ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

COVID-19: Vaccines

COVID-19: Vaccines
Literature review current through: Jan 2024.
This topic last updated: Nov 14, 2023.

INTRODUCTION — Vaccines to prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are considered the most promising approach for curbing the coronavirus disease 2019 (COVID-19) pandemic. Several COVID-19 vaccines are available globally. The World Health Organization (WHO) maintains an updated list of vaccine candidates under evaluation and available for administration [1].

This topic will cover vaccines for SARS-CoV-2, with a focus on vaccines available in the United States. Other aspects related to the 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 prevent attachment to the host cell and neutralize the virus [3]. Over time, the messenger ribonucleic acid (mRNA) vaccines have been reformulated to target the mutations in the spike proteins of the various SARS-CoV-2 variants. (See 'Available vaccines' below.)

Vaccine platforms – COVID-19 vaccines have been and are being developed using several different platforms [3]. Some of these are traditional approaches, such as inactivated virus or live attenuated viruses, which have been used for influenza vaccines and measles vaccines, 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 mRNA and DNA (deoxyribonucleic acid) vaccines, had never been employed in a licensed vaccine prior to the COVID-19 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 when administered systemically [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 (severe acute respiratory syndrome coronavirus 1) and MERS-CoV (Middle East respiratory syndrome-related coronavirus) raised concerns for enhanced disease with vaccination when some vaccinated animals developed non-neutralizing antibody and Th2 cell responses that were associated with eosinophilic lung inflammation following challenge with wild-type virus [6-8]. No vaccine-enhanced disease was seen in any human COVID-19 vaccine studies. Nevertheless, specific immunologic parameters were targeted for animal and human studies to reduce the risk of enhanced disease [9]. The desired immune responses included neutralizing antibody and Th1-polarized cellular immune responses.

APPROACH TO VACCINATION IN THE UNITED STATES

Available vaccines — In the United States, three COVID-19 vaccines are available (table 2):

Two mRNA vaccines

Moderna COVID-19 vaccine (2023-2024 Formula, based on XBB.1.5) – This is approved for individuals 12 years and older and available under emergency use authorization (EUA) for children aged 6 months to 11 years [10,11].

Pfizer-BioNTech COVID-19 vaccine (2023-2024 Formula, based on XBB.1.5) – This is approved for individuals 12 years and older and available under EUA for children aged 6 months to 11 years [12,13].

Each mRNA vaccine began as a monovalent formulation with an antigenic target based on the original Wuhan SARS-CoV-2 strain. In September 2022, bivalent formulations with antigenic targets based on the original Wuhan SARS-CoV-2 strain and the BA.4/BA.5 Omicron subvariants were also introduced and became the only mRNA COVID-19 vaccine formulations available in April 2023. As of September 2023, the bivalent formulations were replaced with a monovalent formulation with an antigenic target based on the XBB.1.5 Omicron variant. That variant was selected as the model for the updated formulation in the summer of 2023, based on projections for fall 2023 variant proportions [14].

An adjuvanted recombinant protein vaccine (monovalent):

Novavax COVID-19 vaccine, adjuvanted (2023-2024 Formula) – This is available under EUA for individuals aged 12 years or older [15].

The Novavax COVID-19 has also been updated to target the XBB.1.5 Omicron variant.

As of May 2023, all doses of the adenoviral vector vaccine Janssen/Johnson & Johnson COVID-19 vaccine (Ad26.COV2.S) have expired [16], and this vaccine is no longer available in the United States.

Indications and vaccine selection — We suggest COVID-19 vaccination with a 2023-2024 formula vaccine for all individuals aged six months and older; this is consistent with recommendations from the Centers for Disease Control and Prevention (CDC). We particularly encourage vaccination in individuals 65 years or older, immunocompromised individuals, and individuals with multiple medical comorbidities, as they are at highest risk of severe outcomes with COVID-19 and are most likely to benefit from vaccination (table 3). In the United States, the available vaccines are Moderna COVID-19 vaccine 2023-2024 Formula, Pfizer COVID-19 vaccine 2023-2024 Formula, and Novavax COVID-19 vaccine 2023-2024 Formula (table 2).

Vaccines available in the United States reduce the risk of COVID-19, especially severe/critical disease, and have been associated with substantial reductions in COVID-19-associated hospitalizations and deaths [17-24], even in the context of variants that partially evade vaccine-induced immune responses. The vaccines are safe, with only very rare associated severe adverse events. The benefits and potential risks of vaccines are discussed elsewhere. (See 'Benefits of vaccination' below and 'Risks of vaccination' below.)

Although the risk of severe disease has decreased substantially since the start of the pandemic, COVID-19 remains an important cause of hospitalization and death, with the highest rates in individuals older than 65 years or younger than six months. In the United States, COVID-19-associated hospitalizations substantially exceeded influenza-associated hospitalizations for all individuals in 2022 and for adults in the first half of 2023. A similar proportion of either COVID-19- or influenza-associated hospitalizations results in intensive care unit (ICU) admission and death [25]. Although most individuals with COVID-19-associated hospitalization have medical comorbidities (and most hospitalized adults have multiple comorbidities), approximately 40 to 60 percent of hospitalized children aged six months to four years have no underlying conditions, and 15 to 50 percent of those aged 6 months to 49 years admitted to the ICU had no underlying conditions. Thus, although the potential benefit of COVID-19 vaccination in reducing the risk of severe disease is greatest in those at highest risk, the risk of severe disease exists for all individuals.

Dose and interval (for immunocompetent individuals) — Vaccine dosing and intervals are also listed in the table (table 2). Recommended vaccine schedules for individuals with immunocompromising conditions are discussed elsewhere. (See 'Immunocompromised individuals' below.)

Children aged six months to four years — For children under five years of age, 2023-2024 formula mRNA vaccines are recommended for all vaccine doses [26]. The dosing schedule depends on their prior COVID-19 vaccination status.

For immunocompetent children who have not previously received a COVID-19 vaccine:

Moderna COVID-19 vaccine (2023-2024 Formula) – Two intramuscular doses of 25 mcg (0.25 mL of the dark blue capped vial) given four to eight weeks apart [10].

Pfizer COVID-19 vaccine (2023-2024 Formula) – Three intramuscular doses of 3 mcg (0.3 mL of the yellow capped vial) [12]. The first two doses are given three to eight weeks apart, and the third dose is given at least eight weeks after the second.

For immunocompetent children who have previously received at least one vaccine dose, the number of subsequent doses with the 2023-2024 formula mRNA vaccines depends on how many doses and which vaccine they had previously received:

Prior Moderna COVID-19 vaccine doses [10]:

-Those who have received a single prior dose receive a single intramuscular dose of 25 mcg (0.25 mL of the dark blue-capped vial) Moderna COVID-19 vaccine (2023-2024 Formula) four to eight weeks later.

-Those who have received two or more prior doses receive a single intramuscular dose of 25 mcg (0.25 mL of the dark blue-capped vial) Moderna COVID-19 vaccine (2023-2024 Formula) at least eight weeks after the last dose.

Prior Pfizer COVID-19 vaccine doses [12]:

-Those who have received a single prior dose receive two intramuscular doses of 3 mcg (0.3 mL of the yellow-capped vial) Pfizer COVID-19 vaccine (2023-2024 Formula): one dose three weeks after the last dose and a second dose at least eight weeks later.

-Those who have received two or more prior doses receive a single intramuscular dose of 3 mcg (0.3 mL of the yellow-capped vial) Pfizer COVID-19 vaccine (2023-2024 Formula). It is given at least eight weeks after the last dose.

Immunocompetent children in this age group who have received at least two doses of Moderna COVID-19 vaccine, at least one of which is the 2023-2024 formula mRNA vaccine, or at least three doses of Pfizer COVID-19 vaccine, at least one of which is the 2023-2024 formula mRNA vaccine, do not need additional vaccinations. All vaccine doses should be from the same manufacturer [26].

Vaccine recommendations for children with immunocompromising conditions (table 4) are discussed in detail elsewhere [26]. (See 'Immunocompromised individuals' below.)

Clinical data evaluating COVID-19 vaccine effectiveness in young children are limited. Support for their use comes mainly from immunogenicity bridging studies, some unpublished, in these age groups. (See 'Children' below.)

Although a single mRNA vaccine dose is recommended for other age groups, serologic surveillance suggests that young children in the United States are less likely to have pre-existing immunity (whether from infection or vaccination) than older children and adults, so they may be more likely to need more than one dose to maximize immunogenicity [27]. Additionally, a prime-boost model of vaccination in general is considered important for optimizing vaccine response in young children.

Children aged 5 to 11 years — For children aged 5 to 11 years, a single dose of a 2023-2024 formula mRNA vaccine is recommended, regardless of whether the recipient is unvaccinated or had previously received one or more vaccine doses [26].

Moderna COVID-19 vaccine (2023-2024 Formula) – A single intramuscular dose of 25 mcg (0.25 mL of the dark blue-capped vial) [10].

Pfizer COVID-19 vaccine (2023-2024 Formula) – A single intramuscular dose of 10 mcg (0.3 mL of the blue-capped vial) [12].

Vaccine recommendations for children with immunocompromising conditions (table 4) are discussed in detail elsewhere [26]. (See 'Immunocompromised individuals' below.)

Prior recommendations had included a two-dose vaccine series for unvaccinated individuals. However, seroprevalence data suggest that the vast majority (over 96 percent) of individuals in the United States have pre-existing immunity to SARS-CoV-2, either through vaccination or infection [27]. Thus, a single vaccine dose, which has been associated with increases in relative effectiveness against infection and severe disease in individuals with pre-existing immunity from prior vaccination, is expected to be sufficient to improve protection for immunocompetent individuals who have not yet been vaccinated. (See 'Protection against severe disease or death' below.)

