNA vaccines and very rare risks of thrombosis with thrombocytopenia and possibly Guillain-Barre syndrome with adenoviral vector vaccines. Although myriad adverse events have been reported in individuals following vaccine administration, no other severe events have been clearly associated with vaccination after hundreds of millions of doses administered.

As an example, many patients of child-bearing potential are concerned that COVID-19 vaccination could adversely impact fertility because of specious reports on social media. However, in epidemiologic studies, there is no association between COVID-19 vaccination and fertility problems in either females or males [316,317].

Thrombosis with thrombocytopenia — ChadOx1 nCoV-19/AZD1222 (AstraZeneca COVID-19 vaccine) and Ad26.COV2.S (Janssen COVID-19 vaccine, also referred to as the Johnson & Johnson vaccine) have each been associated with an extremely small risk of unusual types of thrombotic events associated with thrombocytopenia. A similar risk has not been identified with the mRNA vaccines. Many of these cases have been associated with autoantibodies directed against the platelet factor 4 (PF4) antigen, similar to those found in patients with autoimmune heparin-induced thrombocytopenia (HIT) [318-321]. Some experts refer to this syndrome as vaccine-associated immune thrombotic thrombocytopenia (VITT); others have used the term thrombosis with thrombocytopenia syndrome (TTS).

In reported cases, thrombosis often occurred at unusual sites, including the cerebral venous sinuses and mesenteric vessels, and at more than one site [322-324]. Most of the initially reported events occurred within two weeks of receipt of the initial vaccine dose and in females under 60 years of age, although subsequent cases have been reported following a longer post-vaccine interval and in males and older females. Some fatal cases have been reported.

In the United States, the risks of this syndrome following Ad26.COV2.S receipt was assessed as 3.8 cases and 0.57 deaths per million doses overall, and 9 to 10.6 cases and 1.8 to 1.93 deaths per million doses for females 30 to 49 years old [325]. Regulatory bodies in the United States and Europe have concluded that the population and individual benefits of these vaccines (compared with no vaccination), including reductions in death and critical illness, outweigh the risk of these rare events [326-328]. Additionally, the risk of hospitalization or death associated with thrombocytopenia or thromboembolic complications associated with SARS-CoV-2 infection is higher than that associated with adenoviral vaccines [329]. Nevertheless, recipients of these vaccines should be aware of the possible association and seek immediate care for signs and symptoms suggestive of thrombocytopenia (eg, new petechiae or bruising) or thrombotic complications (including shortness of breath, chest pain, lower extremity edema, persistent severe abdominal pain, unabating severe headache, severe backache, new focal neurologic symptoms, and seizures) [328].

The incidence, risk factors, clinical features, evaluation, and management of VITT/TTS are discussed in detail elsewhere. (See "COVID-19: Vaccine-induced immune thrombotic thrombocytopenia (VITT)".)

A clear, causal relation between either of these vaccines and thromboembolic disorders overall (eg, pulmonary embolism and deep vein thrombosis) has not been identified [207,330]. For ChadOx1 nCoV-19/AZD1222, studies have reported conflicting findings regarding this risk, as discussed elsewhere. (See 'ChAdOx1 nCoV-19/AZD1222 (University of Oxford, AstraZeneca, and the Serum Institute of India)' above.)

Myocarditis — Myocarditis and pericarditis, mainly in male adolescents and young adults, have been reported more frequently than expected following receipt of the mRNA vaccines, BNT162b2 (Pfizer COVID-19 vaccine) and mRNA-1273 (Moderna COVID-19 vaccine) [331,332]. Cases were also noted in NVX-CoV2373 (Novavax COVID-19 vaccine) recipients during the phase III trials [185]. A similar pattern of cases has not been reported following receipt of Ad26.COV2.S (Janssen/Johnson & Johnson COVID-19 vaccine). Given the infrequency and the mild nature of the myocarditis and pericarditis cases, the benefits of vaccination greatly exceed the small increased risk [332]. For those who develop myocarditis or pericarditis following an mRNA vaccine or NVX-CoV2373, we suggest that any subsequent COVID-19 vaccine dose be deferred in most cases; it is reasonable for such individuals to choose to receive an additional dose (ie, the second dose of the primary series or any booster doses) once the episode has completely resolved if the risk of severe COVID-19 is high [42]. Individuals with a history of resolved myocarditis or pericarditis unrelated to COVID-19 vaccination can receive a COVID-19 vaccine.

In a review of the Vaccine Adverse Event Reporting System (VAERS), a passive surveillance system in the United States to which patients and providers can submit reports of events, among over 192 million people who had received an mRNA vaccine between December 2020 and August 2021, there were 1626 cases that met the definition of myocarditis following vaccine receipt [333]. The majority of these cases occurred after the second dose, the median age was 21 years, and 82 percent occurred in males. The estimated rate among males by age group was:

12 to 15 years old – 70.7 cases per million doses of BNT162b2

16 to 17 years old – 105.9 cases per million doses of BNT162b2

18 to 24 years old – 52.4 to 56.3 cases per million doses BNT162b2 and mRNA-1273, respectively

Among females of the same age groups, the estimated case rates ranged from 6.4 to 11 cases per million doses. The number of events observed exceeded the expected baseline rate among males aged 18 to 49 years and females aged 19 to 29 years.

Studies from other countries have also suggested an increased rate of myocarditis following BNT162b2 vaccination compared with the expected background rate [334-337]. Observational data also suggest that the risk may be higher with mRNA-1273 than BNT162b2 [338-341].

For all age groups, the risk of myocarditis or pericarditis following mRNA vaccination is estimated in some, but not all [340], studies to be less than the risk associated with SARS-CoV-2 infection [342].

Among the cases that have been reported, most were mild [332,334,335,343]. Onset was generally within the first week after vaccine receipt. Most patients who presented for care responded well to medical treatment and had rapid symptom improvement. There have been very rare reports of fulminant myocarditis in individuals who had received an mRNA vaccine within the preceding weeks, although a causal relationship is difficult to establish [344].

The clinical presentation was illustrated in a retrospective study of 139 adolescents and young adults ≤21 years old with suspected vaccine-associated myocarditis based on elevated troponins within 30 days of vaccination without alternative diagnosis [345]. Almost all presented with chest pain, with symptom onset a median of two days after vaccine receipt. Electrocardiogram was abnormal in 70 percent (ST segment elevations or T wave abnormalities), cardiac magnetic resonance imaging was abnormal in 77 percent (late gadolinium enhancement and myocardial edema), but systolic function on echocardiogram was normal in 80 percent. Nineteen percent were managed in the intensive care unit, although only two patients required inotropic or vasopressor support. Median hospital stay was two days, and those with decreased systolic function had normalized ejection fraction on follow-up. Ongoing monitoring is necessary to assess for long-term sequelae.

The possibility of myocarditis should be considered in adolescents and young adults who develop new chest pain, shortness of breath, or palpitations after receiving an mRNA vaccine. The possibility of other causes of myocarditis (including SARS-CoV-2 infection) should also be considered. The diagnosis and management of myocarditis are discussed in detail elsewhere. (See "Clinical manifestations and diagnosis of myocarditis in children" and "Clinical manifestations and diagnosis of myocarditis in adults" and "Treatment and prognosis of myocarditis in children" and "Treatment and prognosis of myocarditis in adults".)

Guillain-Barre syndrome — A potential association between the adenovirus vector vaccines (Ad26.COV2.S [Janssen/Johnson & Johnson COVID-19 vaccine] and ChAdOx1 nCoV-19/AZD1222 [AstraZeneca COVID-19 vaccine]) and Guillain-Barre syndrome (GBS) is being investigated, but a causal relationship has not been established. A similar signal has not been observed with the mRNA COVID-19 vaccines. The US Food and Drug Administration (FDA) and Centers for Disease Control and Prevention (CDC) and European regulators affirm that the benefits of these vaccines outweigh their risks [346,347]. Cases of GBS, including recurrent cases, have also been reported in the setting of SARS-CoV-2 infection [348,349], and observational data suggest the risk of GBS after infection exceeds the risk after vaccination [350]. Pending additional data, for individuals with a documented history of GBS, we suggest using COVID-19 vaccines other than adenovirus vector vaccines; if only an adenovirus vector vaccine is available, we individualize the decision to administer it based on that person’s risk for severe COVID-19 and GBS history. The general approach to vaccination in individuals with a history of GBS is discussed elsewhere. (See "Guillain-Barré syndrome in adults: Treatment and prognosis", section on 'Subsequent immunizations'.)

In the United States, as of July 24, 2021, there had been 132 preliminary reports of GBS among Ad26.COV2.S recipients after approximately 13.2 million doses [351]. The estimated rate was 9.8 cases per million doses, a rate that is approximately four times the background rate. The median age was 56 years (interquartile range 45 to 62 years), the median time to onset was 13 days following vaccination, 35 percent had a life-threatening case, and 1 patient died. In an earlier report of 100 cases, a quarter of the patients reported bilateral facial weakness [352]. Another analysis suggested an incidence of 32 cases per 100,000 person-years within the three weeks following Ad26.COV2.S receipt; the estimate for mRNA vaccines was 1.3 per 100,000 person-years [353].

In Europe, a total of 227 cases of GBS in ChAdOx1 nCoV-19/AZD1222 recipients had been reported to regulators as of June 27, 2021, at which point approximately 51 million doses had been administered [347]. Other scattered reports have also described GBS, including variant GBS with bilateral facial weakness, following ChAdOx1 nCoV-19/AZD1222 vaccination [354,355]. However, in a cohort study including over 4 million individuals who received ChAdOx1 nCoV-19/AZD1222, the rate of GBS following the vaccine dose was not higher than the expected pre-pandemic background rate [356].

STEPS TO VACCINE AVAILABILITY AND DELIVERY

Establishing efficacy and licensing a vaccine – Initial estimates of vaccine efficacy are established by phase III trials. In the United States, the minimum criteria for licensure defined by the US Food and Drug Administration (FDA) were at least 50 percent efficacy in preventing microbiologically confirmed symptomatic SARS-CoV-2 infection, with a lower bound of a 95% confidence interval of 30 percent, and at least six months of follow-up for safety assessment [357]. The World Health Organization (WHO) has proposed the same minimal efficacy targets [358].

Once safety and efficacy meeting the criteria have been demonstrated, the FDA makes decisions on vaccine licensure, relying on guidance from the Vaccines and Related Biologic Products Advisory Committee (VRBPAC), a standing advisory group of experienced clinicians, vaccine experts, epidemiologists, and other subject matter experts. Similar approaches are taken by regulatory bodies in Canada and European countries for the licensure of their vaccines.

In addition to the traditional process to issue a license for a vaccine, the FDA can issue an emergency use authorization (EUA), which is designed to make products available during public health emergencies [359]. For a COVID-19 vaccine to receive EUA, it must meet the prespecified efficacy criteria defined for the primary endpoint with a median of two months of follow-up for half of the trial participants [360].

Allocation priorities – When vaccine supplies are limited, it is essential that vaccine deployment be equitable and efficient. Several expert organizations have released guidance documents for vaccine allocation approaches that maximize the individual and societal benefits of vaccination [361-363]. These prioritize vaccination according to risks of acquiring infection, severe morbidity and mortality, negative societal impact (eg, if essential critical societal functions depend on an individual or groups of individuals), and transmission to others; they also emphasize equitable vaccine allocation to populations disproportionately impacted by the pandemic because of structural inequities and social determinants of health, including Black, Latin American, and Indigenous populations. The framework proposed by the WHO also takes into account global equity concerns, including assurance of vaccine access to low- and middle-income countries [363].

Vaccine reimbursement – In the United States, COVID-19 vaccines will be free of charge for any individual for whom the Advisory Committee on Immunization Practices (ACIP) recommends vaccination [364]. Vaccine providers can get administration costs reimbursed by public or private insurers, or for uninsured patients, by the Health Resources and Services Administration's Provider Relief Fund [365].

POST-LICENSURE ISSUES

Combating vaccine hesitancy — Vaccine hesitancy presents a major obstacle to achieving vaccination coverage that is broad enough to result in herd immunity and slow community transmission. In general, vaccine hesitancy has become more common worldwide and was cited by the World Health Organization (WHO) as a top 10 global health threat in 2019 [366]. With COVID-19 vaccines, the accelerated nature of development and misinformation have further contributed to concerns or skepticism about safety and utility among vaccine-hesitant individuals. Efforts to optimize COVID-19 vaccine uptake should identify reasons for and characteristics associated with vaccine refusal and use that information to tailor approaches to individuals and populations.

Based on evidence from other vaccines, health care providers can improve vaccine acceptance in individual patients by making direct recommendations for vaccination, identifying concerns, educating patients on vaccine risks and benefits, and dispelling misconceptions about the disease and the vaccine. (See "Standard childhood vaccines: Parental hesitancy or refusal", section on 'Target education'.)

Communication points that may be helpful when speaking with patients who are uncertain about whether to receive a COVID-19 vaccine can be found here or on the Centers for Disease Control and Prevention (CDC) website [367,368].

Willingness to accept a COVID-19 vaccine has varied by country [369]. In the United States, rates of vaccine hesitancy have decreased over the course of the pandemic but remain substantial [370-373].

COVID-19 vaccine hesitancy has been associated with younger age (eg, <60 years old), lower levels of education, lower household income, rural residence, and lack of health insurance [370,374-376]. In a CDC survey, the main reasons for reporting non-intent to receive vaccine were concerns about vaccine side effects and safety and lack of trust in the process [374].

Ongoing safety assessment — Adequately assessing vaccine safety is critical to the success of immunization programs. Although existing comprehensive systems to monitor vaccine safety are in place, they are being enhanced for the rollout of the COVID-19 vaccine program. It is particularly important to identify rare adverse events that are causally related to vaccine administration and assess their incidence and risk factors to inform potential vaccine contraindications.