Rationale for vaccination in children is discussed elsewhere. (See 'Children' below.)

Adolescents and adults aged 12 years and older — For individuals older than 12 years of age, a single dose of a 2023-2024 formula mRNA vaccine is recommended, regardless of whether the recipient is unvaccinated or had previously received one or more vaccine doses [26]. If the individual had received prior vaccine doses, the 2023-2024 formula vaccine dose is given at least two months after the last dose:

Moderna COVID-19 vaccine (2023-2024 Formula) – A single intramuscular dose of 50 mcg (0.5 mL) [10].

Pfizer COVID-19 vaccine (2023-2024 Formula) – A single intramuscular dose of 30 mcg (0.3 mL) [12].

The Novavax COVID-19 vaccine (2023-2024 Formula) is another option for this age group. For individuals who have not previously received a COVID-19 vaccine, it is given as two intramuscular doses of 5 mcg spike protein/50 mcg adjuvant three to eight weeks apart; for individuals who have received at least one prior COVID-19 vaccine but not a 2023-2024 formula vaccine, it is given as a single dose at least two months after the last dose [15].

Vaccine recommendations for individuals with immunocompromising conditions (table 4) are discussed in detail elsewhere [26]. (See 'Immunocompromised individuals' below.)

Although prior recommendations had included a two-dose primary series for unvaccinated individuals, seroprevalence data suggest that the vast majority (over 96 percent) of individuals in the United States have pre-existing immunity to SARS-CoV-2, either through vaccination or infection [28,29]. Thus, a single vaccine dose, which has been associated with increases in relative effectiveness against infection and severe disease in individuals with pre-existing immunity from prior vaccination, is expected to be sufficient to improve protection in immunocompetent individuals without a history of COVID-19 vaccination. More than one dose of a 2023-2024 formula vaccine is not recommended for immunocompetent adults, regardless of age.

Among individuals who have already been vaccinated, the rationale for an additional updated vaccine dose is to restore protection that wanes with time and to try to improve the immune response against currently circulating viral variants. This is especially relevant in adults ≥65 years old, among whom the rate of COVID-19-associated hospitalization and death is higher than in any other age group [30]. In the United States, approximately 95 percent of adults ≥65 years old have received a complete primary COVID-19 vaccine series [27], so an additional vaccine dose serves as booster dose, which has been associated with increases in relative effectiveness against infection and severe disease, as discussed in detail elsewhere. (See 'Protection against severe disease or death' 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 3). (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 5).

Mixing vaccine types — For individuals six years of age and older who had previously received an older monovalent or bivalent vaccine, any of the 2023-2024 formula vaccines can be subsequently used; they do not have to use the vaccine from the same manufacturer as the original doses. (See 'Dose and interval (for immunocompetent individuals)' above.)

For those who received a non-mRNA vaccine for the primary series, studies indicate robust antibody responses with an mRNA booster dose (ie, a heterologous boost) [31-33] and greater effectiveness than with homologous boosting [34-36]. As an example, in a study of nearly five million United States veterans, receipt of an mRNA vaccine boost after a primary Janssen/Johnson & Johnson COVID-19 vaccine dose was associated with a lower risk of infection than receipt of a Janssen/Johnson & Johnson COVID-19 vaccine 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 [34]. Immunogenicity studies support these findings and suggested higher binding and neutralizing antibody levels with a Moderna COVID-19 vaccine boost than with a Pfizer COVID-19 vaccine boost [32]. No safety concerns were identified; the frequency and duration of systemic symptoms (eg, fever, chills, myalgias) may be slightly higher with Moderna COVID-19 vaccine booster doses.

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 [26].

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 was given concomitantly with either an influenza vaccine or placebo [37].

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 [26]. 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 of 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'.)

Considerations for 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 [26].

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 [26]. 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, are at increased risk for SARS-CoV-2 exposure, and if the MIS was not associated with COVID-19 vaccination [26]. In a study of 186 individuals ≥5 years old with a history of MIS ≥90 days prior to vaccine receipt, the side effect profile of mRNA vaccination was similar to that in the general population; no cases of myocarditis or recurrent MIS were observed [38].

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 likely improves the durability and breadth of protection [39-41]. In multiple observational studies of individuals with prior infection, vaccination has been associated with a lower risk of subsequent reinfection and hospitalization [42-49]; it has also been associated with a lower risk of breakthrough infection compared with vaccination in individuals without prior infection [50,51].

Vaccination has been associated with greater protection against hospitalization for COVID-19 compared with prior infection in some studies [52]. However, one study suggested that when the 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) [53].

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 [54].

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 [26,55,56]. 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 postexposure quarantine period to avoid inadvertent exposures to others in the event of infection [26]. 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, postexposure vaccination would likely not reduce the risk of infection following that specific exposure.

Immunocompromised individuals — We recommend that individuals who have an immunocompromising condition or are taking immunosuppressive agents undergo COVID-19 vaccination (table 4). 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. Given the potential for reduced vaccine response in such individuals, the recommended vaccine schedule is distinct, as outlined below:

Conditions that warrant an adjusted vaccine schedule – Moderate to severe immunocompromising conditions that warrant a modified vaccine schedule include active use of chemotherapy for cancer, hematologic malignancies, hematopoietic stem cell or solid organ transplant, advanced HIV (human immunodeficiency virus) infection with CD4 cell count <200 cells/microL or untreated HIV, moderate or severe primary immunodeficiency disorder, and use of immunosuppressive medications (eg, mycophenolate mofetil, rituximab, prednisone >20 mg/day for >14 days) (table 4) [57]. This list is not exhaustive, however; other conditions, such as impaired splenic function [58], may also warrant additional vaccine doses than recommended in the general population.

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 [59-66]. In a cohort study of over one 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 [59]. Lower vaccine effectiveness against hospitalization for COVID-19 in immunocompromised patients has also been suggested by smaller case-control studies [60]. In studies of individuals hospitalized with COVID-19 despite vaccination, a high proportion (eg, 40 percent) have been immunocompromised [61].

In particular, transplant recipients and individuals taking B cell-depleting agents (particularly within the prior six months) may have a suboptimal vaccine response [65,67-72]. Among transplant recipients, factors associated with a higher rate of nonresponse include use of antimetabolites (eg, mycophenolate mofetil, azathioprine) and a shorter time since transplantation. (See "COVID-19: Issues related to solid organ transplantation", section on 'Vaccination'.)

Recommended vaccine series in immunocompromised individuals – The CDC recommends that individuals with moderate to severe immunocompromising conditions (table 4) receive at least three vaccine doses, at least one of which is a 2023-2024 formula mRNA vaccine (Moderna COVID-19 vaccine 2023-2024 Formula or Pfizer COVID-19 vaccine 2023-2024 Formula). Thus, initial vaccine recommendations depend on the vaccination history:

Unvaccinated individuals should receive three 2023-2024 formula vaccine doses.

-If Moderna COVID-19 vaccine is used, the second dose is given four weeks after the first and the third is given at least four weeks later.

-If Pfizer COVID-19 vaccine is used, the second dose is given three weeks after the first, and the third is given at least eight weeks later (for individuals six months to four years of age) or at least four weeks later (for individuals five years of age and older).

Individuals who have received only one previous vaccine dose should receive two 2023-2024 formula vaccine doses.

-If Moderna COVID-19 vaccine is used, the first 2023-2024 formula dose is given four weeks after the last vaccine dose, and the second 2023-2024 formula dose is given at least four weeks later.

-If Pfizer COVID-19 vaccine is used, the first 2023-2024 formula dose is given three weeks after the last vaccine dose, and the second 2023-2024 formula dose is given at least eight weeks later (for individuals six months to four years of age) or at least four weeks later (for individuals five years of age and older).

Individuals who have received two or more previous vaccine doses should receive one 2023-2024 formula vaccine dose.

-For those who received two prior vaccine doses, the 2023-2024 formula vaccine dose is given at least four weeks after the last dose.

-For those who received three or more prior vaccine doses, the 2023-2024 formula vaccine dose is given at least eight weeks after the last dose.

The specific dose given for each vaccine depends on the age of the individual and is the same as for individuals without immunocompromising conditions (table 2). For individuals aged 12 years and older, the Novavax COVID-19 vaccine (2023-2024 Formula) is another option.

Support for multiple vaccine doses for individuals with immunocompromising conditions comes from multiple observational studies in which receipt of three doses of mRNA monovalent vaccines was associated with higher vaccine effectiveness than two doses [73,74]. In studies of transplant recipients who received a third dose of mRNA monovalent 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 [75-79]. 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 [80].

Additional vaccine doses in individuals who have received at least three vaccine doses, including at least one 2023-2024 formula vaccine dose – For individuals with moderate to severe immunocompromising conditions (table 4) who have received the vaccines recommended above, the CDC allows for an additional 2023-2024 formula vaccine dose at least two months later.

We individualize the decision to administer this additional dose and are more likely to suggest it for individuals with expected high risk of exposure and profound immunosuppression (such as B cell depletion, recent or complicated hematopoietic stem cell transplant [HCT], or chimeric antigen receptor [CAR]-T cell therapy). The clinical effect of multiple 2023-2024 formula vaccines is uncertain, and data are limited. The rationale is that, in such patients, repeated vaccination can boost the immune response. (See 'Protection against severe disease or death' below.)

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 [81]. (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 stem cell transplant (HCT) or chimeric antigen receptor (CAR)-T cell therapy, the CDC recommends repeat vaccination at least three months after the transplant or CAR-T administration [26]. For those who were vaccinated during a limited course of a B cell-depleting therapy, repeat vaccination is suggested six months following therapy. (See "Immunizations in hematopoietic cell transplant candidates and recipients".)