In the United States, there are several systems in place to assess safety in the post-licensure setting; some are passive (ie, rely on others reporting the event) and others are active (ie, review databases or conduct studies to identify events) [377]. These include the Vaccine Adverse Event Reporting System (VAERS), a passive surveillance system in which providers, parents, and patients report adverse events. VAERS is intended to raise hypotheses about whether receipt of a vaccine could cause the adverse event rather than evaluate causation. The Vaccine Safety Datalink (VSD) is a collaborative project between the CDC's Immunization Safety Office and eight health care organizations to actively monitor the safety of vaccines and conduct studies about rare and serious post-vaccination adverse events. The Clinical Immunization Safety Assessment project (CISA) is a national network of vaccine safety experts from the CDC's Immunization Safety Office, seven academic medical research centers, and subject matter experts, and it provides a comprehensive vaccine safety public health service to the nation.

In addition, specific post-licensure vaccine safety systems have been implemented for the introduction of COVID-19 vaccines, similar to those established during the 2009 H1N1 influenza pandemic [378,379]. These systems will be coordinated through the CDC and will enlist multiple other health care groups to provide ongoing data on vaccine safety. These systems and information sources add an additional layer of safety monitoring [380,381].

V-SAFE is a new smartphone-based health checker for people who have received a COVID-19 vaccine. The CDC will send text messages and web-based surveys to vaccine recipients through V-SAFE to check in regarding health problems following vaccination. The system will also provide telephone follow-up to anyone who reports clinically significant adverse events.

Enhanced reporting through National Healthcare Safety Network (NHSN) sites – A monitoring system for health care workers and long-term care facility residents that reports to the VAERS.

Monitoring of larger insurer/payer databases through the US Food and Drug Administration – A system of administrative and claims-based data for surveillance and research.

Since most vaccine-preventable diseases are transmitted person-to-person, effective vaccination not only protects the recipient but also indirectly protects others who cannot be vaccinated or do not respond adequately by preventing another source for transmission ("herd immunity") [382]. Therefore, if someone is injured by vaccine, society owes that person compensation. This is the basis for the National Vaccine Injury Compensation Program (NVICP) [383]. This program also reduces liability for the vaccine provider and the manufacturer, since it is a no-fault alternative to the traditional legal system for resolving vaccine injury claims. With COVID-19 vaccines, another compensation system called the Countermeasures Injury Compensation Program (CICP) may be used [384].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: COVID-19 – Index of guideline topics".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topics (see "Patient education: COVID-19 vaccines (The Basics)" and "Patient education: COVID-19 overview (The Basics)" and "Patient education: COVID-19 and pregnancy (The Basics)" and "Patient education: COVID-19 and children (The Basics)")

SUMMARY AND RECOMMENDATIONS

Antigenic target – The primary antigenic target for COVID-19 vaccines is the large surface spike protein (figure 1), which binds to the angiotensin-converting enzyme 2 (ACE2) receptor on host cells (figure 2). (See 'General principles' above.)

Indications – Several vaccines using different platforms (figure 3) have high vaccine efficacy against laboratory-confirmed symptomatic COVID-19 and substantially reduce the risk of severe COVID-19 (table 2). (See 'Immunogenicity, efficacy, and safety of select vaccines' above.)

For individuals 12 years and older, we recommend COVID-19 vaccination (Grade 1A). For individuals 6 months to 11 years old, we also recommend COVID-19 vaccination (Grade 1B). (See 'Approach to vaccination in the United States' above and 'Immunogenicity, efficacy, and safety of select vaccines' above.)

Selection and administration of the primary series – In the United States, the following COVID-19 vaccines are available (table 2) (see 'Indications and vaccine selection' above and 'Dose and interval' above):

Two mRNA vaccines:

-BNT162b2 (Pfizer COVID-19 vaccine): The primary series includes two intramuscular injections given at least three weeks apart for individuals aged five years or older; for children aged six months to four years old, three vaccine doses are given for the primary series. This vaccine is associated with a rare risk of myocarditis. (See 'BNT162b2 (Pfizer-BioNTech COVID-19 vaccine)' above.)

-mRNA-1273 (Moderna COVID-19 vaccine): The primary series includes two intramuscular injections given at least one month apart for individuals six months or older. This vaccine is associated with a rare risk of myocarditis. (See 'mRNA-1273 (Moderna COVID-19 vaccine)' above.)

For both mRNA vaccines, healthy individuals <65 years old can extend the interval between the two doses to eight weeks; this approach may be preferable for adolescents and young adults (especially males age 12 to 39 years), as it may be associated with a lower risk of myocarditis.

An adjuvanted recombinant protein vaccine NVX-CoV2373 (Novavax COVID-19 vaccine): The primary series includes two intramuscular injections given three to eight weeks apart for individuals aged 12 years or older. The vaccine may be associated with a rare risk of myocarditis. (See 'NVX-CoV2373 (Novavax COVID-19 vaccine)' above.)

An adenoviral vector vaccine Ad26.COV2.S (Janssen COVID-19 vaccine, also referred to as the Johnson & Johnson vaccine): The primary series includes a single intramuscular injection for individuals 18 years or older. This vaccine is associated with a rare risk of thrombosis with thrombocytopenia and possibly Guillain-Barre syndrome. (See 'Ad26.COV2.S (Janssen/Johnson & Johnson COVID-19 vaccine)' above.)

For those eligible for all vaccines, we suggest an mRNA vaccine (BNT162b2 or mRNA-1273) or NVX-CoV2373 (Grade 2C). All three are highly effective; experience is greatest with the mRNA vaccines. NVX-CoV2373 may be an attractive option for individuals who prefer a more established vaccine platform. We reserve Ad26.COV2.S for those who cannot use the others, as it may be less effective against severe disease than mRNA vaccines and its associated rare adverse effects appear more severe. Nevertheless, Ad26.COV2.S is an effective and safe option for most individuals if the others are unavailable or contraindicated. (See 'Specific safety concerns' above.)

Vaccine schedules for immunocompetent (figure 4) and immunocompromised (figure 5) adults are presented in the figures.

Different vaccines are available elsewhere; a list of vaccines that have been authorized in at least one country can be found at covid19.trackvaccines.org/vaccines. Clinicians outside the United States should refer to local guidelines for vaccine recommendations in their location. (See 'Approach to vaccination in other countries' above.)

Adjustments for immunocompromised patients – For individuals with certain immunocompromising conditions (table 3), we suggest administering a three-dose primary mRNA vaccine series rather than a two-dose series (figure 5) (Grade 2C). Similarly, we suggest an additional mRNA vaccine dose for such individuals who previously received a single Ad26.COV2.S dose (Grade 2C). Immunocompromised individuals are less likely to respond adequately to routine vaccination; additional doses are associated with improved vaccine effectiveness. These additional doses do not replace the booster doses. (See 'Immunocompromised individuals' above.)

Booster doses – For individuals who have received a primary COVID-19 vaccine series, we recommend administration of a booster dose when eligible (Grade 1B). A bivalent vaccine (either Pfizer bivalent vaccine or Moderna bivalent vaccine for those ≥5 years old) is used for the booster dose, given at least two months after the primary series. The approach to booster doses for younger children depends on the primary series received. (See 'Booster dose' above.)

For individuals ≥5 years old who have received a primary series and prior booster doses with a monovalent vaccine, we also suggest a booster dose with a bivalent mRNA vaccine (either Pfizer bivalent vaccine for those ≥5 years old or Moderna bivalent vaccine for those ≥6 years old) at least two months after the last vaccine dose (Grade 2C). Individuals who have immunocompromising conditions (table 3) or would otherwise be at high risk of severe disease (eg, because of multiple medical comorbidities) and who live in regions with high or increasing rates of SARS-CoV-2 transmission are likely to benefit most from a bivalent vaccine booster.

The intended role of a bivalent booster dose is to reverse waning immunity following a prior vaccine dose and improve the breadth of protection against SARS-CoV-2 variants. In the United States, the antigenic target of the bivalent vaccines is based on the original SARS-CoV-2 strain and the BA.4/BA.5 Omicron subvariants. (See 'Bivalent mRNA vaccine booster effectiveness and immunogenicity' above.)

Deviations from dosing recommendations – If the vaccine is administered in a manner different from the recommended approach, the dose or series generally does not have to be repeated. Centers for Disease Control and Prevention (CDC) recommendations on how to manage vaccination errors or deviations are presented in the table (table 4). (See 'Dose and interval' above.)

Expected side effects Vaccine recipients should be advised that side effects are common and include local and systemic reactions, including pain at the injection site, fever, fatigue, and headache. Analgesics or antipyretics (eg, nonsteroidal anti-inflammatory drugs [NSAIDs] or acetaminophen) can be taken if these reactions develop, although prophylactic use of these agents before vaccine receipt is generally discouraged because of the uncertain impact on the host immune response to vaccination. (See 'Patient counseling' above.)

Contraindications and precautions – The primary contraindications to COVID-19 vaccination are severe or immediate allergic reactions to the vaccine or any of its components. All individuals should be monitored for an immediate reaction for at least 15 minutes following vaccination. Individuals without a contraindication who have a history of anaphylaxis of any kind, an immediate allergic reaction to other vaccines or injectable therapies, or a contraindication to a COVID-19 vaccine class other than the one they are receiving should be monitored for 30 minutes. (See 'Contraindications and precautions (including allergies)' above and 'Monitoring for immediate reactions to vaccine' above.)

  1. World Health Organization. Draft landscape of COVID-19 candidate vaccines. https://www.who.int/publications/m/item/draft-landscape-of-covid-19-candidate-vaccines (Accessed on October 20, 2020).
  2. Zhou P, Yang XL, Wang XG, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020; 579:270.
  3. Krammer F. SARS-CoV-2 vaccines in development. Nature 2020; 586:516.
  4. Edwards KM, Dupont WD, Westrich MK, et al. A randomized controlled trial of cold-adapted and inactivated vaccines for the prevention of influenza A disease. J Infect Dis 1994; 169:68.
  5. Ohmit SE, Victor JC, Rotthoff JR, et al. Prevention of antigenically drifted influenza by inactivated and live attenuated vaccines. N Engl J Med 2006; 355:2513.
  6. Graepel KW, Kochhar S, Clayton EW, Edwards KE. Balancing Expediency and Scientific Rigor in Severe Acute Respiratory Syndrome Coronavirus 2 Vaccine Development. J Infect Dis 2020; 222:180.
  7. Graham BS. Rapid COVID-19 vaccine development. Science 2020; 368:945.
  8. Halstead SB, Katzelnick L. COVID-19 Vaccines: Should We Fear ADE? J Infect Dis 2020; 222:1946.
  9. Brighton Collaboration. Accelerated Assessment of the Risk of Disease Enhancement with COVID-19 Vaccines​, March 2020. https://brightoncollaboration.us/brighton-collaboration-cepi-covid-19-web-conference/ (Accessed on October 20, 2020).
  10. US Food and Drug Administration. Pfizer-BioNTech Fact Sheets (English) and FAQs. https://www.fda.gov/emergency-preparedness-and-response/coronavirus-disease-2019-covid-19/comirnaty-and-pfizer-biontech-covid-19-vaccine#additional (Accessed on February 03, 2022).
  11. EMERGENCY USE AUTHORIZATION (EUA) for PFIZER-BIONTECH COVID-19 VACCINE, BIVALENT (ORIGINAL AND OMICRON BA.4/BA.5). https://www.fda.gov/media/161327/download (Accessed on September 02, 2022).
  12. US Food and Drug Administration. Spikevax and Moderna COVID-19 Vaccine. https://www.fda.gov/emergency-preparedness-and-response/coronavirus-disease-2019-covid-19/spikevax-and-moderna-covid-19-vaccine (Accessed on February 03, 2022).
  13. EMERGENCY USE AUTHORIZATION (EUA) for MODERNA COVID-19 VACCINE, BIVALENT (ORIGINAL AND OMICRON BA.4/BA.5) BOOSTER DOSE FOR 6 YEARS OF AGE AND OLDER. https://www.fda.gov/media/161318/download (Accessed on October 13, 2022).
  14. Emergency Use Authorization (EUA) of the Novavax COVID-19 vaccine, adjuvanted to prevent coronavirus disease 2019 (COVID-19). https://www.fda.gov/media/159897/download (Accessed on July 21, 2022).
  15. US Food and Drug Administration. Emergency use authorization (EUA) of the Janssen COVID-19 vaccine to prevent coronavirus disease 2019 (COVID-19). https://www.fda.gov/media/146304/download (Accessed on February 03, 2022).
  16. Thompson MG, Stenehjem E, Grannis S, et al. Effectiveness of Covid-19 Vaccines in Ambulatory and Inpatient Care Settings. N Engl J Med 2021; 385:1355.
  17. Haas EJ, Angulo FJ, McLaughlin JM, et al. Impact and effectiveness of mRNA BNT162b2 vaccine against SARS-CoV-2 infections and COVID-19 cases, hospitalisations, and deaths following a nationwide vaccination campaign in Israel: an observational study using national surveillance data. Lancet 2021; 397:1819.
  18. Vasileiou E, Simpson CR, Shi T, et al. Interim findings from first-dose mass COVID-19 vaccination roll-out and COVID-19 hospital admissions in Scotland: a national prospective cohort study. Lancet 2021; 397:1646.
  19. Dagan N, Barda N, Kepten E, et al. BNT162b2 mRNA Covid-19 Vaccine in a Nationwide Mass Vaccination Setting. N Engl J Med 2021; 384:1412.
  20. McNamara LA, Wiegand RE, Burke RM, et al. Estimating the early impact of the US COVID-19 vaccination programme on COVID-19 cases, emergency department visits, hospital admissions, and deaths among adults aged 65 years and older: an ecological analysis of national surveillance data. Lancet 2022; 399:152.
  21. Ioannou GN, Locke ER, O'Hare AM, et al. COVID-19 Vaccination Effectiveness Against Infection or Death in a National U.S. Health Care System : A Target Trial Emulation Study. Ann Intern Med 2022; 175:352.
  22. Tenforde MW, Self WH, Gaglani M, et al. Effectiveness of mRNA Vaccination in Preventing COVID-19-Associated Invasive Mechanical Ventilation and Death - United States, March 2021-January 2022. MMWR Morb Mortal Wkly Rep 2022; 71:459.
  23. Havers FP, Pham H, Taylor CA, et al. COVID-19-Associated Hospitalizations Among Vaccinated and Unvaccinated Adults 18 Years or Older in 13 US States, January 2021 to April 2022. JAMA Intern Med 2022; 182:1071.
  24. Link-Gelles R. Updates on COVID-19 Vaccine Effectiveness during Omicron. ACIP meeting, September 1, 2022. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2022-09-01/04-COVID-Link-Gelles-508.pdf (Accessed on September 02, 2022).
  25. Xu S, Huang R, Sy LS, et al. COVID-19 Vaccination and Non-COVID-19 Mortality Risk - Seven Integrated Health Care Organizations, United States, December 14, 2020-July 31, 2021. MMWR Morb Mortal Wkly Rep 2021; 70:1520.
  26. Grannis SJ, Rowley EA, Ong TC, et al. Interim Estimates of COVID-19 Vaccine Effectiveness Against COVID-19-Associated Emergency Department or Urgent Care Clinic Encounters and Hospitalizations Among Adults During SARS-CoV-2 B.1.617.2 (Delta) Variant Predominance - Nine States, June-August 2021. MMWR Morb Mortal Wkly Rep 2021; 70:1291.
  27. Pilishvili T, Gierke R, Fleming-Dutra KE, et al. Effectiveness of mRNA Covid-19 Vaccine among U.S. Health Care Personnel. N Engl J Med 2021; 385:e90.
  28. Dickerman BA, Gerlovin H, Madenci AL, et al. Comparative Effectiveness of BNT162b2 and mRNA-1273 Vaccines in U.S. Veterans. N Engl J Med 2022; 386:105.
  29. Bajema KL, Dahl RM, Evener SL, et al. Comparative Effectiveness and Antibody Responses to Moderna and Pfizer-BioNTech COVID-19 Vaccines among Hospitalized Veterans - Five Veterans Affairs Medical Centers, United States, February 1-September 30, 2021. MMWR Morb Mortal Wkly Rep 2021; 70:1700.
  30. Abu-Raddad LJ, Chemaitelly H, Bertollini R, National Study Group for COVID-19 Vaccination. Effectiveness of mRNA-1273 and BNT162b2 Vaccines in Qatar. N Engl J Med 2022; 386:799.
  31. Wang L, Davis PB, Kaelber DC, et al. Comparison of mRNA-1273 and BNT162b2 Vaccines on Breakthrough SARS-CoV-2 Infections, Hospitalizations, and Death During the Delta-Predominant Period. JAMA 2022; 327:678.
  32. Lin DY, Gu Y, Wheeler B, et al. Effectiveness of Covid-19 Vaccines over a 9-Month Period in North Carolina. N Engl J Med 2022; 386:933.
  33. Britton A, Fleming-Dutra KE, Shang N, et al. Association of COVID-19 Vaccination With Symptomatic SARS-CoV-2 Infection by Time Since Vaccination and Delta Variant Predominance. JAMA 2022; 327:1032.
  34. Botton J, Semenzato L, Jabagi MJ, et al. Effectiveness of Ad26.COV2.S Vaccine vs BNT162b2 Vaccine for COVID-19 Hospitalizations. JAMA Netw Open 2022; 5:e220868.
  35. Islam N, Sheils NE, Jarvis MS, Cohen K. Comparative effectiveness over time of the mRNA-1273 (Moderna) vaccine and the BNT162b2 (Pfizer-BioNTech) vaccine. Nat Commun 2022; 13:2377.
  36. Dickerman BA, Madenci AL, Gerlovin H, et al. Comparative Safety of BNT162b2 and mRNA-1273 Vaccines in a Nationwide Cohort of US Veterans. JAMA Intern Med 2022; 182:739.
  37. Buchan SA, Seo CY, Johnson C, et al. Epidemiology of Myocarditis and Pericarditis Following mRNA Vaccination by Vaccine Product, Schedule, and Interdose Interval Among Adolescents and Adults in Ontario, Canada. JAMA Netw Open 2022; 5:e2218505.
  38. Grunau B, Goldfarb DM, Asamoah-Boaheng M, et al. Immunogenicity of Extended mRNA SARS-CoV-2 Vaccine Dosing Intervals. JAMA 2022; 327:279.
  39. Payne RP, Longet S, Austin JA, et al. Immunogenicity of standard and extended dosing intervals of BNT162b2 mRNA vaccine. Cell 2021; 184:5699.
  40. Amirthalingam G, Bernal JL, Andrews NJ, et al. Serological responses and vaccine effectiveness for extended COVID-19 vaccine schedules in England. Nat Commun 2021; 12:7217.
  41. Skowronski DM, Febriani Y, Ouakki M, et al. Two-Dose Severe Acute Respiratory Syndrome Coronavirus 2 Vaccine Effectiveness With Mixed Schedules and Extended Dosing Intervals: Test-Negative Design Studies From British Columbia and Quebec, Canada. Clin Infect Dis 2022; 75:1980.
  42. Interim Clinical Considerations for Use of COVID-19 Vaccines Currently Authorized in the United States. https://www.cdc.gov/vaccines/covid-19/clinical-considerations/covid-19-vaccines-us.html (Accessed on December 13, 2022).
  43. https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-authorizes-updated-bivalent-covid-19-vaccines-children-down-6-months (Accessed on December 08, 2022).
  44. EMERGENCY USE AUTHORIZATION (EUA). PFIZER-BIONTECH COVID-19 VACCINE, BIVALENT (ORIGINAL AND OMICRON BA.4/BA.5) BOOSTER DOSE FOR 5 THROUGH 11 YEARS OF AGE. https://www.fda.gov/media/162250/download (Accessed on October 13, 2022).
  45. Mallory RM, Formica N, Pfeiffer S, et al. Safety and immunogenicity following a homologous booster dose of a SARS-CoV-2 recombinant spike protein vaccine (NVX-CoV2373): a secondary analysis of a randomised, placebo-controlled, phase 2 trial. Lancet Infect Dis 2022; 22:1565.
  46. Atmar RL, Lyke KE, Deming ME, et al. Homologous and Heterologous Covid-19 Booster Vaccinations. N Engl J Med 2022; 386:1046.
  47. Sablerolles RSG, Rietdijk WJR, Goorhuis A, et al. Immunogenicity and Reactogenicity of Vaccine Boosters after Ad26.COV2.S Priming. N Engl J Med 2022; 386:951.
  48. Munro APS, Feng S, Janani L, et al. Safety, immunogenicity, and reactogenicity of BNT162b2 and mRNA-1273 COVID-19 vaccines given as fourth-dose boosters following two doses of ChAdOx1 nCoV-19 or BNT162b2 and a third dose of BNT162b2 (COV-BOOST): a multicentre, blinded, phase 2, randomised trial. Lancet Infect Dis 2022; 22:1131.
  49. Mayr FB, Talisa VB, Shaikh O, et al. Effectiveness of Homologous or Heterologous Covid-19 Boosters in Veterans. N Engl J Med 2022; 386:1375.
  50. Natarajan K, Prasad N, Dascomb K, et al. Effectiveness of Homologous and Heterologous COVID-19 Booster Doses Following 1 Ad.26.COV2.S (Janssen [Johnson & Johnson]) Vaccine Dose Against COVID-19-Associated Emergency Department and Urgent Care Encounters and Hospitalizations Among Adults - VISION Network, 10 States, December 2021-March 2022. MMWR Morb Mortal Wkly Rep 2022; 71:495.
  51. Accorsi EK, Britton A, Shang N, et al. Effectiveness of Homologous and Heterologous Covid-19 Boosters against Omicron. N Engl J Med 2022; 386:2433.
  52. Borobia AM, Carcas AJ, Pérez-Olmeda M, et al. Immunogenicity and reactogenicity of BNT162b2 booster in ChAdOx1-S-primed participants (CombiVacS): a multicentre, open-label, randomised, controlled, phase 2 trial. Lancet 2021; 398:121.
  53. Shaw RH, Stuart A, Greenland M, et al. Heterologous prime-boost COVID-19 vaccination: initial reactogenicity data. Lancet 2021; 397:2043.
  54. Normark J, Vikström L, Gwon YD, et al. Heterologous ChAdOx1 nCoV-19 and mRNA-1273 Vaccination. N Engl J Med 2021; 385:1049.
  55. Costa Clemens SA, Weckx L, Clemens R, et al. Heterologous versus homologous COVID-19 booster vaccination in previous recipients of two doses of CoronaVac COVID-19 vaccine in Brazil (RHH-001): a phase 4, non-inferiority, single blind, randomised study. Lancet 2022; 399:521.
  56. Tan CS, Collier AY, Yu J, et al. Durability of Heterologous and Homologous COVID-19 Vaccine Boosts. JAMA Netw Open 2022; 5:e2226335.
  57. Lazarus R, Baos S, Cappel-Porter H, et al. Safety and immunogenicity of concomitant administration of COVID-19 vaccines (ChAdOx1 or BNT162b2) with seasonal influenza vaccines in adults in the UK (ComFluCOV): a multicentre, randomised, controlled, phase 4 trial. Lancet 2021; 398:2277.
  58. Zhong D, Xiao S, Debes AK, et al. Durability of Antibody Levels After Vaccination With mRNA SARS-CoV-2 Vaccine in Individuals With or Without Prior Infection. JAMA 2021; 326:2524.
  59. Reynolds CJ, Pade C, Gibbons JM, et al. Prior SARS-CoV-2 infection rescues B and T cell responses to variants after first vaccine dose. Science 2021.
  60. Stamatatos L, Czartoski J, Wan YH, et al. mRNA vaccination boosts cross-variant neutralizing antibodies elicited by SARS-CoV-2 infection. Science 2021.
  61. Chin ET, Leidner D, Zhang Y, et al. Effectiveness of the mRNA-1273 Vaccine during a SARS-CoV-2 Delta Outbreak in a Prison. N Engl J Med 2021; 385:2300.
  62. Hammerman A, Sergienko R, Friger M, et al. Effectiveness of the BNT162b2 Vaccine after Recovery from Covid-19. N Engl J Med 2022; 386:1221.