Continued use of protective measures – We advise immunocompromised patients to maintain personal measures to try to minimize exposure to SARS-CoV-2 (eg, avoiding crowds when possible, masking if the likelihood of exposure is high) 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.

Limited role for post-vaccination serology – At this time, antibody testing is not recommended to determine response to vaccination [26]. Precise immune correlates of protection against severe disease remain uncertain; vaccination may elicit cellular but not humoral responses among some immunocompromised patients, and those responses may protect against severe infection [82]. Furthermore, heterogeneity in the sensitivity and specificity of available serologic tests complicates interpretation of results. (See 'Antibody responses and immune correlates of protection' below and 'Limited role for post-vaccination testing' above.)

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 inborn errors of immunity", 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. Dosing in children depends on age and is discussed elsewhere. (See 'Dose and interval (for immunocompetent individuals)' above.)

Benefits of COVID-19 vaccination in children – COVID-19 vaccination is associated with reductions in severe adverse outcomes related to COVID-19 among children and adolescents. Randomized trials demonstrated that antibody responses to the mRNA vaccines in children six months and older are as high as (or higher than) those in older individuals [83-88]. Efficacy data from these trials were limited, in part because of the low number of COVID-19 cases among study participants. However, subsequent observational studies suggest that vaccination is associated with reductions in COVID-19-related hospitalization, ICU admission, and death in adolescents, and reductions in hospitalization in younger school-aged children [89-101]. As an example, in a meta-analysis of 17 studies that included over 11 million vaccinated and 2.5 million unvaccinated children between 5 and 11 years old, COVID-19 vaccination was associated with lower rates of COVID-19 associated hospitalization (OR 0.32) and MIS (OR 0.05) [100]. Limited observational data suggest that vaccination is also associated with reductions in medically attended and severe infection among children younger than five years old [102,103]. As in adults, vaccine effectiveness in children of all ages wanes over time [98]. (See 'Benefits of vaccination' below.)

For children under five years old, assessment of vaccine efficacy is primarily extrapolated from studies demonstrating that the neutralizing activity elicited by vaccination is comparable with or higher than levels associated with protection in older populations.

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 their vaccination [104]. Furthermore, even with the lower risk of severe disease among children, the number of COVID-19 deaths among those 6 months to 18 years old during the pandemic (even during 2022 when disease severity was lower) exceeded the prevaccination era mortality rates of infections for which childhood vaccines are routinely recommended (eg, rotavirus, meningococcal disease, varicella) [105].

Risks and concerns in children – The safety profile of COVID-19 vaccines in children is similar to that in older individuals; mild local and systemic reactions are common but serious adverse events are rare. (See 'Risks of vaccination' below.)

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 [106]. No cases of myocarditis thought related to vaccine were reported in the trials of the mRNA vaccines in young children [107,108]. In a review of the Vaccine Adverse Event Reporting System (VAERS) following administration of approximately 8.7 million doses of Pfizer COVID-19 vaccine to children aged 5 to 11 years in the United States, there were 11 verified reports of myocarditis in this age group [109]; no cases were reported following 1.5 million doses among children six months to five years of age [110]. 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 [111,112]. (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 were another concern. Vaccine trials in this age group have not identified a potential signal, although rare case reports of MIS in children and adults following vaccination highlight the importance of monitoring for this possible adverse effect [113-115]. Nevertheless, evidence suggests that vaccination may protect against MIS-C [116,117]. In a study of 102 patients aged 12 to 18 years hospitalized with MIS-C, 95 percent were unvaccinated; of the five patients with MIS-C who had previously received primary series of Pfizer COVID-19 vaccine, none required invasive respiratory or cardiovascular support [117]. 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 Advisory Committee on Immunization Practices (ACIP)-approved vaccines for eligible children through 18 years of age [118,119]. Eligible children include those on Medicaid, those who are completely uninsured, and American Indian/Alaskan Natives. Approximately 50 percent of United States children are covered by the VFC. In addition, federal grants to states can be used to purchase vaccines to cover other children.

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 [120,121]. Nevertheless, almost all immunocompetent recipients develop sufficiently high antibody levels, regardless of side effects. Among mRNA vaccines, Pfizer COVID-19 vaccine may be associated with slightly lower rates of local and systemic reactions compared with mRNA-1273 [122]. Rates of reactions for the distinct vaccines are discussed in detail elsewhere. (See 'Common adverse effects' 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 [26]. 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 [123,124]. Aspirin is not recommended for individuals ≤18 years old because of the risk of Reye syndrome.

Syncope – Syncope has been reported following receipt of other injectable vaccines, particularly among adolescents and young adults [125]. Monitoring individuals who have previously had syncope or may be at higher risk (eg, adolescents) for 15 minutes following COVID-19 vaccine receipt 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. These issues and safety concerns with other COVID-19 vaccines are discussed in detail elsewhere. (See 'Rare but serious associated events' 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 venous thromboembolic events without thrombocytopenia, deep vein thrombosis or pulmonary embolism, Bell palsy, and tinnitus) have been reported in vaccine recipients but have not been identified as causally related vaccine-associated adverse events. (See 'Other reactions (without established association)' below.)

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

Contraindications and precautions (including allergies)

Contraindications – The following are the only contraindications to COVID-19 vaccination [26]:

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 and do not represent a contraindication to vaccine. (See "COVID-19: Allergic reactions to SARS-CoV-2 vaccines", section on 'Delayed-onset urticarial reactions'.)

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.

The mRNA vaccines, Pfizer-BioNTech COVID-19 vaccine and Moderna COVID-19 vaccine, each contain polyethylene glycol (PEG). However, it is a smaller molecular weight and less allergenic PEG than that present in other products, and multiple studies have documented safe receipt of the mRNA vaccines in individuals with known or suspected allergy to other PEG-containing products. The Novavax COVID-19 vaccine contains polysorbate, which is structurally related to PEG. Allergic reaction to polysorbate remains listed as a contraindication to Novavax COVID-19 vaccine and a precaution to mRNA vaccines. These issues are discussed in detail elsewhere. (See "COVID-19: Allergic reactions to SARS-CoV-2 vaccines", section on 'Uncertain role of polyethylene glycol'.)

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. Many individuals with apparent anaphylaxis after COVID-19 vaccination were able to receive a subsequent dose uneventfully after the reaction was determined not to be immunoglobulin E (IgE)-mediated. 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-C, or Guillain-Barré syndrome (GBS). These issues are discussed elsewhere. (See 'Myocarditis' below and 'History of SARS-CoV-2 infection' above and 'Guillain-Barré syndrome' below.)

Caution may be warranted prior to administering any vaccine in certain rare but life-threatening conditions, such as acquired thrombotic thrombocytopenic purpura and capillary leak syndrome, exacerbations of which have been reported following COVID-19 vaccination [126,127]. (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 Novavax COVID-19 vaccine. Although very rare cases of unusual types of thrombosis associated with thrombocytopenia have been reported following vaccination with certain adenoviral vector vaccines, 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 [128], 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 one week after mRNA COVID-19 vaccination have been reported, with recurrence occurring in some individuals after repeat vaccination [129]. This may occur more frequently with Moderna COVID-19 vaccine than with Pfizer COVID-19 vaccine [130]. 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 [26]. (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 [26]. 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 [131]. 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:

Allergy-related contraindication to a different type of COVID-19 vaccine (see 'Contraindications and precautions (including allergies)' above)

Nonsevere, immediate (onset within four hours) allergic reaction after a previous dose of COVID-19 vaccine

History of anaphylaxis after non-COVID-19 vaccines or injectable therapies

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 6). (See "Anaphylaxis: Acute diagnosis" and "Anaphylaxis: Emergency treatment" and 'Ongoing safety assessment and reporting of adverse events' 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.

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.)

BENEFITS OF VACCINATION

Protection against severe disease or death — Vaccines available in the United States reduce the risk of severe or critical COVID-19 and have been associated with substantial reductions in COVID-19-associated hospitalizations and deaths [17-24,132], even in the context of variants that partially evade vaccine-induced immune responses. Booster doses have been associated with restoration of vaccine effectiveness against severe infection that wanes over time. Hospitalization and mortality rates for COVID-19 have been consistently higher among unvaccinated compared with vaccinated individuals, particularly in those who received a vaccine dose within the several months preceding [133]. In addition to direct reductions in COVID-19-associated morbidity and mortality, vaccination has been associated with lower non-COVID-19 mortality rates as well [134].

Risk reduction compared with no vaccination – The large randomized trials evaluating the mRNA vaccines (the Pfizer COVID-19 vaccine and the Moderna COVID-19 vaccine) and the Novavax COVID-19 vaccine demonstrated 96 to 100 percent vaccine efficacy against severe infection; however, rates of severe infection in these trials were very low [135-139]. Thus, most data demonstrating reductions in severe disease and death with vaccination are from large observational and population-based studies [17-24,132].

As an example, in a study that evaluated over 8000 COVID-19-associated deaths in the United States between September 2022 and April 2023, receipt of vaccination, including a bivalent booster dose, was consistently associated with lower mortality compared with no vaccination; without vaccination, the risk of death was 7.3 to 16.3 times greater, depending on the prevalent Omicron sublineage [138]. In another 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 86 to 90 percent against hospitalization and 90 to 93 percent against death [139]. At 12 months, the same measures were 60 to 65 percent and 70 to 75 percent.