  63. Gazit S, Shlezinger R, Perez G, et al. The Incidence of SARS-CoV-2 Reinfection in Persons With Naturally Acquired Immunity With and Without Subsequent Receipt of a Single Dose of BNT162b2 Vaccine : A Retrospective Cohort Study. Ann Intern Med 2022; 175:674.
  64. Hall V, Foulkes S, Insalata F, et al. Protection against SARS-CoV-2 after Covid-19 Vaccination and Previous Infection. N Engl J Med 2022; 386:1207.
  65. Cerqueira-Silva T, Andrews JR, Boaventura VS, et al. Effectiveness of CoronaVac, ChAdOx1 nCoV-19, BNT162b2, and Ad26.COV2.S among individuals with previous SARS-CoV-2 infection in Brazil: a test-negative, case-control study. Lancet Infect Dis 2022; 22:791.
  66. Nordström P, Ballin M, Nordström A. Risk of SARS-CoV-2 reinfection and COVID-19 hospitalisation in individuals with natural and hybrid immunity: a retrospective, total population cohort study in Sweden. Lancet Infect Dis 2022; 22:781.
  67. Carazo S, Skowronski DM, Brisson M, et al. Estimated Protection of Prior SARS-CoV-2 Infection Against Reinfection With the Omicron Variant Among Messenger RNA-Vaccinated and Nonvaccinated Individuals in Quebec, Canada. JAMA Netw Open 2022; 5:e2236670.
  68. Chin ET, Leidner D, Lamson L, et al. Protection against Omicron from Vaccination and Previous Infection in a Prison System. N Engl J Med 2022; 387:1770.
  69. Abu-Raddad LJ, Chemaitelly H, Ayoub HH, et al. Association of Prior SARS-CoV-2 Infection With Risk of Breakthrough Infection Following mRNA Vaccination in Qatar. JAMA 2021; 326:1930.
  70. Goldberg Y, Mandel M, Bar-On YM, et al. Protection and Waning of Natural and Hybrid Immunity to SARS-CoV-2. N Engl J Med 2022; 386:2201.
  71. Bozio CH, Grannis SJ, Naleway AL, et al. Laboratory-Confirmed COVID-19 Among Adults Hospitalized with COVID-19-Like Illness with Infection-Induced or mRNA Vaccine-Induced SARS-CoV-2 Immunity - Nine States, January-September 2021. MMWR Morb Mortal Wkly Rep 2021; 70:1539.
  72. León TM, Dorabawila V, Nelson L, et al. COVID-19 Cases and Hospitalizations by COVID-19 Vaccination Status and Previous COVID-19 Diagnosis - California and New York, May-November 2021. MMWR Morb Mortal Wkly Rep 2022; 71:125.
  73. UK Health Security Agency. The effectiveness of vaccination against long COVID: A rapid evidence briefing. https://ukhsa.koha-ptfs.co.uk/cgi-bin/koha/opac-retrieve-file.pl?id=fe4f10cd3cd509fe045ad4f72ae0dfff (Accessed on February 24, 2022).
  74. Krammer F, Srivastava K, Alshammary H, et al. Antibody Responses in Seropositive Persons after a Single Dose of SARS-CoV-2 mRNA Vaccine. N Engl J Med 2021; 384:1372.
  75. Menni C, Klaser K, May A, et al. Vaccine side-effects and SARS-CoV-2 infection after vaccination in users of the COVID Symptom Study app in the UK: a prospective observational study. Lancet Infect Dis 2021; 21:939.
  76. CDC - An Additional Dose of mRNA COVID-19 Vaccine Following a Primary Series in Immunocompromised People. Available at: https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2021-08-13/02-COVID-Dooling-508.pdf (Accessed on August 14, 2021).
  77. Centers for Disease Control and Prevention. Updated healthcare infection prevention and control recommendations in response to COVID-19 vaccination. Available at: https://www.cdc.gov/coronavirus/2019-ncov/hcp/infection-control-after-vaccination.html (Accessed on May 05, 2021).
  78. https://www.hematology.org/covid-19/ash-astct-covid-19-and-vaccines (Accessed on February 21, 2022).
  79. CDC Data and clinical considerations for additional doses in immunocompromised people. Available at: https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2021-07/07-COVID-Oliver-508.pdf (Accessed on July 23, 2021).
  80. Tenforde MW, Patel MM, Gaglani M, et al. Effectiveness of a Third Dose of Pfizer-BioNTech and Moderna Vaccines in Preventing COVID-19 Hospitalization Among Immunocompetent and Immunocompromised Adults - United States, August-December 2021. MMWR Morb Mortal Wkly Rep 2022; 71:118.
  81. Britton A, Embi PJ, Levy ME, et al. Effectiveness of COVID-19 mRNA Vaccines Against COVID-19-Associated Hospitalizations Among Immunocompromised Adults During SARS-CoV-2 Omicron Predominance - VISION Network, 10 States, December 2021-August 2022. MMWR Morb Mortal Wkly Rep 2022; 71:1335.
  82. Kamar N, Abravanel F, Marion O, et al. Three Doses of an mRNA Covid-19 Vaccine in Solid-Organ Transplant Recipients. N Engl J Med 2021; 385:661.
  83. Werbel WA, Boyarsky BJ, Ou MT, et al. Safety and Immunogenicity of a Third Dose of SARS-CoV-2 Vaccine in Solid Organ Transplant Recipients: A Case Series. Ann Intern Med 2021; 174:1330.
  84. Longlune N, Nogier MB, Miedougé M, et al. High immunogenicity of a messenger RNA-based vaccine against SARS-CoV-2 in chronic dialysis patients. Nephrol Dial Transplant 2021; 36:1704.
  85. Benotmane I, Gautier G, Perrin P, et al. Antibody Response After a Third Dose of the mRNA-1273 SARS-CoV-2 Vaccine in Kidney Transplant Recipients With Minimal Serologic Response to 2 Doses. JAMA 2021.
  86. Hall VG, Ferreira VH, Ku T, et al. Randomized Trial of a Third Dose of mRNA-1273 Vaccine in Transplant Recipients. N Engl J Med 2021; 385:1244.
  87. Caillard S, Thaunat O, Benotmane I, et al. Antibody Response to a Fourth Messenger RNA COVID-19 Vaccine Dose in Kidney Transplant Recipients: A Case Series. Ann Intern Med 2022; 175:455.
  88. American College of Rheumatology. COVID-19 Vaccine Clinical Guidance Summary for Patients with Rheumatic and Musculoskeletal Diseases. https://www.rheumatology.org/Portals/0/Files/COVID-19-Vaccine-Clinical-Guidance-Rheumatic-Diseases-Summary.pdf (Accessed on March 04, 2021).
  89. Chodick G, Tene L, Rotem RS, et al. The Effectiveness of the Two-Dose BNT162b2 Vaccine: Analysis of Real-World Data. Clin Infect Dis 2022; 74:472.
  90. Tenforde MW, Patel MM, Ginde AA, et al. Effectiveness of Severe Acute Respiratory Syndrome Coronavirus 2 Messenger RNA Vaccines for Preventing Coronavirus Disease 2019 Hospitalizations in the United States. Clin Infect Dis 2022; 74:1515.
  91. Brosh-Nissimov T, Orenbuch-Harroch E, Chowers M, et al. BNT162b2 vaccine breakthrough: clinical characteristics of 152 fully vaccinated hospitalized COVID-19 patients in Israel. Clin Microbiol Infect 2021; 27:1652.
  92. Embi PJ, Levy ME, Naleway AL, et al. Effectiveness of 2-Dose Vaccination with mRNA COVID-19 Vaccines Against COVID-19-Associated Hospitalizations Among Immunocompromised Adults - Nine States, January-September 2021. MMWR Morb Mortal Wkly Rep 2021; 70:1553.
  93. Aslam S, Adler E, Mekeel K, Little SJ. Clinical effectiveness of COVID-19 vaccination in solid organ transplant recipients. Transpl Infect Dis 2021; 23:e13705.
  94. Sun J, Zheng Q, Madhira V, et al. Association Between Immune Dysfunction and COVID-19 Breakthrough Infection After SARS-CoV-2 Vaccination in the US. JAMA Intern Med 2022; 182:153.
  95. Lee ARYB, Wong SY, Chai LYA, et al. Efficacy of covid-19 vaccines in immunocompromised patients: systematic review and meta-analysis. BMJ 2022; 376:e068632.
  96. Risk M, Hayek SS, Schiopu E, et al. COVID-19 vaccine effectiveness against omicron (B.1.1.529) variant infection and hospitalisation in patients taking immunosuppressive medications: a retrospective cohort study. Lancet Rheumatol 2022; 4:e775.
  97. Boyarsky BJ, Werbel WA, Avery RK, et al. Antibody Response to 2-Dose SARS-CoV-2 mRNA Vaccine Series in Solid Organ Transplant Recipients. JAMA 2021; 325:2204.
  98. Marion O, Del Bello A, Abravanel F, et al. Safety and Immunogenicity of Anti-SARS-CoV-2 Messenger RNA Vaccines in Recipients of Solid Organ Transplants. Ann Intern Med 2021; 174:1336.
  99. Redjoul R, Le Bouter A, Beckerich F, et al. Antibody response after second BNT162b2 dose in allogeneic HSCT recipients. Lancet 2021; 398:298.
  100. Mazzola A, Todesco E, Drouin S, et al. Poor Antibody Response After Two Doses of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Vaccine in Transplant Recipients. Clin Infect Dis 2022; 74:1093.
  101. Moor MB, Suter-Riniker F, Horn MP, et al. Humoral and cellular responses to mRNA vaccines against SARS-CoV-2 in patients with a history of CD20 B-cell-depleting therapy (RituxiVac): an investigator-initiated, single-centre, open-label study. Lancet Rheumatol 2021; 3:e789.
  102. Anderson EJ, Campbell JD, Creech CB, et al. Warp Speed for Coronavirus Disease 2019 (COVID-19) Vaccines: Why Are Children Stuck in Neutral? Clin Infect Dis 2021; 73:336.
  103. Olivier S. Evidence to Recommendations (EtR) Framework: Pfizer-BioNTech COVID-19 vaccine in children aged 5–11 years. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2021-11-2-3/08-COVID-Oliver-508.pdf (Accessed on November 03, 2021).
  104. Olivier S. Evidence to Recommendation Framework: Moderna COVID-19 vaccine in children ages 6 months – 5 years & Pfizer-BioNTech COVID-19 vaccine in children ages 6 months – 4 years. June 17, 2022. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2022-06-17-18/03-COVID-Oliver-508.pdf (Accessed on June 19, 2022).
  105. Nygaard U, Holm M, Dungu KHS, et al. Risk of Myopericarditis After COVID-19 Vaccination in Danish Children Aged 5 to 11 Years. Pediatrics 2022; 150.
  106. Vaccines and Related Biological Products Advisory Committee Meeting. FDA Briefing Document: EUA amendment request for use of the Moderna COVID-19 Vaccine in children 6 months through 17 years of age. June 14-15, 2022 https://www.fda.gov/media/159189/download (Accessed on June 16, 2022).
  107. Vaccines and Related Biological Products Advisory Committee Meeting. FDA Briefing Document: EUA amendment request for Pfizer-BioNTech COVID-19 Vaccine for use in children 6 months through 4 years of age, June 15, 2022. https://www.fda.gov/media/159195/download (Accessed on June 17, 2022).
  108. Hause AM, Baggs J, Marquez P, et al. COVID-19 Vaccine Safety in Children Aged 5-11 Years - United States, November 3-December 19, 2021. MMWR Morb Mortal Wkly Rep 2021; 70:1755.
  109. Centers for Disease Control and Prevention. COVID-19 vaccine safety update: Primary series in young children and booster doses in older children and adults. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2022-09-01/05-COVID-Shimabukuro-508.pdf (Accessed on September 05, 2022).
  110. Yang H. Benefits-Risks of Pfizer-BioNTech COVID-19 Vaccine for Ages 5 to 11 Years. https://www.fda.gov/media/153507/download (Accessed on November 02, 2021).
  111. Salzman MB, Huang CW, O'Brien CM, Castillo RD. Multisystem Inflammatory Syndrome after SARS-CoV-2 Infection and COVID-19 Vaccination. Emerg Infect Dis 2021; 27:1944.
  112. Levy M, Recher M, Hubert H, et al. Multisystem Inflammatory Syndrome in Children by COVID-19 Vaccination Status of Adolescents in France. JAMA 2022; 327:281.
  113. Zambrano LD, Newhams MM, Olson SM, et al. Effectiveness of BNT162b2 (Pfizer-BioNTech) mRNA Vaccination Against Multisystem Inflammatory Syndrome in Children Among Persons Aged 12-18 Years - United States, July-December 2021. MMWR Morb Mortal Wkly Rep 2022; 71:52.
  114. Centers for Disease Control and Prevention. Vaccines for Children Program (VFC). https://www.cdc.gov/vaccines/programs/vfc/ (Accessed on November 05, 2020).
  115. Whitney CG, Zhou F, Singleton J, et al. Benefits from immunization during the vaccines for children program era - United States, 1994-2013. MMWR Morb Mortal Wkly Rep 2014; 63:352.
  116. Hermann EA, Lee B, Balte PP, et al. Association of Symptoms After COVID-19 Vaccination With Anti-SARS-CoV-2 Antibody Response in the Framingham Heart Study. JAMA Netw Open 2022; 5:e2237908.
  117. Debes AK, Xiao S, Colantuoni E, et al. Association of Vaccine Type and Prior SARS-CoV-2 Infection With Symptoms and Antibody Measurements Following Vaccination Among Health Care Workers. JAMA Intern Med 2021; 181:1660.
  118. Chapin-Bardales J, Gee J, Myers T. Reactogenicity Following Receipt of mRNA-Based COVID-19 Vaccines. JAMA 2021; 325:2201.
  119. Prymula R, Siegrist CA, Chlibek R, et al. Effect of prophylactic paracetamol administration at time of vaccination on febrile reactions and antibody responses in children: two open-label, randomised controlled trials. Lancet 2009; 374:1339.
  120. Doedée AM, Boland GJ, Pennings JL, et al. Effects of prophylactic and therapeutic paracetamol treatment during vaccination on hepatitis B antibody levels in adults: two open-label, randomized controlled trials. PLoS One 2014; 9:e98175.
  121. Centers for Disease Control and Prevention (CDC). Syncope after vaccination--United States, January 2005-July 2007. MMWR Morb Mortal Wkly Rep 2008; 57:457.