Risk of severe disease among vaccinated individuals – Among vaccinated individuals, the overall risk of severe COVID-19 is low [140-144]. In a study of over one 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 [144]. A history of vaccination has also been associated with lower rates of myocardial infarction, acute stroke, and other major adverse cardiovascular outcomes in patients with COVID-19 [143,145]. Risk factors for severe COVID-19 after vaccination are similar to those for unvaccinated individuals: older age (>65 years), immunocompromising conditions, and multiple comorbidities [144,146].

Impact of variants and time since vaccination (and effect of booster doses) – Vaccine effectiveness against severe disease wanes with time since the last vaccination, although the levels of protection against hospitalization and death are higher and sustained for longer than protection against symptomatic infection [147-153]. Booster doses, both with monovalent and bivalent vaccines, have consistently been associated with greater protection against severe disease compared with more remote vaccination, although the effect of boosting also wanes over time [139,154-167]. The pace and degree of waning following primary and booster vaccinations have varied across studies, however, and waning may be faster in the setting of Omicron compared with other variants [168-177]. In a systematic review of observational studies evaluating protection against hospitalization after emergence of the Omicron variants, pooled vaccine effectiveness following a booster dose was 89 percent in the first month and 71 percent by four to six months [178]. However, in another study of over 65,000 hospitalized adults during Omicron prevalence in the United States, vaccine effectiveness (which included a bivalent booster vaccine dose) against COVID-19-associated hospitalization compared with no vaccination waned from 62 percent during the first 60 days after the vaccine dose to 24 percent three to six months later [179].

Decreases in observed vaccine effectiveness against severe disease 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 more associated with milder infection.

Protection against symptomatic infection — Although the initial clinical trials reported extremely high efficacy rates with the Pfizer COVID-19 vaccine (90 to 100 percent [83,84,135,180-182]), the Moderna COVID-19 vaccine (86 to 100 percent [107,136,183]), and the Novavax COVID-19 vaccine (80 to 90 percent [137,184,185]) in preventing laboratory-confirmed symptomatic infection within the first two months of infection, their observed effectiveness against infection has decreased over time because of waning immunity and immune evasion by certain circulating SARS-CoV-2 variants.

Impact of variants and time since vaccination (and effect of booster doses) – Multiple observational studies have consistently suggested that vaccine protection against symptomatic infection is lower with Omicron compared with other variants and wanes over time (after both primary series and booster vaccinations and with both monovalent and bivalent vaccines) in children and adults [186-198]. In a systematic review of 40 studies evaluating vaccine effectiveness since the emergence of the Omicron variant, pooled estimates for vaccine efficacy against symptomatic disease decreased from 53 to 14 percent at one and six months after completing the primary vaccine series; a booster dose was associated with restoration of effectiveness, although this also waned to <20 percent after nine months [197]. The estimated half-life of vaccine effectiveness was 111 days.

The relative effects of booster doses to temporarily restore vaccine effectiveness against infection are greater in the few months following the booster dose and if more time has elapsed since the prior vaccine dose [89,157,159,162,164,166,199-205]. As an example, 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, vaccine effectiveness of an additional booster dose (with a bivalent vaccine) against symptomatic COVID-19 was greater if more time had elapsed between the last vaccine dose and the additional booster (28 to 31 percent if the interval was two to three months versus 43 to 56 percent if it was more than eight months) [199].

Updating a vaccine to better match the antigenic target with circulating variants is presumed to be associated with a boost in effectiveness against symptomatic infection, although this is likely short-lived as well. (See 'Antibody responses and immune correlates of protection' below.)

Impact on character and duration of breakthrough infection (including risk of long-COVID-19) – Multiple observational studies suggest that breakthrough infection in vaccinated individuals is associated with fewer and shorter duration of symptoms, a lower likelihood of "long COVID-19" (otherwise unexplained symptoms that persist at least two to three months after infection), and a higher likelihood of asymptomatic infection compared with infection in unvaccinated individuals [206-209].

Impact on transmission risk — Widespread vaccination reduces the overall transmission risk since vaccinated individuals are less likely to become infected. Data accumulated prior to the emergence of the Omicron variant also suggested that individuals who developed infection despite vaccination may be less likely to transmit to others [210-215].

Vaccination may also reduce the likelihood of transmission in the setting of Omicron infection [216-218]. In a study of individuals in a state prison system in the United States conducted when BA.1 and BA.2 Omicron subvariants were dominant, the overall secondary attack rate from 1126 index SARS-CoV-2 cases to their cellmates was 30 percent [216]. The risk of transmission from index cases who had been vaccinated was compared with index cases who had neither vaccination nor previous infection (22 percent lower in those with vaccination without prior infection and 40 percent lower in those with both vaccination and prior infection). The risk of transmission from vaccinated individuals was lower following booster vaccination than primary series alone. Prior infection alone was also associated with a lower transmission risk.

Comparative efficacy between vaccine types — Although precise comparative efficacy is uncertain because the different vaccines have not been compared head to head in clinical trials, limited evidence from observational studies suggests that Moderna COVID-19 vaccine may be slightly more effective than Pfizer COVID-19 vaccine [219-226]. However, it is unclear whether the difference is clinically significant. As an example, in a study from the United States that compared over 400,000 veterans who received one of the two mRNA vaccines, Moderna COVID-19 vaccine 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) [221]. Safety of the mRNA vaccines in the same cohort was largely comparable, with only small differences in serious adverse events of uncertain clinical significance [227].

Because it has not been available for as long as the other vaccines, data on Novavax COVID-19 vaccine are more limited, although large, randomized trials support its safety and efficacy.

Antibody responses and 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 [228,229]. Data from these studies can help assess likely efficacy of new vaccines or formulations (eg, with new antigenic targets) 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 the clinical trials. Furthermore, the immune correlates of protection against severe infection have not been fully elucidated. Vaccine- or infection-induced cellular immunity often appears robust against variants that escape antibody binding, and this may contribute to the reduced risk of severe disease from such variants [230-233].

Using COVID-19 vaccines with altered spike targets to more closely match circulating SARS-CoV-2 variants is analogous to updating the seasonal influenza vaccine each year; influenza vaccines are made available globally based on immunogenicity studies prior to repeated clinical evaluation because of extensive experience with the preceding versions.

In human and animal immunogenicity studies, the 2023-2024 formula COVID-19 vaccines that are based on the spike protein of the XBB.1.5 variant elicited robust neutralizing antibody levels against related Omicron variants that are prevalent in the fall of 2023 (eg, EG.5.1, FL.1.5.1, XBB.1.16) as well as the Omicron variant BA.2.86, which had raised concern for having the potential to escape immunity from other variants [234-236].

RISKS OF VACCINATION

Common adverse effects — Local and systemic adverse effects are relatively common with the mRNA vaccines (Pfizer COVID-19 vaccine and Moderna COVID-19 vaccine) and the recombinant Novavax COVID-19 vaccine. Most are of mild or moderate severity (ie, do not prevent daily activities or require pain relievers) and are limited to the first two to three days after vaccination [122,237,238]. Approximately 10 to 20 percent of recipients have adverse effects that limit school attendance or daily life activities [239,240].

Common injection site reactions include mainly pain, as well as redness, swelling, and pruritus. Among the mRNA vaccines, slightly higher rates have been reported for the Moderna COVID-19 vaccine (in 75 to 80 percent) than for the Pfizer COVID-19 vaccine (65 percent) [122].

Similarly, reported rates of nonsevere systemic adverse effects, such as fatigue, headache, and myalgia (50 to 60 versus 40 to 50 percent), and fevers, chills, and joint pain (30 to 40 versus 20 percent) appear slightly higher with Moderna COVID-19 vaccine than Pfizer COVID-19 vaccine [122].

In general, the rates of common reactions are lower among older (eg, >65 years) compared with younger adults and are lower among younger children than adolescents [83,182]. Among young children, irritability, crying, drowsiness, and loss of appetite are common; febrile seizures are rare [107,108].

Review of active and passive surveillance systems in the United States suggests that the safety profile of the bivalent mRNA vaccines was similar to that of the monovalent mRNA vaccines in children and adults [239,240]. Local injection reactions, headache, myalgia, and fever are the most common side effects and are short lived, although they limit school attendance or daily life activities in 10 to 20 percent of recipients.

Rare but serious associated events — COVID-19 vaccines are exceedingly safe. The primary safety concerns are a very rare risk of myocarditis with mRNA vaccines and possibly recombinant protein vaccines and very rare risks of thrombosis with thrombocytopenia and Guillain-Barré syndrome (GBS) with adenoviral vector vaccines (which are not used in the United States). Active and passive surveillance systems in adults and children have failed to find other clear associations with major adverse events [241].

Myocarditis — Myocarditis and pericarditis, mainly in male adolescents and young adults, have been reported more frequently than expected following receipt of the mRNA vaccines, Pfizer COVID-19 vaccine and Moderna COVID-19 vaccine [242,243]. Cases were also noted in Novavax COVID-19 vaccine recipients during the phase III trials [244], and surveillance suggested a possible increased risk following receipt of Janssen/Johnson & Johnson COVID-19 vaccine, which is no longer used in the United States [16]. Given the infrequency and the typically mild nature of the myocarditis and pericarditis cases, the benefits of vaccination greatly exceed the small increased risk [243].

For those who develop myocarditis or pericarditis following an mRNA vaccine or Novavax COVID-19 vaccine, 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 [26]. 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 [245]. 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 Pfizer COVID-19 vaccine

16 to 17 years old – 105.9 cases per million doses of Pfizer COVID-19 vaccine

18 to 24 years old – 52.4 to 56.3 cases per million doses Pfizer COVID-19 vaccine and Moderna COVID-19 vaccine, 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.