  122. CDC. Stay Up to Date with Your COVID-19 Vaccines. https://www.cdc.gov/coronavirus/2019-ncov/vaccines/stay-up-to-date.html#footnote02 (Accessed on April 27, 2022).
  123. Maayan H, Kirgner I, Gutwein O, et al. Acquired thrombotic thrombocytopenic purpura: A rare disease associated with BNT162b2 vaccine. J Thromb Haemost 2021; 19:2314.
  124. Matheny M, Maleque N, Channell N, et al. Severe Exacerbations of Systemic Capillary Leak Syndrome After COVID-19 Vaccination: A Case Series. Ann Intern Med 2021; 174:1476.
  125. American Society of Hematology. Thrombosis with Thrombocytopenia Syndrome (also termed Vaccine-induced Thrombotic Thrombocytopenia). https://www.hematology.org/covid-19/vaccine-induced-immune-thrombotic-thrombocytopenia (Accessed on April 24, 2021).
  126. Blumenthal KG, Freeman EE, Saff RR, et al. Delayed Large Local Reactions to mRNA-1273 Vaccine against SARS-CoV-2. N Engl J Med 2021; 384:1273.
  127. Blumenthal KG, Saff RR, Freeman EE. Delayed Large Local Reactions to mRNA Vaccines. Reply. N Engl J Med 2021; 384:e98.
  128. Casajuana J, Iglesias B, Fàbregas M, et al. Safety of intramuscular influenza vaccine in patients receiving oral anticoagulation therapy: a single blinded multi-centre randomized controlled clinical trial. BMC Blood Disord 2008; 8:1.
  129. Banerji A, Wickner PG, Saff R, et al. mRNA Vaccines to Prevent COVID-19 Disease and Reported Allergic Reactions: Current Evidence and Suggested Approach. J Allergy Clin Immunol Pract 2021; 9:1423.
  130. Shimabukuro T. COVID-19 vaccine safety update, Advisory Committee on Immunization Practices (ACIP) meeting, January 27, 2021. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2021-01/06-COVID-Shimabukuro.pdf (Accessed on January 28, 2021).
  131. Shimabukuro T. Allergic reactions including anaphylaxis after receipt of the first dose of Moderna COVID-19 vaccine - United States, December 21, 2020-January 10, 2021. Am J Transplant 2021; 21:1326.
  132. Gee J, Marquez P, Su J, et al. First Month of COVID-19 Vaccine Safety Monitoring - United States, December 14, 2020-January 13, 2021. MMWR Morb Mortal Wkly Rep 2021; 70:283.
  133. FDA Briefing Document. Pfizer-BioNTech COVID-19 Vaccine. Vaccines and Related Biological Products Advisory Committee Meeting. December 10, 2020 https://www.fda.gov/media/144245/download (Accessed on December 09, 2020).
  134. Polack FP, Thomas SJ, Kitchin N, et al. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N Engl J Med 2020; 383:2603.
  135. Frenck RW Jr, Klein NP, Kitchin N, et al. Safety, Immunogenicity, and Efficacy of the BNT162b2 Covid-19 Vaccine in Adolescents. N Engl J Med 2021; 385:239.
  136. US FDA. Emergency Use Authorization (EUA) of the Pfizer-BioNTech COVID-19 vaccine to prevent coronavirus disease 2019 (COVID-19) for 5 through 11 years of age. https://www.fda.gov/media/153714/download (Accessed on November 02, 2021).
  137. Thomas SJ, Moreira ED Jr, Kitchin N, et al. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine through 6 Months. N Engl J Med 2021; 385:1761.
  138. Reis BY, Barda N, Leshchinsky M, et al. Effectiveness of BNT162b2 Vaccine against Delta Variant in Adolescents. N Engl J Med 2021; 385:2101.
  139. Olson SM, Newhams MM, Halasa NB, et al. Effectiveness of Pfizer-BioNTech mRNA Vaccination Against COVID-19 Hospitalization Among Persons Aged 12-18 Years - United States, June-September 2021. MMWR Morb Mortal Wkly Rep 2021; 70:1483.
  140. Olson SM, Newhams MM, Halasa NB, et al. Effectiveness of BNT162b2 Vaccine against Critical Covid-19 in Adolescents. N Engl J Med 2022; 386:713.
  141. Hall VJ, Foulkes S, Saei A, et al. COVID-19 vaccine coverage in health-care workers in England and effectiveness of BNT162b2 mRNA vaccine against infection (SIREN): a prospective, multicentre, cohort study. Lancet 2021; 397:1725.
  142. Amit S, Regev-Yochay G, Afek A, et al. Early rate reductions of SARS-CoV-2 infection and COVID-19 in BNT162b2 vaccine recipients. Lancet 2021; 397:875.
  143. Rinott E, Youngster I, Lewis YE. Reduction in COVID-19 Patients Requiring Mechanical Ventilation Following Implementation of a National COVID-19 Vaccination Program - Israel, December 2020-February 2021. MMWR Morb Mortal Wkly Rep 2021; 70:326.
  144. Britton A, Jacobs Slifka KM, Edens C, et al. Effectiveness of the Pfizer-BioNTech COVID-19 Vaccine Among Residents of Two Skilled Nursing Facilities Experiencing COVID-19 Outbreaks - Connecticut, December 2020-February 2021. MMWR Morb Mortal Wkly Rep 2021; 70:396.
  145. Benenson S, Oster Y, Cohen MJ, Nir-Paz R. BNT162b2 mRNA Covid-19 Vaccine Effectiveness among Health Care Workers. N Engl J Med 2021; 384:1775.
  146. Tenforde MW, Olson SM, Self WH, et al. Effectiveness of Pfizer-BioNTech and Moderna Vaccines Against COVID-19 Among Hospitalized Adults Aged ≥65 Years - United States, January-March 2021. MMWR Morb Mortal Wkly Rep 2021; 70:674.
  147. Butt AA, Omer SB, Yan P, et al. SARS-CoV-2 Vaccine Effectiveness in a High-Risk National Population in a Real-World Setting. Ann Intern Med 2021; 174:1404.
  148. Chodick G, Tene L, Patalon T, et al. Assessment of Effectiveness of 1 Dose of BNT162b2 Vaccine for SARS-CoV-2 Infection 13 to 24 Days After Immunization. JAMA Netw Open 2021; 4:e2115985.
  149. Chung H, He S, Nasreen S, et al. Effectiveness of BNT162b2 and mRNA-1273 covid-19 vaccines against symptomatic SARS-CoV-2 infection and severe covid-19 outcomes in Ontario, Canada: test negative design study. BMJ 2021; 374:n1943.
  150. Fowlkes AL, Yoon SK, Lutrick K, et al. Effectiveness of 2-Dose BNT162b2 (Pfizer BioNTech) mRNA Vaccine in Preventing SARS-CoV-2 Infection Among Children Aged 5-11 Years and Adolescents Aged 12-15 Years - PROTECT Cohort, July 2021-February 2022. MMWR Morb Mortal Wkly Rep 2022; 71:422.
  151. Price AM, Olson SM, Newhams MM, et al. BNT162b2 Protection against the Omicron Variant in Children and Adolescents. N Engl J Med 2022; 386:1899.
  152. Shi DS, Whitaker M, Marks KJ, et al. Hospitalizations of Children Aged 5-11 Years with Laboratory-Confirmed COVID-19 - COVID-NET, 14 States, March 2020-February 2022. MMWR Morb Mortal Wkly Rep 2022; 71:574.
  153. Dorabawila V, Hoefer D, Bauer UE, et al. Risk of Infection and Hospitalization Among Vaccinated and Unvaccinated Children and Adolescents in New York After the Emergence of the Omicron Variant. JAMA 2022; 327:2242.
  154. Fleming-Dutra KE, Britton A, Shang N, et al. Association of Prior BNT162b2 COVID-19 Vaccination With Symptomatic SARS-CoV-2 Infection in Children and Adolescents During Omicron Predominance. JAMA 2022; 327:2210.
  155. Cohen-Stavi CJ, Magen O, Barda N, et al. BNT162b2 Vaccine Effectiveness against Omicron in Children 5 to 11 Years of Age. N Engl J Med 2022; 387:227.
  156. Sacco C, Del Manso M, Mateo-Urdiales A, et al. Effectiveness of BNT162b2 vaccine against SARS-CoV-2 infection and severe COVID-19 in children aged 5-11 years in Italy: a retrospective analysis of January-April, 2022. Lancet 2022; 400:97.
  157. Tan SHX, Cook AR, Heng D, et al. Effectiveness of BNT162b2 Vaccine against Omicron in Children 5 to 11 Years of Age. N Engl J Med 2022; 387:525.
  158. Lin DY, Gu Y, Xu Y, et al. Effects of Vaccination and Previous Infection on Omicron Infections in Children. N Engl J Med 2022; 387:1141.
  159. Chemaitelly H, AlMukdad S, Ayoub HH, et al. Covid-19 Vaccine Protection among Children and Adolescents in Qatar. N Engl J Med 2022; 387:1865.
  160. Walsh EE, Frenck RW Jr, Falsey AR, et al. Safety and Immunogenicity of Two RNA-Based Covid-19 Vaccine Candidates. N Engl J Med 2020; 383:2439.
  161. 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; 385:e84.
  162. Wall EC, Wu M, Harvey R, et al. Neutralising antibody activity against SARS-CoV-2 VOCs B.1.617.2 and B.1.351 by BNT162b2 vaccination. Lancet 2021; 397:2331.
  163. Edara VV, Pinsky BA, Suthar MS, et al. Infection and Vaccine-Induced Neutralizing-Antibody Responses to the SARS-CoV-2 B.1.617 Variants. N Engl J Med 2021; 385:664.
  164. Planas D, Veyer D, Baidaliuk A, et al. Reduced sensitivity of SARS-CoV-2 variant Delta to antibody neutralization. Nature 2021; 596:276.
  165. Pfizer-BioNTech COVID-19 vaccine demonstrates strong immune response, high efficacy and favorable safety in children 6 months to under 5 years of age following third dose. Pfizer. Available at: https://www.pfizer.com/news/press-release/press-release-detail/pfizer-biontech-covid-19-vaccine-demonstrates-strong-immune (Accessed on June 02, 2022).
  166. CDC COVID-19 Response Team, Food and Drug Administration. Allergic Reactions Including Anaphylaxis After Receipt of the First Dose of Pfizer-BioNTech COVID-19 Vaccine - United States, December 14-23, 2020. MMWR Morb Mortal Wkly Rep 2021; 70:46.
  167. Klein NP, Lewis N, Goddard K, et al. Surveillance for Adverse Events After COVID-19 mRNA Vaccination. JAMA 2021; 326:1390.
  168. Barda N, Dagan N, Ben-Shlomo Y, et al. Safety of the BNT162b2 mRNA Covid-19 Vaccine in a Nationwide Setting. N Engl J Med 2021; 385:1078.
  169. Baden LR, El Sahly HM, Essink B, et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N Engl J Med 2021; 384:403.
  170. 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; 385:1774.
  171. Pilishvili T, Fleming-Dutra KE, Farrar JL, et al. Interim Estimates of Vaccine Effectiveness of Pfizer-BioNTech and Moderna COVID-19 Vaccines Among Health Care Personnel - 33 U.S. Sites, January-March 2021. MMWR Morb Mortal Wkly Rep 2021; 70:753.
  172. Thompson MG, Burgess JL, Naleway AL, et al. Prevention and Attenuation of Covid-19 with the BNT162b2 and mRNA-1273 Vaccines. N Engl J Med 2021; 385:320.
  173. Bruxvoort KJ, Sy LS, Qian L, et al. Effectiveness of mRNA-1273 against delta, mu, and other emerging variants of SARS-CoV-2: test negative case-control study. BMJ 2021; 375:e068848.
  174. Jackson LA, Anderson EJ, Rouphael NG, et al. An mRNA Vaccine against SARS-CoV-2 - Preliminary Report. N Engl J Med 2020; 383:1920.
  175. Anderson EJ, Rouphael NG, Widge AT, et al. Safety and Immunogenicity of SARS-CoV-2 mRNA-1273 Vaccine in Older Adults. N Engl J Med 2020; 383:2427.
  176. Ali K, Berman G, Zhou H, et al. Evaluation of mRNA-1273 SARS-CoV-2 Vaccine in Adolescents. N Engl J Med 2021; 385:2241.
  177. Creech CB, Anderson E, Berthaud V, et al. Evaluation of mRNA-1273 Covid-19 Vaccine in Children 6 to 11 Years of Age. N Engl J Med 2022; 386:2011.
  178. Anderson EJ, Creech CB, Berthaud V, et al. Evaluation of mRNA-1273 Vaccine in Children 6 Months to 5 Years of Age. N Engl J Med 2022; 387:1673.
  179. 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; 384:2259.
  180. Steensels D, Pierlet N, Penders J, et al. Comparison of SARS-CoV-2 Antibody Response Following Vaccination With BNT162b2 and mRNA-1273. JAMA 2021; 326:1533.
  181. Richards NE, Keshavarz B, Workman LJ, et al. Comparison of SARS-CoV-2 Antibody Response by Age Among Recipients of the BNT162b2 vs the mRNA-1273 Vaccine. JAMA Netw Open 2021; 4:e2124331.
  182. FDA Briefing Document. Moderna COVID-19 Vaccine. https://www.fda.gov/media/144434/download (Accessed on December 16, 2020).
  183. Dunkle LM, Kotloff KL, Gay CL, et al. Efficacy and Safety of NVX-CoV2373 in Adults in the United States and Mexico. N Engl J Med 2022; 386:531.
  184. Heath PT, Galiza EP, Baxter DN, et al. Safety and Efficacy of NVX-CoV2373 Covid-19 Vaccine. N Engl J Med 2021; 385:1172.
  185. Vaccines and Related Biological Products Advisory Committee Meeting. FDA Briefing Document: Novavax COVID-19 Vaccine. June 7, 2022 https://www.fda.gov/media/158912/download (Accessed on June 16, 2022).
  186. Sadoff J, Gray G, Vandebosch A, et al. Safety and Efficacy of Single-Dose Ad26.COV2.S Vaccine against Covid-19. N Engl J Med 2021; 384:2187.
  187. Sadoff J, Gray G, Vandebosch A, et al. Final Analysis of Efficacy and Safety of Single-Dose Ad26.COV2.S. N Engl J Med 2022; 386:847.
  188. Corchado-Garcia J, Zemmour D, Hughes T, et al. Analysis of the Effectiveness of the Ad26.COV2.S Adenoviral Vector Vaccine for Preventing COVID-19. JAMA Netw Open 2021; 4:e2132540.
  189. Bekker LG, Garrett N, Goga A, et al. Effectiveness of the Ad26.COV2.S vaccine in health-care workers in South Africa (the Sisonke study): results from a single-arm, open-label, phase 3B, implementation study. Lancet 2022; 399:1141.
  190. Sadoff J, Le Gars M, Shukarev G, et al. Interim Results of a Phase 1-2a Trial of Ad26.COV2.S Covid-19 Vaccine. N Engl J Med 2021; 384:1824.
  191. Stephenson KE, Le Gars M, Sadoff J, et al. Immunogenicity of the Ad26.COV2.S Vaccine for COVID-19. JAMA 2021; 325:1535.
  192. Barouch DH, Stephenson KE, Sadoff J, et al. Durable Humoral and Cellular Immune Responses 8 Months after Ad26.COV2.S Vaccination. N Engl J Med 2021; 385:951.
  193. Collier AY, Yu J, McMahan K, et al. Differential Kinetics of Immune Responses Elicited by Covid-19 Vaccines. N Engl J Med 2021; 385:2010.
  194. Shay DK, Gee J, Su JR, et al. Safety Monitoring of the Janssen (Johnson & Johnson) COVID-19 Vaccine - United States, March-April 2021. MMWR Morb Mortal Wkly Rep 2021; 70:680.
  195. Hause AM, Gee J, Johnson T, et al. Anxiety-Related Adverse Event Clusters After Janssen COVID-19 Vaccination - Five U.S. Mass Vaccination Sites, April 2021. MMWR Morb Mortal Wkly Rep 2021; 70:685.
  196. Takuva S, Takalani A, Seocharan I, et al. Safety evaluation of the single-dose Ad26.COV2.S vaccine among healthcare workers in the Sisonke study in South Africa: A phase 3b implementation trial. PLoS Med 2022; 19:e1004024.
  197. World Health Organization. Interim recommendations for use of the AZD1222 (ChAdOx1-S (recombinant)) vaccine against COVID-19 developed by Oxford University and AstraZeneca. https://www.who.int/publications/i/item/WHO-2019-nCoV-vaccines-SAGE_recommendation-AZD1222-2021.1 (Accessed on February 11, 2021).
  198. Voysey M, Clemens SAC, Madhi SA, et al. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet 2021; 397:99.
  199. Falsey AR, Sobieszczyk ME, Hirsch I, et al. Phase 3 Safety and Efficacy of AZD1222 (ChAdOx1 nCoV-19) Covid-19 Vaccine. N Engl J Med 2021; 385:2348.
  200. 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; 397:881.
  201. Lopez Bernal J, Andrews N, Gower C, et al. Effectiveness of the Pfizer-BioNTech and Oxford-AstraZeneca vaccines on covid-19 related symptoms, hospital admissions, and mortality in older adults in England: test negative case-control study. BMJ 2021; 373:n1088.
  202. Katikireddi SV, Cerqueira-Silva T, Vasileiou E, et al. Two-dose ChAdOx1 nCoV-19 vaccine protection against COVID-19 hospital admissions and deaths over time: a retrospective, population-based cohort study in Scotland and Brazil. Lancet 2022; 399:25.
  203. Folegatti PM, Ewer KJ, Aley PK, et al. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet 2020; 396:467.
  204. Flaxman A, Marchevsky NG, Jenkin D, et al. Reactogenicity and immunogenicity after a late second dose or a third dose of ChAdOx1 nCoV-19 in the UK: a substudy of two randomised controlled trials (COV001 and COV002). Lancet 2021; 398:981.
  205. Ramasamy MN, Minassian AM, Ewer KJ, et al. Safety and immunogenicity of ChAdOx1 nCoV-19 vaccine administered in a prime-boost regimen in young and old adults (COV002): a single-blind, randomised, controlled, phase 2/3 trial. Lancet 2021; 396:1979.
  206. Wall EC, Wu M, Harvey R, et al. AZD1222-induced neutralising antibody activity against SARS-CoV-2 Delta VOC. Lancet 2021; 398:207.
  207. European Medicines Agency. https://www.ema.europa.eu/en/news/covid-19-vaccine-astrazeneca-benefits-still-outweigh-risks-despite-possible-link-rare-blood-clots (Accessed on March 18, 2021).
  208. Whiteley WN, Ip S, Cooper JA, et al. Association of COVID-19 vaccines ChAdOx1 and BNT162b2 with major venous, arterial, or thrombocytopenic events: A population-based cohort study of 46 million adults in England. PLoS Med 2022; 19:e1003926.
  209. Pottegård A, Lund LC, Karlstad Ø, et al. Arterial events, venous thromboembolism, thrombocytopenia, and bleeding after vaccination with Oxford-AstraZeneca ChAdOx1-S in Denmark and Norway: population based cohort study. BMJ 2021; 373:n1114.
  210. Hviid A, Hansen JV, Thiesson EM, Wohlfahrt J. Association of AZD1222 and BNT162b2 COVID-19 Vaccination With Thromboembolic and Thrombocytopenic Events in Frontline Personnel : A Retrospective Cohort Study. Ann Intern Med 2022; 175:541.
  211. Zhu FC, Guan XH, Li YH, et al. Immunogenicity and safety of a recombinant adenovirus type-5-vectored COVID-19 vaccine in healthy adults aged 18 years or older: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2020; 396:479.
  212. Halperin SA, Ye L, MacKinnon-Cameron D, et al. Final efficacy analysis, interim safety analysis, and immunogenicity of a single dose of recombinant novel coronavirus vaccine (adenovirus type 5 vector) in adults 18 years and older: an international, multicentre, randomised, double-blinded, placebo-controlled phase 3 trial. Lancet 2022; 399:237.
  213. Sputnik Vaccine. The Gamaleya Center statement on increasing the interval between the first and second Sputnik V vaccine shots. https://sputnikvaccine.com/newsroom/pressreleases/the-gamaleya-center-statement-on-increasing-the-interval-between-first-and-second-sputnik-v-vaccine-/ (Accessed on May 06, 2021).
  214. Logunov DY, Dolzhikova IV, Zubkova OV, et al. Safety and immunogenicity of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: two open, non-randomised phase 1/2 studies from Russia. Lancet 2020; 396:887.
  215. Al Kaabi N, Zhang Y, Xia S, et al. Effect of 2 Inactivated SARS-CoV-2 Vaccines on Symptomatic COVID-19 Infection in Adults: A Randomized Clinical Trial. JAMA 2021; 326:35.
  216. Tanriover MD, Doğanay HL, Akova M, et al. Efficacy and safety of an inactivated whole-virion SARS-CoV-2 vaccine (CoronaVac): interim results of a double-blind, randomised, placebo-controlled, phase 3 trial in Turkey. Lancet 2021; 398:213.
  217. Baraniuk C. What do we know about China's covid-19 vaccines? BMJ 2021; 373:n912.
  218. Fadlyana E, Rusmil K, Tarigan R, et al. A phase III, observer-blind, randomized, placebo-controlled study of the efficacy, safety, and immunogenicity of SARS-CoV-2 inactivated vaccine in healthy adults aged 18-59 years: An interim analysis in Indonesia. Vaccine 2021; 39:6520.
  219. Jara A, Undurraga EA, González C, et al. Effectiveness of an Inactivated SARS-CoV-2 Vaccine in Chile. N Engl J Med 2021; 385:875.
  220. Ranzani OT, Hitchings MDT, Dorion M, et al. Effectiveness of the CoronaVac vaccine in older adults during a gamma variant associated epidemic of covid-19 in Brazil: test negative case-control study. BMJ 2021; 374:n2015.
  221. Ella R, Reddy S, Blackwelder W, et al. Efficacy, safety, and lot-to-lot immunogenicity of an inactivated SARS-CoV-2 vaccine (BBV152): interim results of a randomised, double-blind, controlled, phase 3 trial. Lancet 2021; 398:2173.
  222. Mallapaty S. India's DNA COVID vaccine is a world first - more are coming. Nature 2021; 597:161.
  223. Khobragade A, Bhate S, Ramaiah V, et al. Efficacy, safety, and immunogenicity of the DNA SARS-CoV-2 vaccine (ZyCoV-D): the interim efficacy results of a phase 3, randomised, double-blind, placebo-controlled study in India. Lancet 2022; 399:1313.
  224. Rosenberg ES, Holtgrave DR, Dorabawila V, et al. New COVID-19 Cases and Hospitalizations Among Adults, by Vaccination Status - New York, May 3-July 25, 2021. MMWR Morb Mortal Wkly Rep 2021; 70:1150.
  225. Nanduri S, Pilishvili T, Derado G, et al. Effectiveness of Pfizer-BioNTech and Moderna Vaccines in Preventing SARS-CoV-2 Infection Among Nursing Home Residents Before and During Widespread Circulation of the SARS-CoV-2 B.1.617.2 (Delta) Variant - National Healthcare Safety Network, March 1-August 1, 2021. MMWR Morb Mortal Wkly Rep 2021; 70:1163.
  226. Chemaitelly H, Tang P, Hasan MR, et al. Waning of BNT162b2 Vaccine Protection against SARS-CoV-2 Infection in Qatar. N Engl J Med 2021; 385:e83.
  227. Tartof SY, Slezak JM, Fischer H, et al. Effectiveness of mRNA BNT162b2 COVID-19 vaccine up to 6 months in a large integrated health system in the USA: a retrospective cohort study. Lancet 2021; 398:1407.
  228. Baden LR, El Sahly HM, Essink B, et al. Phase 3 Trial of mRNA-1273 during the Delta-Variant Surge. N Engl J Med 2021; 385:2485.
  229. Cohn BA, Cirillo PM, Murphy CC, et al. SARS-CoV-2 vaccine protection and deaths among US veterans during 2021. Science 2022; 375:331.
  230. Israel A, Merzon E, Schäffer AA, et al. Elapsed time since BNT162b2 vaccine and risk of SARS-CoV-2 infection: test negative design study. BMJ 2021; 375:e067873.
  231. Abu-Raddad LJ, Chemaitelly H, Bertollini R, National Study Group for COVID-19 Vaccination. Waning mRNA-1273 Vaccine Effectiveness against SARS-CoV-2 Infection in Qatar. N Engl J Med 2022; 386:1091.
  232. Nordström P, Ballin M, Nordström A. Risk of infection, hospitalisation, and death up to 9 months after a second dose of COVID-19 vaccine: a retrospective, total population cohort study in Sweden. Lancet 2022; 399:814.
  233. Feikin DR, Higdon MM, Abu-Raddad LJ, et al. Duration of effectiveness of vaccines against SARS-CoV-2 infection and COVID-19 disease: results of a systematic review and meta-regression. Lancet 2022; 399:924.
  234. Tenforde MW, Self WH, Naioti EA, et al. Sustained Effectiveness of Pfizer-BioNTech and Moderna Vaccines Against COVID-19 Associated Hospitalizations Among Adults - United States, March-July 2021. MMWR Morb Mortal Wkly Rep 2021; 70:1156.
  235. Goldberg Y, Mandel M, Bar-On YM, et al. Waning Immunity after the BNT162b2 Vaccine in Israel. N Engl J Med 2021; 385:e85.
  236. Andrews N, Tessier E, Stowe J, et al. Duration of Protection against Mild and Severe Disease by Covid-19 Vaccines. N Engl J Med 2022; 386:340.
  237. Ferdinands JM, Rao S, Dixon BE, et al. Waning 2-Dose and 3-Dose Effectiveness of mRNA Vaccines Against COVID-19-Associated Emergency Department and Urgent Care Encounters and Hospitalizations Among Adults During Periods of Delta and Omicron Variant Predominance - VISION Network, 10 States, August 2021-January 2022. MMWR Morb Mortal Wkly Rep 2022; 71:255.
  238. Ng OT, Marimuthu K, Lim N, et al. Analysis of COVID-19 Incidence and Severity Among Adults Vaccinated With 2-Dose mRNA COVID-19 or Inactivated SARS-CoV-2 Vaccines With and Without Boosters in Singapore. JAMA Netw Open 2022; 5:e2228900.
  239. Lin DY, Gu Y, Xu Y, et al. Association of Primary and Booster Vaccination and Prior Infection With SARS-CoV-2 Infection and Severe COVID-19 Outcomes. JAMA 2022; 328:1415.
  240. Ridgway JP, Tideman S, French T, et al. Odds of Hospitalization for COVID-19 After 3 vs 2 Doses of mRNA COVID-19 Vaccine by Time Since Booster Dose. JAMA 2022; 328:1559.
  241. Ferdinands JM, Rao S, Dixon BE, et al. Waning of vaccine effectiveness against moderate and severe covid-19 among adults in the US from the VISION network: test negative, case-control study. BMJ 2022; 379:e072141.
  242. Accorsi EK, Britton A, Fleming-Dutra KE, et al. Association Between 3 Doses of mRNA COVID-19 Vaccine and Symptomatic Infection Caused by the SARS-CoV-2 Omicron and Delta Variants. JAMA 2022; 327:639.
  243. Thompson MG, Natarajan K, Irving SA, et al. Effectiveness of a Third Dose of mRNA Vaccines Against COVID-19-Associated Emergency Department and Urgent Care Encounters and Hospitalizations Among Adults During Periods of Delta and Omicron Variant Predominance - VISION Network, 10 States, August 2021-January 2022. MMWR Morb Mortal Wkly Rep 2022; 71:139.
  244. Tseng HF, Ackerson BK, Luo Y, et al. Effectiveness of mRNA-1273 against SARS-CoV-2 Omicron and Delta variants. Nat Med 2022; 28:1063.
  245. Lauring AS, Tenforde MW, Chappell JD, et al. Clinical severity of, and effectiveness of mRNA vaccines against, covid-19 from omicron, delta, and alpha SARS-CoV-2 variants in the United States: prospective observational study. BMJ 2022; 376:e069761.
  246. Abu-Raddad LJ, Chemaitelly H, Ayoub HH, et al. Effect of mRNA Vaccine Boosters against SARS-CoV-2 Omicron Infection in Qatar. N Engl J Med 2022; 386:1804.
  247. Andrews N, Stowe J, Kirsebom F, et al. Covid-19 Vaccine Effectiveness against the Omicron (B.1.1.529) Variant. N Engl J Med 2022; 386:1532.
  248. Altarawneh HN, Chemaitelly H, Ayoub HH, et al. Effects of Previous Infection and Vaccination on Symptomatic Omicron Infections. N Engl J Med 2022; 387:21.
  249. Discovery Health, South Africa’s largest private health insurance administrator, releases at-scale, real-world analysis of Omicron outbreak based on 211,000 COVID-19 test results in South Africa, including collaboration with South Africa. https://www.discovery.co.za/corporate/health-insights-omicron-outbreak-analysis (Accessed on December 17, 2021).
  250. Collie S, Champion J, Moultrie H, et al. Effectiveness of BNT162b2 Vaccine against Omicron Variant in South Africa. N Engl J Med 2022; 386:494.