Estimated rates of myocarditis from the Vaccine Safety Datalink (VSD), an active surveillance system in the United States, which recorded a total of 320 cases of myocarditis following seven million vaccine doses, were somewhat higher, possibly because the system does not rely on the patients or providers to make special efforts to report cases [246]. Among males, the estimated rates of myocarditis within the week following a second vaccine dose were 150.5 cases (ages 12 to 15 years), 127.1 (ages 16 to 17 years), and 81.4 (ages 18 to 29 years) per million doses of BNT162b2 and 97.0 (ages 18 to 29 years) per million doses of mRNA-1273.

Studies from other countries have also suggested an increased rate of myocarditis following Pfizer COVID-19 vaccine receipt compared with the expected background rate [100,247-251]. Observational data also suggest that the risk may be higher with Moderna COVID-19 vaccine than Pfizer COVID-19 vaccine [252-255]. The risk also appears to be higher following the second dose and with shorter intervals between doses (eg, less than 30 versus more than 60 days) [256].

For all age groups, the risk of myocarditis or pericarditis following mRNA vaccination is estimated in some, but not all [254], studies to be less than the risk associated with SARS-CoV-2 infection [257].

Among the cases that have been reported, most are mild [243,247,248,258,259]. 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 persistent or fulminant myocarditis in individuals who had received an mRNA vaccine within the preceding weeks [260,261]. Although a study from Korea reported that 20 percent of the 480 cases of vaccine-associated myocarditis identified nationwide were severe, with 36 fulminant cases and 21 deaths, that report is an outlier compared with those from other countries [262].

The typical 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 [263]. Almost all presented with chest pain, with symptom onset a median of two days after vaccine receipt, which was often receipt of the second vaccine dose. Electrocardiogram was abnormal in 70 percent (ST-segment elevations or T-wave abnormalities), cardiac magnetic resonance imaging (MRI) 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 (ICU), 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".)

Events associated with vaccines not used in the United States

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) [264-267]. Some experts refer to this syndrome as vaccine-associated immune thrombotic thrombocytopenia (VITT); others have used the term thrombosis with thrombocytopenia syndrome (TTS). Neither of these vaccines is used in the United States.

In reported cases, thrombosis often occurred at unusual sites, including the cerebral venous sinuses and mesenteric vessels, and at more than one site [268-270]. 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 [271]. 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 [272-274]. 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 [275]. 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) [274].

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 [276,277]. For ChadOx1 nCoV-19/AZD1222, studies have reported conflicting findings regarding this risk.

Guillain-Barré syndrome — The adenovirus vector vaccines (Ad26.COV2.S [Janssen/Johnson & Johnson COVID-19 vaccine] and ChAdOx1 nCoV-19/AZD1222 [AstraZeneca COVID-19 vaccine]) have been associated with GBS. 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 [278,279]. Cases of GBS, including recurrent cases, have also been reported in the setting of SARS-CoV-2 infection [280,281], and observational data suggest the risk of GBS after infection exceeds the risk after vaccination [282]. Pending additional data, for individuals with a documented history of GBS, we suggest using COVID-19 vaccines other than adenovirus vector vaccines (adenovirus vector vaccines are not available in the United States). 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'.)

Ad26.COV2.S – When Ad26.COV2.S was available in the United States, studies of different surveillance systems suggested a higher incidence of GBS following that vaccine compared with others [283-285]. In an evaluation of almost 500 million COVID-19 vaccine doses administered in the United States, 295 verified cases of GBS were reported to a passive surveillance system [285]. The rate of GBS reported within 212 days after vaccination was higher for Ad26.COV2.S than for BNT162b2 or mRNA-1273 (3.29 cases compared with 0.29 and 0.35 cases per million doses). The number of cases observed after Ad26.COV2.S was 3.8 times the expected rate in the general population. The median age was 59 years (interquartile range [IQR] 46 to 68), and the median time from Ad26.COV2.S receipt and GBS was 10 days (IQR 5 to 17). Two deaths occurred in individuals with GBS reported after Ad26.COV2.S. In an earlier report of 100 cases, a quarter of the patients reported bilateral facial weakness [286]. This vaccine is no longer available in the United States.

ChAdOx1 nCoV-19/AZD1222 – Reviews of surveillance systems in Europe and the United Kingdom have suggested an excess number of GBS cases following ChAdOx1 nCoV-19/AZD1222 vaccination (an excess of 0.58 cases per 100,000 doses according to one analysis) [279,287]. Other scattered reports have also described GBS, including variant GBS with bilateral facial weakness, following ChAdOx1 nCoV-19/AZD1222 vaccination [288,289]. However, in a cohort study including over four million individuals who received ChAdOx1 nCoV-19/AZD1222, the rate of GBS following the vaccine dose was not higher than the expected prepandemic background rate [290].

Other reactions (without established association) — Although myriad adverse events have been reported in individuals following vaccine administration, no other severe events than those listed above have been clearly associated with vaccination after hundreds of millions of doses administered.

No clear signal of ischemic stroke with the bivalent mRNA vaccines – One active surveillance system (the VSD) reported a slightly higher-than-expected number of ischemic strokes in individuals ≥65 years of age following receipt of the bivalent Pfizer-BioNTech COVID-19 vaccine; however, the rate excess decreased with time and there was no increased incidence of other cardiovascular events [291,292]. Furthermore, additional analyses did not identify a similar signal in several other surveillance systems or with the bivalent Moderna COVID-19 vaccine [291,293-296].

Possible anaphylaxis – Anaphylaxis has been reported following administration of both mRNA COVID-19 vaccines [297]. 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 [237,298,299]; rates ranging from 2.5 to 7.9 events per million doses have been reported, depending on the surveillance system [300-302]. The vast majority of these events occurred in individuals with a history of prior allergic reactions and occurred within 30 minutes. However, many of those who had a reaction after the first vaccination did not suffer a similar reaction after the second dose. Data suggest that the majority are not IgE-mediated reactions [303] and complement-mediated mechanisms have not been documented. Some cases have been reported as stress-related events [304,305]. Possible anaphylaxis and other reported allergic reactions (eg, pruritus, rash, scratchy sensations in the throat, and mild respiratory symptoms) are discussed elsewhere [306]. (See "COVID-19: Allergic reactions to SARS-CoV-2 vaccines", section on 'mRNA vaccines'.)

No clear impact on fertility – As an example, many patients of child-bearing potential are concerned that COVID-19 vaccination could adversely impact fertility. However, in epidemiologic studies, there is no association between COVID-19 vaccination and fertility problems in either females or males [307,308].

Other major adverse events have also not been consistently associated with mRNA or recombinant protein vaccines. Rare cases of Bell palsy were noted in the trials for both Pfizer COVID-19 vaccine and Moderna COVID-19 vaccine, although 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 palsy [298,300]. No other major vaccine-associated adverse events have been clearly identified in post-vaccine surveillance [300].

Ongoing safety assessment and reporting of adverse events — Adequately assessing vaccine safety is critical to the success of immunization programs. Although existing comprehensive systems to monitor vaccine safety are in place, they were 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) [309]. These include:

Vaccine Adverse Event Reporting System – this is 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. 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 VAERS.

Vaccine Safety Datalink – this 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. Analysis of the VSD can help determine if adverse events are causally or only coincidentally related to vaccination by comparing the incidence of given clinical syndrome in vaccinees at time points soon after vaccination with time points before or well after vaccination [300].

Clinical Immunization Safety Assessment project (CISA) – this 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 [310,311]. These systems have been coordinated through the CDC and have enlisted multiple other health care groups to provide ongoing data on vaccine safety. These systems and information sources add an additional layer of safety monitoring [312,313].

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 FDA – 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") [314]. Therefore, if someone is injured by vaccine, society owes that person compensation. This is the basis for the National Vaccine Injury Compensation Program (NVICP) [315]. 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 [316].

COMBATING VACCINE HESITANCY — Vaccine hesitancy presents a major obstacle to achieving broad vaccination coverage. 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 [317]. 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: Caregiver 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 [318,319].

Willingness to accept a COVID-19 vaccine has varied by country [320,321]. In the United States, rates of vaccine hesitancy have decreased over the course of the pandemic but remain substantial [322-325]. 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 [322,326-328]. In a CDC survey, the main reasons for reporting nonintent to receive vaccine were concerns about vaccine side effects and safety and lack of trust in the process [326]. Vaccination rates in the United States increase with age; rates among children have been especially low. As of April 2023, fewer than 35 percent of children 5 to 11 years had received a primary vaccine series and fewer than 5 percent had received a bivalent vaccine dose; rates among younger children were much lower [329].

Among individuals who have received a primary COVID-19 vaccine series, major reasons for not receiving a booster dose are lack of awareness about eligibility or availability, belief that they remain protected against severe infection without it, and uncertainty about the vaccine safety and efficacy [330]. These findings highlight the role for clinicians in educating patients on emerging vaccine data and recommendations.

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). Over time, the mRNA vaccines have been reformulated to target the mutations in the spike proteins of various SARS-CoV-2 variants. As of September 2023, COVID-19 vaccines are only available as updated 2023-2024 formulations (Moderna COVID-19 vaccine 2023-2024 Formula, Pfizer COVID-19 vaccine 2023-2024 Formula, and Novavax COVID-19 vaccine 2023-2024 Formula), which contain the spike protein from Omicron variant XBB.1.5. (See 'General principles' above and 'Available vaccines' above.)

Indications for vaccination – For all individuals aged six months and older, we suggest a 2023-2024 formula COVID-19 vaccine (Grade 2C). In particular, individuals who are at highest risk of severe outcomes with COVID-19, specifically individuals 65 years or older, immunocompromised individuals, and individuals with multiple medical comorbidities (table 3) are most likely to benefit from vaccination.