  251. Surie D, Bonnell L, Adams K, et al. Effectiveness of Monovalent mRNA Vaccines Against COVID-19-Associated Hospitalization Among Immunocompetent Adults During BA.1/BA.2 and BA.4/BA.5 Predominant Periods of SARS-CoV-2 Omicron Variant in the United States - IVY Network, 18 States, December 26, 2021-August 31, 2022. MMWR Morb Mortal Wkly Rep 2022; 71:1327.
  252. Garcia-Beltran WF, St Denis KJ, Hoelzemer A, et al. mRNA-based COVID-19 vaccine boosters induce neutralizing immunity against SARS-CoV-2 Omicron variant. Cell 2022; 185:457.
  253. Nemet I, Kliker L, Lustig Y, et al. Third BNT162b2 Vaccination Neutralization of SARS-CoV-2 Omicron Infection. N Engl J Med 2022; 386:492.
  254. Dejnirattisai W, Shaw RH, Supasa P, et al. Reduced neutralisation of SARS-CoV-2 omicron B.1.1.529 variant by post-immunisation serum. Lancet 2022; 399:234.
  255. Hoffmann M, Krüger N, Schulz S, et al. The Omicron variant is highly resistant against antibody-mediated neutralization: Implications for control of the COVID-19 pandemic. Cell 2022; 185:447.
  256. Rössler A, Riepler L, Bante D, et al. SARS-CoV-2 Omicron Variant Neutralization in Serum from Vaccinated and Convalescent Persons. N Engl J Med 2022; 386:698.
  257. Muik A, Lui BG, Wallisch AK, et al. Neutralization of SARS-CoV-2 Omicron by BNT162b2 mRNA vaccine-elicited human sera. Science 2022; 375:678.
  258. Wu M, Wall EC, Carr EJ, et al. Three-dose vaccination elicits neutralising antibodies against omicron. Lancet 2022; 399:715.
  259. Pajon R, Doria-Rose NA, Shen X, et al. SARS-CoV-2 Omicron Variant Neutralization after mRNA-1273 Booster Vaccination. N Engl J Med 2022; 386:1088.
  260. Walls AC, Sprouse KR, Bowen JE, et al. SARS-CoV-2 breakthrough infections elicit potent, broad, and durable neutralizing antibody responses. Cell 2022; 185:872.
  261. Yu J, Collier AY, Rowe M, et al. Neutralization of the SARS-CoV-2 Omicron BA.1 and BA.2 Variants. N Engl J Med 2022; 386:1579.
  262. Wang Q, Guo Y, Iketani S, et al. Antibody evasion by SARS-CoV-2 Omicron subvariants BA.2.12.1, BA.4 and BA.5. Nature 2022; 608:603.
  263. Hachmann NP, Miller J, Collier AY, et al. Neutralization Escape by SARS-CoV-2 Omicron Subvariants BA.2.12.1, BA.4, and BA.5. N Engl J Med 2022; 387:86.
  264. Qu P, Faraone J, Evans JP, et al. Neutralization of the SARS-CoV-2 Omicron BA.4/5 and BA.2.12.1 Subvariants. N Engl J Med 2022; 386:2526.
  265. Bowen JE, Addetia A, Dang HV, et al. Omicron spike function and neutralizing activity elicited by a comprehensive panel of vaccines. Science 2022; 377:890.
  266. Goel RR, Painter MM, Apostolidis SA, et al. mRNA vaccines induce durable immune memory to SARS-CoV-2 and variants of concern. Science 2021; 374:abm0829.
  267. Liu J, Chandrashekar A, Sellers D, et al. Vaccines elicit highly conserved cellular immunity to SARS-CoV-2 Omicron. Nature 2022; 603:493.
  268. Keeton R, Tincho MB, Ngomti A, et al. T cell responses to SARS-CoV-2 spike cross-recognize Omicron. Nature 2022; 603:488.
  269. Tarke A, Coelho CH, Zhang Z, et al. SARS-CoV-2 vaccination induces immunological T cell memory able to cross-recognize variants from Alpha to Omicron. Cell 2022; 185:847.
  270. Joint Statement from HHS Public Health and Medical Experts on COVID-19 Booster Shots. https://www.cdc.gov/media/releases/2021/s0818-covid-19-booster-shots.html (Accessed on August 20, 2021).
  271. Moreira ED Jr, Kitchin N, Xu X, et al. Safety and Efficacy of a Third Dose of BNT162b2 Covid-19 Vaccine. N Engl J Med 2022; 386:1910.
  272. Bar-On YM, Goldberg Y, Mandel M, et al. Protection against Covid-19 by BNT162b2 Booster across Age Groups. N Engl J Med 2021; 385:2421.
  273. Barda N, Dagan N, Cohen C, et al. Effectiveness of a third dose of the BNT162b2 mRNA COVID-19 vaccine for preventing severe outcomes in Israel: an observational study. Lancet 2021; 398:2093.
  274. Patalon T, Gazit S, Pitzer VE, et al. Odds of Testing Positive for SARS-CoV-2 Following Receipt of 3 vs 2 Doses of the BNT162b2 mRNA Vaccine. JAMA Intern Med 2022; 182:179.
  275. Spitzer A, Angel Y, Marudi O, et al. Association of a Third Dose of BNT162b2 Vaccine With Incidence of SARS-CoV-2 Infection Among Health Care Workers in Israel. JAMA 2022; 327:341.
  276. Tai CG, Maragakis LL, Connolly S, et al. Association Between COVID-19 Booster Vaccination and Omicron Infection in a Highly Vaccinated Cohort of Players and Staff in the National Basketball Association. JAMA 2022; 328:209.
  277. Arbel R, Hammerman A, Sergienko R, et al. BNT162b2 Vaccine Booster and Mortality Due to Covid-19. N Engl J Med 2021; 385:2413.
  278. Muhsen K, Maimon N, Mizrahi A, et al. Effects of BNT162b2 Covid-19 Vaccine Booster in Long-Term Care Facilities in Israel. N Engl J Med 2022; 386:399.
  279. Au WY, Cheung PP. Effectiveness of heterologous and homologous covid-19 vaccine regimens: living systematic review with network meta-analysis. BMJ 2022; 377:e069989.
  280. Oliver S, Updates to the Evidence to Recommendation Framework: Pfizer-BioNTech COVID-19 booster in children aged 5-11 years. ACIP meeting, May 19, 2022. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2022-05-19/06-COVID-Oliver-508.pdf (Accessed on June 02, 2022).
  281. Bar-On YM, Goldberg Y, Mandel M, et al. Protection by a Fourth Dose of BNT162b2 against Omicron in Israel. N Engl J Med 2022; 386:1712.
  282. Arbel R, Sergienko R, Friger M, et al. Effectiveness of a second BNT162b2 booster vaccine against hospitalization and death from COVID-19 in adults aged over 60 years. Nat Med 2022; 28:1486.
  283. Magen O, Waxman JG, Makov-Assif M, et al. Fourth Dose of BNT162b2 mRNA Covid-19 Vaccine in a Nationwide Setting. N Engl J Med 2022; 386:1603.
  284. Grewal R, Kitchen SA, Nguyen L, et al. Effectiveness of a fourth dose of covid-19 mRNA vaccine against the omicron variant among long term care residents in Ontario, Canada: test negative design study. BMJ 2022; 378:e071502.
  285. Muhsen K, Maimon N, Mizrahi AY, et al. Association of Receipt of the Fourth BNT162b2 Dose With Omicron Infection and COVID-19 Hospitalizations Among Residents of Long-term Care Facilities. JAMA Intern Med 2022; 182:859.
  286. Cohen MJ, Oster Y, Moses AE, et al. Association of Receiving a Fourth Dose of the BNT162b Vaccine With SARS-CoV-2 Infection Among Health Care Workers in Israel. JAMA Netw Open 2022; 5:e2224657.
  287. Canetti M, Barda N, Gilboa M, et al. Six-Month Follow-up after a Fourth BNT162b2 Vaccine Dose. N Engl J Med 2022; 387:2092.
  288. Hause AM, Baggs J, Marquez P, et al. Safety Monitoring of COVID-19 Vaccine Booster Doses Among Adults - United States, September 22, 2021-February 6, 2022. MMWR Morb Mortal Wkly Rep 2022; 71:249.
  289. Hause AM, Baggs J, Marquez P, et al. Safety Monitoring of COVID-19 mRNA Vaccine Second Booster Doses Among Adults Aged ≥50 Years - United States, March 29, 2022-July 10, 2022. MMWR Morb Mortal Wkly Rep 2022; 71:971.
  290. Oliver S. Updates to the Evidence to Recommendation Framework: Pfizer-BioNTech vaccine booster doses in 12–15 year olds. Presented at ACIP meeting January 5, 2022. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2022-01-05/06_COVID_Oliver_2022-01-05.pdf (Accessed on January 05, 2022).
  291. Friedensohn L, Levin D, Fadlon-Derai M, et al. Myocarditis Following a Third BNT162b2 Vaccination Dose in Military Recruits in Israel. JAMA 2022; 327:1611.
  292. Hause AM, Baggs J, Marquez P, et al. Safety Monitoring of COVID-19 Vaccine Booster Doses Among Persons Aged 12-17 Years - United States, December 9, 2021-February 20, 2022. MMWR Morb Mortal Wkly Rep 2022; 71:347.
  293. Link-Gelles R, Ciesla AA, Fleming-Dutra KE, et al. Effectiveness of Bivalent mRNA Vaccines in Preventing Symptomatic SARS-CoV-2 Infection - Increasing Community Access to Testing Program, United States, September-November 2022. MMWR Morb Mortal Wkly Rep 2022; 71:1526.
  294. Fast Facts for Health Care Providers: Data Points on the Bivalent COVID-19 Vaccinations. https://www.fda.gov/media/163290/download?utm_medium=email&utm_source=govdelivery (Accessed on December 07, 2022).
  295. Zou J, Kurhade C, Patel S, et al. Improved Neutralization of Omicron BA.4/5, BA.4.6, BA.2.75.2, BQ.1.1, and XBB.1 with Bivalent BA.4/5 Vaccine. UNPUBLISHED. https://www.biorxiv.org/content/10.1101/2022.11.17.516898v1 (Accessed on December 08, 2022).
  296. Davis-Gardner M, Lai L, Wali B, et al. mRNA bivalent booster enhances neutralization against BA.2.75.2 and BQ.1.1.UNPUBLISHED. https://www.biorxiv.org/content/10.1101/2022.10.31.514636v1 (Accessed on December 08, 2022).
  297. Kurhade C, Zou J, Xia H, et al. Low neutralization of SARS-CoV-2 Omicron BA.2.75.2, BQ.1.1, and XBB.1 by parental mRNA vaccine or a BA.5-bivalent booster. Nat Med 2022.
  298. Collier AY, Miller J, Hachmann NP, et al. Immunogenicity of the BA.5 Bivalent mRNA Vaccine Boosters.UNPUBLISHED. https://www.biorxiv.org/content/10.1101/2022.10.24.513619v1 (Accessed on December 08, 2022).
  299. CDC COVID-19 Vaccine Breakthrough Case Investigations Team. COVID-19 Vaccine Breakthrough Infections Reported to CDC - United States, January 1-April 30, 2021. MMWR Morb Mortal Wkly Rep 2021; 70:792.
  300. Bergwerk M, Gonen T, Lustig Y, et al. Covid-19 Breakthrough Infections in Vaccinated Health Care Workers. N Engl J Med 2021; 385:1474.
  301. Tenforde MW, Self WH, Adams K, et al. Association Between mRNA Vaccination and COVID-19 Hospitalization and Disease Severity. JAMA 2021; 326:2043.
  302. Kim YE, Huh K, Park YJ, et al. Association Between Vaccination and Acute Myocardial Infarction and Ischemic Stroke After COVID-19 Infection. JAMA 2022; 328:887.
  303. Yek C, Warner S, Wiltz JL, et al. Risk Factors for Severe COVID-19 Outcomes Among Persons Aged ≥18 Years Who Completed a Primary COVID-19 Vaccination Series - 465 Health Care Facilities, United States, December 2020-October 2021. MMWR Morb Mortal Wkly Rep 2022; 71:19.
  304. Grange Z, Buelo A, Sullivan C, et al. Characteristics and risk of COVID-19-related death in fully vaccinated people in Scotland. Lancet 2021; 398:1799.
  305. Antonelli M, Penfold RS, Merino J, et al. Risk factors and disease profile of post-vaccination SARS-CoV-2 infection in UK users of the COVID Symptom Study app: a prospective, community-based, nested, case-control study. Lancet Infect Dis 2022; 22:43.
  306. Al-Aly Z, Bowe B, Xie Y. Long COVID after breakthrough SARS-CoV-2 infection. Nat Med 2022; 28:1461.
  307. Azzolini E, Levi R, Sarti R, et al. Association Between BNT162b2 Vaccination and Long COVID After Infections Not Requiring Hospitalization in Health Care Workers. JAMA 2022; 328:676.
  308. Eyre DW, Taylor D, Purver M, et al. Effect of Covid-19 Vaccination on Transmission of Alpha and Delta Variants. N Engl J Med 2022; 386:744.
  309. Singanayagam A, Hakki S, Dunning J, et al. Community transmission and viral load kinetics of the SARS-CoV-2 delta (B.1.617.2) variant in vaccinated and unvaccinated individuals in the UK: a prospective, longitudinal, cohort study. Lancet Infect Dis 2022; 22:183.
  310. Kissler SM, Fauver JR, Mack C, et al. Viral Dynamics of SARS-CoV-2 Variants in Vaccinated and Unvaccinated Persons. N Engl J Med 2021; 385:2489.
  311. Puhach O, Adea K, Hulo N, et al. Infectious viral load in unvaccinated and vaccinated individuals infected with ancestral, Delta or Omicron SARS-CoV-2. Nat Med 2022; 28:1491.
  312. Jung J, Kim JY, Park H, et al. Transmission and Infectious SARS-CoV-2 Shedding Kinetics in Vaccinated and Unvaccinated Individuals. JAMA Netw Open 2022; 5:e2213606.
  313. Chia PY, Ong SWX, Chiew CJ, et al. Virological and serological kinetics of SARS-CoV-2 Delta variant vaccine breakthrough infections: a multicentre cohort study. Clin Microbiol Infect 2022; 28:612.e1.
  314. Feng S, Phillips DJ, White T, et al. Correlates of protection against symptomatic and asymptomatic SARS-CoV-2 infection. Nat Med 2021; 27:2032.
  315. Gilbert PB, Montefiori DC, McDermott AB, et al. Immune correlates analysis of the mRNA-1273 COVID-19 vaccine efficacy clinical trial. Science 2022; 375:43.
  316. Wesselink AK, Hatch EE, Rothman KJ, et al. A Prospective Cohort Study of COVID-19 Vaccination, SARS-CoV-2 Infection, and Fertility. Am J Epidemiol 2022; 191:1383.
  317. Chen F, Zhu S, Dai Z, et al. Effects of COVID-19 and mRNA vaccines on human fertility. Hum Reprod 2021; 37:5.
  318. Updated GTH statement on vaccination with the AstraZeneca COVID-19 vaccine, as of March 22, 2021. https://gth-online.org/wp-content/uploads/2021/03/GTH_Stellungnahme_AstraZeneca_engl._3_22_2021.pdf (Accessed on March 28, 2021).
  319. Pai M, Grill A, Ivers A, et al. Vaccine-Induced Prothrombotic Immune Thrombocytopenia (VIPIT) Following AstraZeneca COVID-19 Vaccination. https://covid19-sciencetable.ca/sciencebrief/vaccine-induced-prothrombotic-immune-thrombocytopenia-vipit-following-astrazeneca-covid-19-vaccination/ (Accessed on March 31, 2021).
  320. Schultz NH, Sørvoll IH, Michelsen AE, et al. Thrombosis and Thrombocytopenia after ChAdOx1 nCoV-19 Vaccination. N Engl J Med 2021; 384:2124.
  321. Greinacher A, Thiele T, Warkentin TE, et al. Thrombotic Thrombocytopenia after ChAdOx1 nCov-19 Vaccination. N Engl J Med 2021; 384:2092.
  322. Muir KL, Kallam A, Koepsell SA, Gundabolu K. Thrombotic Thrombocytopenia after Ad26.COV2.S Vaccination. N Engl J Med 2021; 384:1964.
  323. See I, Su JR, Lale A, et al. US Case Reports of Cerebral Venous Sinus Thrombosis With Thrombocytopenia After Ad26.COV2.S Vaccination, March 2 to April 21, 2021. JAMA 2021; 325:2448.
  324. Pavord S, Scully M, Hunt BJ, et al. Clinical Features of Vaccine-Induced Immune Thrombocytopenia and Thrombosis. N Engl J Med 2021; 385:1680.
  325. Updates on Thrombosis with Thrombocytopenia Syndrome (TTS). ACIP Meeting December 16, 2021. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2021-12-16/02-COVID-See-508.pdf (Accessed on December 17, 2021).
  326. MacNeil JR, Su JR, Broder KR, et al. Updated Recommendations from the Advisory Committee on Immunization Practices for Use of the Janssen (Johnson & Johnson) COVID-19 Vaccine After Reports of Thrombosis with Thrombocytopenia Syndrome Among Vaccine Recipients - United States, April 2021. MMWR Morb Mortal Wkly Rep 2021; 70:651.
  327. European Medicines Agency. COVID-19 Vaccine Janssen: EMA finds possible link to very rare cases of unusual blood clots with low blood platelets. https://www.ema.europa.eu/en/news/covid-19-vaccine-janssen-ema-finds-possible-link-very-rare-cases-unusual-blood-clots-low-blood (Accessed on May 10, 2021).
  328. European Medicines Agency. AstraZeneca’s COVID-19 vaccine: EMA finds possible link to very rare cases of unusual blood clots with low blood platelets. https://www.ema.europa.eu/en/news/astrazenecas-covid-19-vaccine-ema-finds-possible-link-very-rare-cases-unusual-blood-clots-low-blood (Accessed on April 07, 2021).
  329. Hippisley-Cox J, Patone M, Mei XW, et al. Risk of thrombocytopenia and thromboembolism after covid-19 vaccination and SARS-CoV-2 positive testing: self-controlled case series study. BMJ 2021; 374:n1931.
  330. Centers for Disease Control and Prevention. Thrombosis with thrombocytopenia syndrome (TTS) following Janssen COVID-19 vaccine. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2021-04-23/03-COVID-Shimabukuro-508.pdf (Accessed on April 23, 2021).
  331. https://www.ema.europa.eu/en/news/meeting-highlights-pharmacovigilance-risk-assessment-committee-prac-3-6-may-2021 (Accessed on May 26, 2021).
  332. Gargano JW, Wallace M, Hadler SC, et al. Use of mRNA COVID-19 Vaccine After Reports of Myocarditis Among Vaccine Recipients: Update from the Advisory Committee on Immunization Practices - United States, June 2021. MMWR Morb Mortal Wkly Rep 2021; 70:977.
  333. Oster ME, Shay DK, Su JR, et al. Myocarditis Cases Reported After mRNA-Based COVID-19 Vaccination in the US From December 2020 to August 2021. JAMA 2022; 327:331.
  334. Mevorach D, Anis E, Cedar N, et al. Myocarditis after BNT162b2 mRNA Vaccine against Covid-19 in Israel. N Engl J Med 2021; 385:2140.
  335. Witberg G, Barda N, Hoss S, et al. Myocarditis after Covid-19 Vaccination in a Large Health Care Organization. N Engl J Med 2021; 385:2132.
  336. Lai FTT, Li X, Peng K, et al. Carditis After COVID-19 Vaccination With a Messenger RNA Vaccine and an Inactivated Virus Vaccine : A Case-Control Study. Ann Intern Med 2022; 175:362.
  337. Mevorach D, Anis E, Cedar N, et al. Myocarditis after BNT162b2 Vaccination in Israeli Adolescents. N Engl J Med 2022; 386:998.
  338. Husby A, Hansen JV, Fosbøl E, et al. SARS-CoV-2 vaccination and myocarditis or myopericarditis: population based cohort study. BMJ 2021; 375:e068665.
  339. https://www.publichealthontario.ca/-/media/documents/ncov/vaccines/2021/11/myocarditis-pericarditis-mrna-vaccines.pdf?sc_lang=en (Accessed on January 28, 2022).
  340. Karlstad Ø, Hovi P, Husby A, et al. SARS-CoV-2 Vaccination and Myocarditis in a Nordic Cohort Study of 23 Million Residents. JAMA Cardiol 2022; 7:600.
  341. Le Vu S, Bertrand M, Jabagi MJ, et al. Age and sex-specific risks of myocarditis and pericarditis following Covid-19 messenger RNA vaccines. Nat Commun 2022; 13:3633.
  342. Block JP, Boehmer TK, Forrest CB, et al. Cardiac Complications After SARS-CoV-2 Infection and mRNA COVID-19 Vaccination - PCORnet, United States, January 2021-January 2022. MMWR Morb Mortal Wkly Rep 2022; 71:517.
  343. Witberg G, Magen O, Hoss S, et al. Myocarditis after BNT162b2 Vaccination in Israeli Adolescents. N Engl J Med 2022; 387:1816.
  344. Verma AK, Lavine KJ, Lin CY. Myocarditis after Covid-19 mRNA Vaccination. N Engl J Med 2021; 385:1332.
  345. Truong DT, Dionne A, Muniz JC, et al. Clinically Suspected Myocarditis Temporally Related to COVID-19 Vaccination in Adolescents and Young Adults: Suspected Myocarditis After COVID-19 Vaccination. Circulation 2022; 145:345.
  346. fda.gov/news-events/press-announcements/coronavirus-covid-19-update-july-13-2021 (Accessed on July 15, 2021).
  347. ema.europa.eu/en/documents/covid-19-vaccine-safety-update/covid-19-vaccine-safety-update-covid-19-vaccine-janssen-14-july-2021_en.pdf (Accessed on July 15, 2021).
  348. Abu-Rumeileh S, Abdelhak A, Foschi M, et al. Guillain-Barré syndrome spectrum associated with COVID-19: an up-to-date systematic review of 73 cases. J Neurol 2021; 268:1133.
  349. McDonnell EP, Altomare NJ, Parekh YH, et al. COVID-19 as a Trigger of Recurrent Guillain-Barré Syndrome. Pathogens 2020; 9.
  350. Patone M, Handunnetthi L, Saatci D, et al. Neurological complications after first dose of COVID-19 vaccines and SARS-CoV-2 infection. Nat Med 2021; 27:2144.
  351. Woo EJ, Mba-Jonas A, Dimova RB, et al. Association of Receipt of the Ad26.COV2.S COVID-19 Vaccine With Presumptive Guillain-Barré Syndrome, February-July 2021. JAMA 2021; 326:1606.
  352. Rosenblum HG, Hadler SC, Moulia D, et al. Use of COVID-19 Vaccines After Reports of Adverse Events Among Adult Recipients of Janssen (Johnson & Johnson) and mRNA COVID-19 Vaccines (Pfizer-BioNTech and Moderna): Update from the Advisory Committee on Immunization Practices - United States, July 2021. MMWR Morb Mortal Wkly Rep 2021; 70:1094.
  353. Hanson KE, Goddard K, Lewis N, et al. Incidence of Guillain-Barré Syndrome After COVID-19 Vaccination in the Vaccine Safety Datalink. JAMA Netw Open 2022; 5:e228879.
  354. Allen CM, Ramsamy S, Tarr AW, et al. Guillain-Barré Syndrome Variant Occurring after SARS-CoV-2 Vaccination. Ann Neurol 2021; 90:315.
  355. Maramattom BV, Krishnan P, Paul R, et al. Guillain-Barré Syndrome following ChAdOx1-S/nCoV-19 Vaccine. Ann Neurol 2021; 90:312.
  356. Li X, Raventós B, Roel E, et al. Association between covid-19 vaccination, SARS-CoV-2 infection, and risk of immune mediated neurological events: population based cohort and self-controlled case series analysis. BMJ 2022; 376:e068373.
  357. US Food and Drug Administration. Development and Licensure of Vaccines to Prevent COVID-19: Guidance for Industry. https://www.fda.gov/media/139638/download (Accessed on October 20, 2020).
  358. WHO Target Product Profiles for COVID-19 Vaccines. April 2020. https://www.who.int/publications/m/item/who-target-product-profiles-for-covid-19-vaccines (Accessed on November 06, 2020).
  359. Goodman JL, Grabenstein JD, Braun MM. Answering Key Questions About COVID-19 Vaccines. JAMA 2020; 324:2027.
  360. US Food and Drug Administration. Emergency Use Authorization for Vaccines to Prevent COVID-19: Guidance for Industry. October 2020. https://www.fda.gov/media/142749/download (Accessed on November 05, 2020).
  361. National Academies of Sciences, Engineering, and Medicine. 2020. Framework for Equitable Allocation of COVID-19 Vaccine. Washington, DC: The National Academies Press. https://www.nap.edu/catalog/25917/framework-for-equitable-allocation-of-covid-19-vaccine (Accessed on November 12, 2020).
  362. Centers for Disease Control and Prevention. How CDC Is Making COVID-19 Vaccine Recommendations. https://www.cdc.gov/coronavirus/2019-ncov/vaccines/recommendations-process.html (Accessed on November 12, 2020).
  363. WHO Strategic Advisory Group of Experts (SAGE) on Immunization. COVID-19 vaccines: Key documents. https://www.who.int/immunization/sage/covid-19_documents/en/ (Accessed on November 12, 2020).
  364. United States Department of Health and Human Services. From the Factory to the Frontlines: The Operation Warp Speed Strategy for Distributing a COVID-19 Vaccine. https://www.hhs.gov/sites/default/files/strategy-for-distributing-covid-19-vaccine.pdf (Accessed on November 18, 2020).
  365. Centers for Disease Control and Prevention. Frequently Asked Questions about COVID-19 Vaccination. https://www.cdc.gov/coronavirus/2019-ncov/vaccines/faq.html (Accessed on November 18, 2020).
  366. World Health Organization. Ten threats to global health in 2019. https://www.who.int/news-room/spotlight/ten-threats-to-global-health-in-2019 (Accessed on November 12, 2020).
  367. https://www.vitaltalk.org/wp-content/uploads/Communication-skills-for-the-COVID-vaccine_v1.2-1.pdf (Accessed on January 21, 2021).
  368. Centers for Disease Control and Prevention. Talking to Recipients about COVID-19 Vaccines. https://www.cdc.gov/vaccines/covid-19/hcp/index.html (Accessed on January 21, 2021).
  369. Lazarus JV, Ratzan SC, Palayew A, et al. A global survey of potential acceptance of a COVID-19 vaccine. Nat Med 2021; 27:225.
  370. Daly M, Jones A, Robinson E. Public Trust and Willingness to Vaccinate Against COVID-19 in the US From October 14, 2020, to March 29, 2021. JAMA 2021; 325:2397.
  371. Szilagyi PG, Thomas K, Shah MD, et al. National Trends in the US Public's Likelihood of Getting a COVID-19 Vaccine-April 1 to December 8, 2020. JAMA 2020.
  372. Gadoth A, Halbrook M, Martin-Blais R, et al. Cross-sectional Assessment of COVID-19 Vaccine Acceptance Among Health Care Workers in Los Angeles. Ann Intern Med 2021; 174:882.
  373. Shaw J, Stewart T, Anderson KB, et al. Assessment of US Healthcare Personnel Attitudes Towards Coronavirus Disease 2019 (COVID-19) Vaccination in a Large University Healthcare System. Clin Infect Dis 2021; 73:1776.
  374. Nguyen KH, Srivastav A, Razzaghi H, et al. COVID-19 Vaccination Intent, Perceptions, and Reasons for Not Vaccinating Among Groups Prioritized for Early Vaccination - United States, September and December 2020. MMWR Morb Mortal Wkly Rep 2021; 70:217.
  375. Fisher KA, Bloomstone SJ, Walder J, et al. Attitudes Toward a Potential SARS-CoV-2 Vaccine : A Survey of U.S. Adults. Ann Intern Med 2020; 173:964.
  376. El-Mohandes A, White TM, Wyka K, et al. COVID-19 vaccine acceptance among adults in four major US metropolitan areas and nationwide. Sci Rep 2021; 11:21844.
  377. Centers for Disease Control and Prevention. Vaccine Safety Datalink (VSD). https://www.cdc.gov/vaccinesafety/ensuringsafety/monitoring/vsd/index.html (Accessed on November 05, 2020).
  378. McCarthy NL, Gee J, Weintraub E, et al. Monitoring vaccine safety using the Vaccine Safety Datalink: utilizing immunization registries for pandemic influenza. Vaccine 2011; 29:4891.
  379. Yih WK, Lee GM, Lieu TA, et al. Surveillance for adverse events following receipt of pandemic 2009 H1N1 vaccine in the Post-Licensure Rapid Immunization Safety Monitoring (PRISM) System, 2009-2010. Am J Epidemiol 2012; 175:1120.
  380. Lee GM, Romero JR, Bell BP. Postapproval Vaccine Safety Surveillance for COVID-19 Vaccines in the US. JAMA 2020; 324:1937.
  381. Centers for Disease Control and Prevention. Enhanced safety monitoring for COVID-19 vaccines in early phase vaccination. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2020-09/COVID-03-Shimabukuro.pdf (Accessed on November 18, 2020).
  382. Fine PEM, Mulholland K, Scott JA, Edmunds WJ. Community Protection. In: Plotkin’s Vaccines, 7th, Plotkin SA, Orenstein WA, Offit PA, Edwards KM (Eds), Elsevier, 2018. p.1512.
  383. Levine EM, Davey AS, Houstan AM. Legal Issues. In: Plotkin’s Vaccines, 7th, Plotkin SA, Orenstein WA, Offit PA, Edwards KM (Eds), Elsevier, 2018. p.1601.
  384. Health Resources and Services Administration. Countermeasures Injury Compensation Program. https://www.hrsa.gov/cicp (Accessed on November 20, 2020).
Topic 129849 Version 169.0

References