Vaccines available in the United States 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, even in the context of variants that partially evade vaccine-induced immune responses. Although effectiveness wanes, vaccine-induced immunity continues to reduce the risk of severe disease, and repeat vaccination is associated with a relative increase in effectiveness over several months. (See 'Indications and vaccine selection' above and 'Benefits of vaccination' above.)

How to vaccinate – Our approach to COVID-19 vaccination in the United States is in accordance with recommendations from the Centers for Disease Control and Prevention (CDC).

Immunocompetent individuals aged six months to four years – Individuals in this age group should receive at least three vaccine doses, at least one of which is a 2023-2024 formula COVID-19 vaccine. The number of doses of the 2023-2024 formula COVID-19 vaccine depends on their vaccination history. (See 'Children aged six months to four years' above.)

Immunocompetent individuals aged five years and older – Individuals in this age group should receive one 2023-2024 formula COVID-19 vaccine. This includes individuals who have already received prior vaccine doses. (See 'Children aged 5 to 11 years' above and 'Adolescents and adults aged 12 years and older' above.)

Individuals aged six months and older with immunocompromising conditions – Individuals with immunocompromising conditions (table 4) should receive at least three mRNA vaccine doses or at least two Novavax vaccine doses, at least one of which is a 2023-2024 formula COVID-19 vaccine. The number of doses of the 2023-2024 formula COVID-19 vaccine depends on their vaccination history. (See 'Immunocompromised individuals' above.)

Vaccine doses are presented in the table (table 2).

Vaccine safety – Severe adverse events with available COVID-19 vaccines are extremely rare and do not outweigh their benefit for recommended indications. The mRNA vaccines and Novavax COVID-19 vaccine have all been associated with a small excess risk of myocarditis, mainly in male adolescents and young adults. The majority of vaccine-associated myocarditis cases are mild and improve within days. (See 'Myocarditis' above.)

Deviations from dosing recommendations – If the vaccine is administered in a manner different from the recommended approach, the dose generally does not have to be repeated. CDC recommendations on how to manage vaccination errors or deviations are presented in the table (table 5). (See 'Technique and potential administration errors' 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 and 'Common adverse effects' 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. Individuals without a contraindication who have a history of anaphylaxis to other vaccines or injectable therapies, an allergy-related contraindication to a COVID-19 vaccine class other than the one they are receiving, or a nonsevere immediate allergic reaction to a prior COVID-19 vaccine 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. SPIKEVAX (COVID-19 Vaccine, mRNA) Suspension for injection, for intramuscular use; 2023-2024 Formula. US Food and Drug Administration (FDA) approved product information. Revised September 11, 2023. https://www.fda.gov/media/155675/download?attachment (Accessed on September 11, 2023).
  11. FACT SHEET FOR HEALTHCARE PROVIDERS ADMINISTERING VACCINE: EMERGENCY USE AUTHORIZATION OF MODERNA COVID-19 VACCINE (2023-2024 FORMULA), FOR INDIVIDUALS 6 MONTHS THROUGH 11 YEARS OF AGE https://www.fda.gov/media/167208/download?attachment (Accessed on September 11, 2023).
  12. COMIRNATY (COVID-19 Vaccine, mRNA) suspension for injection, for intramuscular use; 2023-2024 Formula. US Food and Drug Administration (FDA) approved product information. Revised September 11, 2023. https://www.fda.gov/media/151707/download?attachment (Accessed on September 11, 2023).
  13. FACT SHEET FOR HEALTHCARE PROVIDERS ADMINISTERING VACCINE: EMERGENCY USE AUTHORIZATION OF PFIZER-BIONTECH COVID-19 VACCINE (2023-2024 FORMULA), FOR 6 MONTHS THROUGH 11 YEARS OF AGE https://www.fda.gov/media/167211/download?attachment (Accessed on September 11, 2023).
  14. Recommendation for the 2023-2024 Formula of COVID-19 vaccines in the US. https://www.fda.gov/media/169591/download?attachment (Accessed on September 11, 2023).
  15. Emergency Use Authorization (EUA) of the Novavax COVID-19 vaccine, adjuvanted (2023-2024 Formula), for individuals 12 years of age and older https://www.fda.gov/media/159897/download?attachment (Accessed on October 04, 2023).
  16. 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 March 27, 2023).
  17. 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.
  18. 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.
  19. 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.
  20. 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.
  21. 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.
  22. 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.
  23. 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.
  24. 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.
  25. CDC. COVID-19–Associated Hospitalizations among Infants, Children and Adults — COVID-NET, January– August 2023. Presented September 12, 2023. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2023-09-12/03-COVID-Havers-508.pdf (Accessed on September 12, 2023).
  26. 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 April 26, 2023).
  27. Oliver S. Updates to COVID-19 vaccine policy: Considerations for future planning. Presented at the ACIP meeting on April 19, 2023. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2023-04-19/06-COVID-Oliver-508.pdf (Accessed on April 20, 2023).
  28. Jones JM, Manrique IM, Stone MS, et al. Estimates of SARS-CoV-2 Seroprevalence and Incidence of Primary SARS-CoV-2 Infections Among Blood Donors, by COVID-19 Vaccination Status - United States, April 2021-September 2022. MMWR Morb Mortal Wkly Rep 2023; 72:601.
  29. Klaassen F, Chitwood MH, Cohen T, et al. Changes in Population Immunity Against Infection and Severe Disease From Severe Acute Respiratory Syndrome Coronavirus 2 Omicron Variants in the United States Between December 2021 and November 2022. Clin Infect Dis 2023; 77:355.
  30. Taylor CA, Patel K, Patton ME, et al. COVID-19-Associated Hospitalizations Among U.S. Adults Aged ≥65 Years - COVID-NET, 13 States, January-August 2023. MMWR Morb Mortal Wkly Rep 2023; 72:1089.
  31. Atmar RL, Lyke KE, Deming ME, et al. Homologous and Heterologous Covid-19 Booster Vaccinations. N Engl J Med 2022; 386:1046.
  32. 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.
  33. 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.
  34. 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.
  35. 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.
  36. 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.
  37. 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.
  38. Elias MD, Truong DT, Oster ME, et al. Examination of Adverse Reactions After COVID-19 Vaccination Among Patients With a History of Multisystem Inflammatory Syndrome in Children. JAMA Netw Open 2023; 6:e2248987.
  39. 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.
  40. 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; 372:1418.
  41. Stamatatos L, Czartoski J, Wan YH, et al. mRNA vaccination boosts cross-variant neutralizing antibodies elicited by SARS-CoV-2 infection. Science 2021; 372:1413.
  42. 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.
  43. 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.
  44. 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.
  45. 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.
  46. 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.
  47. 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.
  48. 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.
  49. 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.
  50. 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.
  51. 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.
  52. 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.
  53. 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.
  54. 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).
  55. 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.
  56. 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.
  57. 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).
  58. General Principles of COVID-19 Vaccines for Immunocompromised Patients https://www.hematology.org/covid-19/ash-astct-covid-19-and-vaccines (Accessed on February 21, 2022).
  59. 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.
  60. 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.
  61. 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.
  62. 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.
  63. 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.
  64. 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.
  65. 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.
  66. 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.
  67. 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.
  68. 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.
  69. Redjoul R, Le Bouter A, Beckerich F, et al. Antibody response after second BNT162b2 dose in allogeneic HSCT recipients. Lancet 2021; 398:298.
  70. 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.
  71. 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.
  72. Bitoun S, Henry J, Desjardins D, et al. Rituximab Impairs B Cell Response But Not T Cell Response to COVID-19 Vaccine in Autoimmune Diseases. Arthritis Rheumatol 2022; 74:927.
  73. 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.
  74. 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.
  75. 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.
  76. 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.
  77. 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.
  78. 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.
  79. 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.
  80. 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.
  81. American College of Rheumatology. COVID-19 Vaccine Clinical Guidance Summary for Patients with Rheumatic and Musculoskeletal Diseases. Available at: https://www.rheumatology.org/Portals/0/Files/COVID-19-Vaccine-Clinical-Guidance-Rheumatic-Diseases-Summary.pdf (Accessed on March 04, 2021).
  82. Barnes E, Goodyear CS, Willicombe M, et al. SARS-CoV-2-specific immune responses and clinical outcomes after COVID-19 vaccination in patients with immune-suppressive disease. Nat Med 2023; 29:1760.
  83. 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.
  84. Muñoz FM, Sher LD, Sabharwal C, et al. Evaluation of BNT162b2 Covid-19 Vaccine in Children Younger than 5 Years of Age. N Engl J Med 2023; 388:621.
  85. 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.
  86. 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.
  87. 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.
  88. Walter EB, Talaat KR, Sabharwal C, et al. Evaluation of the BNT162b2 Covid-19 Vaccine in Children 5 to 11 Years of Age. N Engl J Med 2022; 386:35.
  89. 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.
  90. 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.
  91. 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.
  92. 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.
  93. 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.
  94. 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.
  95. 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.
  96. 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.
  97. 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.
  98. 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.
  99. Jang EJ, Choe YJ, Kim RK, Park YJ. BNT162b2 Vaccine Effectiveness Against the SARS-CoV-2 Omicron Variant in Children Aged 5 to 11 Years. JAMA Pediatr 2023; 177:319.
  100. Watanabe A, Kani R, Iwagami M, et al. Assessment of Efficacy and Safety of mRNA COVID-19 Vaccines in Children Aged 5 to 11 Years: A Systematic Review and Meta-analysis. JAMA Pediatr 2023; 177:384.
  101. Piechotta V, Siemens W, Thielemann I, et al. Safety and effectiveness of vaccines against COVID-19 in children aged 5-11 years: a systematic review and meta-analysis. Lancet Child Adolesc Health 2023; 7:379.
  102. Lin DY, Xu Y, Gu Y, et al. Effects of COVID-19 vaccination and previous SARS-CoV-2 infection on omicron infection and severe outcomes in children under 12 years of age in the USA: an observational cohort study. Lancet Infect Dis 2023; 23:1257.
  103. Tartof SY, Frankland TB, Slezak JM, et al. Receipt of BNT162b2 Vaccine and COVID-19 Ambulatory Visits in US Children Younger Than 5 Years. JAMA 2023; 330:1282.
  104. 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.
  105. Wallace M. Evidence to Recommendations Framework: 2023 – 2024 (Monovalent, XBB Containing) COVID-19 Vaccine . Presented at ACIP meeting September 12, 2023. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2023-09-12/11-COVID-Wallace-508.pdf (Accessed on September 12, 2023).
  106. 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.
  107. 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).
  108. 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).
  109. 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.
  110. 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).
  111. 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).
  112. 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).
  113. 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.
  114. Yousaf AR, Cortese MM, Taylor AW, et al. Reported cases of multisystem inflammatory syndrome in children aged 12-20 years in the USA who received a COVID-19 vaccine, December, 2020, through August, 2021: a surveillance investigation. Lancet Child Adolesc Health 2022; 6:303.
  115. Cortese MM, Taylor AW, Akinbami LJ, et al. Surveillance for Multisystem Inflammatory Syndrome in US Children Aged 5-11 Years Who Received Pfizer-BioNTech COVID-19 Vaccine, November 2021 through March 2022. J Infect Dis 2023; 228:143.
  116. 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.
  117. 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.
  118. Centers for Disease Control and Prevention. Vaccines for Children Program (VFC). https://www.cdc.gov/vaccines/programs/vfc/ (Accessed on November 05, 2020).
  119. 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.
  120. 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.
  121. 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.
  122. Chapin-Bardales J, Gee J, Myers T. Reactogenicity Following Receipt of mRNA-Based COVID-19 Vaccines. JAMA 2021; 325:2201.
  123. 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.
  124. 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.
  125. Centers for Disease Control and Prevention (CDC). Syncope after vaccination--United States, January 2005-July 2007. MMWR Morb Mortal Wkly Rep 2008; 57:457.
  126. 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.
  127. 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.
  128. 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).
  129. 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.
  130. Blumenthal KG, Saff RR, Freeman EE. Delayed Large Local Reactions to mRNA Vaccines. Reply. N Engl J Med 2021; 384:e98.
  131. 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.
  132. Bohnert AS, Kumbier K, Rowneki M, et al. Adverse outcomes of SARS-CoV-2 infection with delta and omicron variants in vaccinated versus unvaccinated US veterans: retrospective cohort study. BMJ 2023; 381:e074521.
  133. 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).
  134. 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.
  135. 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.
  136. 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.
  137. 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.
  138. Johnson AG, Linde L, Payne AB, et al. Notes from the Field: Comparison of COVID-19 Mortality Rates Among Adults Aged ≥65 Years Who Were Unvaccinated and Those Who Received a Bivalent Booster Dose Within the Preceding 6 Months - 20 U.S. Jurisdictions, September 18, 2022-April 1, 2023. MMWR Morb Mortal Wkly Rep 2023; 72:667.
  139. 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.
  140. 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.
  141. Bergwerk M, Gonen T, Lustig Y, et al. Covid-19 Breakthrough Infections in Vaccinated Health Care Workers. N Engl J Med 2021; 385:1474.
  142. Tenforde MW, Self WH, Adams K, et al. Association Between mRNA Vaccination and COVID-19 Hospitalization and Disease Severity. JAMA 2021; 326:2043.
  143. 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.
  144. 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.
  145. Jiang J, Chan L, Kauffman J, et al. Impact of Vaccination on Major Adverse Cardiovascular Events in Patients With COVID-19 Infection. J Am Coll Cardiol 2023; 81:928.
  146. 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.
  147. 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.
  148. Goldberg Y, Mandel M, Bar-On YM, et al. Waning Immunity after the BNT162b2 Vaccine in Israel. N Engl J Med 2021; 385:e85.
  149. 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.
  150. 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.
  151. 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.
  152. DeCuir J, Surie D, Zhu Y, et al. Effectiveness of Monovalent mRNA COVID-19 Vaccination in Preventing COVID-19-Associated Invasive Mechanical Ventilation and Death Among Immunocompetent Adults During the Omicron Variant Period - IVY Network, 19 U.S. States, February 1, 2022-January 31, 2023. MMWR Morb Mortal Wkly Rep 2023; 72:463.
  153. Kirsebom FCM, Andrews N, Stowe J, et al. Duration of protection of ancestral-strain monovalent vaccines and effectiveness of bivalent BA.1 boosters against COVID-19 hospitalisation in England: a test-negative case-control study. Lancet Infect Dis 2023; 23:1235.
  154. Surie D, DeCuir J, Zhu Y, et al. Early Estimates of Bivalent mRNA Vaccine Effectiveness in Preventing COVID-19-Associated Hospitalization Among Immunocompetent Adults Aged ≥65 Years - IVY Network, 18 States, September 8-November 30, 2022. MMWR Morb Mortal Wkly Rep 2022; 71:1625.
  155. Tenforde MW, Weber ZA, Natarajan K, et al. Early Estimates of Bivalent mRNA Vaccine Effectiveness in Preventing COVID-19-Associated Emergency Department or Urgent Care Encounters and Hospitalizations Among Immunocompetent Adults - VISION Network, Nine States, September-November 2022. MMWR Morb Mortal Wkly Rep 2022; 71:1616.
  156. Lin DY, Xu Y, Gu Y, et al. Effectiveness of Bivalent Boosters against Severe Omicron Infection. N Engl J Med 2023; 388:764.
  157. Johnson AG, Linde L, Ali AR, et al. COVID-19 Incidence and Mortality Among Unvaccinated and Vaccinated Persons Aged ≥12 Years by Receipt of Bivalent Booster Doses and Time Since Vaccination - 24 U.S. Jurisdictions, October 3, 2021-December 24, 2022. MMWR Morb Mortal Wkly Rep 2023; 72:145.
  158. Arbel R, Peretz A, Sergienko R, et al. Effectiveness of a bivalent mRNA vaccine booster dose to prevent severe COVID-19 outcomes: a retrospective cohort study. Lancet Infect Dis 2023; 23:914.
  159. 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.
  160. 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.
  161. Arbel R, Hammerman A, Sergienko R, et al. BNT162b2 Vaccine Booster and Mortality Due to Covid-19. N Engl J Med 2021; 385:2413.
  162. Ioannou GN, Bohnert ASB, O'Hare AM, et al. Effectiveness of mRNA COVID-19 Vaccine Boosters Against Infection, Hospitalization, and Death: A Target Trial Emulation in the Omicron (B.1.1.529) Variant Era. Ann Intern Med 2022; 175:1693.
  163. Lai FTT, Yan VKC, Ye X, et al. Booster vaccination with inactivated whole-virus or mRNA vaccines and COVID-19-related deaths among people with multimorbidity: a cohort study. CMAJ 2023; 195:E143.
  164. 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.
  165. 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.
  166. 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.
  167. Wee LE, Pang D, Chiew C, et al. Long-term Real-world Protection Afforded by Third mRNA Doses Against Symptomatic Severe Acute Respiratory Syndrome Coronavirus 2 Infections, Coronavirus Disease 19-related Emergency Attendances and Hospitalizations Amongst Older Singaporeans During an Omicron XBB Wave. Clin Infect Dis 2023; 77:1111.
  168. 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.
  169. 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.
  170. 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.
  171. 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.
  172. 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.
  173. 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.
  174. 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.
  175. 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).
  176. 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.
  177. 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.
  178. Wu N, Joyal-Desmarais K, Ribeiro PAB, et al. Long-term effectiveness of COVID-19 vaccines against infections, hospitalisations, and mortality in adults: findings from a rapid living systematic evidence synthesis and meta-analysis up to December, 2022. Lancet Respir Med 2023; 11:439.
  179. Link-Gelles R, Weber ZA, Reese SE, et al. Estimates of Bivalent mRNA Vaccine Durability in Preventing COVID-19-Associated Hospitalization and Critical Illness Among Adults with and Without Immunocompromising Conditions - VISION Network, September 2022-April 2023. MMWR Morb Mortal Wkly Rep 2023; 72:579.
  180. 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).
  181. 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.
  182. 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).
  183. 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.
  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. Áñez G, Dunkle LM, Gay CL, et al. Safety, Immunogenicity, and Efficacy of the NVX-CoV2373 COVID-19 Vaccine in Adolescents: A Randomized Clinical Trial. JAMA Netw Open 2023; 6:e239135.
  186. 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.
  187. 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.
  188. 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.
  189. 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.
  190. 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.
  191. 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.
  192. Cohn BA, Cirillo PM, Murphy CC, et al. SARS-CoV-2 vaccine protection and deaths among US veterans during 2021. Science 2022; 375:331.
  193. 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.
  194. 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.
  195. 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.
  196. 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.
  197. Menegale F, Manica M, Zardini A, et al. Evaluation of Waning of SARS-CoV-2 Vaccine-Induced Immunity: A Systematic Review and Meta-analysis. JAMA Netw Open 2023; 6:e2310650.
  198. Mateo-Urdiales A, Sacco C, Petrone D, et al. Estimated Effectiveness of a Primary Cycle of Protein Recombinant Vaccine NVX-CoV2373 Against COVID-19. JAMA Netw Open 2023; 6:e2336854.
  199. 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.
  200. Lin DY, Xu Y, Gu Y, et al. Durability of Bivalent Boosters against Omicron Subvariants. N Engl J Med 2023; 388:1818.
  201. 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.
  202. 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.
  203. 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.
  204. Amir O, Goldberg Y, Mandel M, et al. Initial protection against SARS-CoV-2 omicron lineage infection in children and adolescents by BNT162b2 in Israel: an observational study. Lancet Infect Dis 2023; 23:67.
  205. Arashiro T, Arima Y, Kuramochi J, et al. Effectiveness of BA.1- and BA.4/BA. 5-Containing Bivalent COVID-19 mRNA Vaccines Against Symptomatic SARS-CoV-2 Infection During the BA.5-Dominant Period in Japan. Open Forum Infect Dis 2023; 10:ofad240.
  206. 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.
  207. Al-Aly Z, Bowe B, Xie Y. Long COVID after breakthrough SARS-CoV-2 infection. Nat Med 2022; 28:1461.
  208. 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.
  209. Byambasuren O, Stehlik P, Clark J, et al. Effect of covid-19 vaccination on long covid: systematic review. BMJ Med 2023; 2:e000385.
  210. 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.
  211. 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.
  212. 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.
  213. 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.
  214. 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.
  215. 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.
  216. Tan ST, Kwan AT, Rodríguez-Barraquer I, et al. Infectiousness of SARS-CoV-2 breakthrough infections and reinfections during the Omicron wave. Nat Med 2023; 29:358.
  217. Hoeve CE, de Gier B, Huiberts AJ, et al. Vaccine Effectiveness Against Severe Acute Respiratory Syndrome Coronavirus 2 Delta and Omicron Infection and Infectiousness Within Households in the Netherlands Between July 2021 and August 2022. J Infect Dis 2023; 228:431.
  218. Mongin D, Bürgisser N, Laurie G, et al. Effect of SARS-CoV-2 prior infection and mRNA vaccination on contagiousness and susceptibility to infection. Nat Commun 2023; 14:5452.
  219. 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.
  220. 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.
  221. 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.
  222. 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.
  223. 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.
  224. 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.
  225. 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.
  226. Hulme WJ, Horne EMF, Parker EPK, et al. Comparative effectiveness of BNT162b2 versus mRNA-1273 covid-19 vaccine boosting in England: matched cohort study in OpenSAFELY-TPP. BMJ 2023; 380:e072808.
  227. 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.
  228. Feng S, Phillips DJ, White T, et al. Correlates of protection against symptomatic and asymptomatic SARS-CoV-2 infection. Nat Med 2021; 27:2032.
  229. 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.
  230. 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.
  231. Liu J, Chandrashekar A, Sellers D, et al. Vaccines elicit highly conserved cellular immunity to SARS-CoV-2 Omicron. Nature 2022; 603:493.
  232. Keeton R, Tincho MB, Ngomti A, et al. T cell responses to SARS-CoV-2 spike cross-recognize Omicron. Nature 2022; 603:488.
  233. 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.
  234. Safety and Immunogenicity of Moderna COVID-19 Vaccine (2023-2024 Formula) Monovalent XBB.1.5 Variant Vaccine. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2023-09-12/08-COVID-Priddy-508.pdf (Accessed on September 12, 2023).
  235. Monovalent XBB.1.5 BNT162b2 COVID-19 Vaccine. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2023-09-12/10-COVID-Modjarrad-508.pdf (Accessed on September 12, 2023).
  236. Data in Support of Novavax XBB.1.5 Vaccine https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2023-09-12/09-COVID-Dubovsky-508.pdf (Accessed on September 12, 2023).
  237. 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.
  238. FDA Briefing Document. Moderna COVID-19 Vaccine. https://www.fda.gov/media/144434/download (Accessed on December 16, 2020).
  239. Hause AM, Marquez P, Zhang B, et al. Safety Monitoring of Bivalent COVID-19 mRNA Vaccine Booster Doses Among Children Aged 5-11 Years - United States, October 12-January 1, 2023. MMWR Morb Mortal Wkly Rep 2023; 72:39.
  240. Hause AM, Marquez P, Zhang B, et al. Safety Monitoring of Bivalent COVID-19 mRNA Vaccine Booster Doses Among Persons Aged ≥12 Years - United States, August 31-October 23, 2022. MMWR Morb Mortal Wkly Rep 2022; 71:1401.
  241. Goddard K, Donahue JG, Lewis N, et al. Safety of COVID-19 mRNA Vaccination Among Young Children in the Vaccine Safety Datalink. Pediatrics 2023; 152.
  242. Meeting highlights from the Pharmacovigilance Risk Assessment Committee (PRAC) 3-6 May 2021 https://www.ema.europa.eu/en/news/meeting-highlights-pharmacovigilance-risk-assessment-committee-prac-3-6-may-2021 (Accessed on May 26, 2021).
  243. 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.
  244. 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).
  245. 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.
  246. Goddard K, Hanson KE, Lewis N, et al. Incidence of Myocarditis/Pericarditis Following mRNA COVID-19 Vaccination Among Children and Younger Adults in the United States. Ann Intern Med 2022; 175:1169.
  247. 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.
  248. 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.
  249. 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.
  250. Mevorach D, Anis E, Cedar N, et al. Myocarditis after BNT162b2 Vaccination in Israeli Adolescents. N Engl J Med 2022; 386:998.
  251. Buchan SA, Alley S, Seo CY, et al. Myocarditis or Pericarditis Events After BNT162b2 Vaccination in Individuals Aged 12 to 17 Years in Ontario, Canada. JAMA Pediatr 2023; 177:410.
  252. Husby A, Hansen JV, Fosbøl E, et al. SARS-CoV-2 vaccination and myocarditis or myopericarditis: population based cohort study. BMJ 2021; 375:e068665.
  253. Myocarditis and Pericarditis after COVID-19 mRNA Vaccines https://www.publichealthontario.ca/-/media/documents/ncov/vaccines/2021/11/myocarditis-pericarditis-mrna-vaccines.pdf?sc_lang=en (Accessed on January 28, 2022).
  254. 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.
  255. 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.
  256. 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.
  257. 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.
  258. Witberg G, Magen O, Hoss S, et al. Myocarditis after BNT162b2 Vaccination in Israeli Adolescents. N Engl J Med 2022; 387:1816.
  259. Husby A, Gulseth HL, Hovi P, et al. Clinical outcomes of myocarditis after SARS-CoV-2 mRNA vaccination in four Nordic countries: population based cohort study. BMJ Med 2023; 2:e000373.
  260. Verma AK, Lavine KJ, Lin CY. Myocarditis after Covid-19 mRNA Vaccination. N Engl J Med 2021; 385:1332.
  261. Wu KY, Butler CR, Koshman S, et al. Persistent myopericarditis after heterologous SARS-CoV-2 mRNA vaccination. CMAJ 2023; 195:E584.
  262. Cho JY, Kim KH, Lee N, et al. COVID-19 vaccination-related myocarditis: a Korean nationwide study. Eur Heart J 2023; 44:2234.
  263. 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.
  264. 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).
  265. 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).
  266. Schultz NH, Sørvoll IH, Michelsen AE, et al. Thrombosis and Thrombocytopenia after ChAdOx1 nCoV-19 Vaccination. N Engl J Med 2021; 384:2124.
  267. Greinacher A, Thiele T, Warkentin TE, et al. Thrombotic Thrombocytopenia after ChAdOx1 nCov-19 Vaccination. N Engl J Med 2021; 384:2092.
  268. Muir KL, Kallam A, Koepsell SA, Gundabolu K. Thrombotic Thrombocytopenia after Ad26.COV2.S Vaccination. N Engl J Med 2021; 384:1964.
  269. 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.
  270. Pavord S, Scully M, Hunt BJ, et al. Clinical Features of Vaccine-Induced Immune Thrombocytopenia and Thrombosis. N Engl J Med 2021; 385:1680.
  271. 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).
  272. 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.
  273. 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).
  274. 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).
  275. 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.
  276. 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).
  277. 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).
  278. FDA NEWS RELEASE. Coronavirus (COVID-19) Update: July 13, 2021 fda.gov/news-events/press-announcements/coronavirus-covid-19-update-july-13-2021 (Accessed on July 15, 2021).
  279. COVID-19 vaccine safety update. COVID-19 VACCINE JANSSEN 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).
  280. 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.
  281. McDonnell EP, Altomare NJ, Parekh YH, et al. COVID-19 as a Trigger of Recurrent Guillain-Barré Syndrome. Pathogens 2020; 9.
  282. 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.
  283. 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.
  284. 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.
  285. Abara WE, Gee J, Marquez P, et al. Reports of Guillain-Barré Syndrome After COVID-19 Vaccination in the United States. JAMA Netw Open 2023; 6:e2253845.
  286. 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.
  287. Keh RYS, Scanlon S, Datta-Nemdharry P, et al. COVID-19 vaccination and Guillain-Barré syndrome: analyses using the National Immunoglobulin Database. Brain 2023; 146:739.
  288. Allen CM, Ramsamy S, Tarr AW, et al. Guillain-Barré Syndrome Variant Occurring after SARS-CoV-2 Vaccination. Ann Neurol 2021; 90:315.
  289. Maramattom BV, Krishnan P, Paul R, et al. Guillain-Barré Syndrome following ChAdOx1-S/nCoV-19 Vaccine. Ann Neurol 2021; 90:312.
  290. 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.
  291. CDC and FDA identify preliminary COVID-19 vaccine safety signal for persons aged 65 years and older. US Food and Drug Administration. Available at: https://www.fda.gov/vaccines-blood-biologics/safety-availability-biologics/cdc-and-fda-identify-preliminary-covid-19-vaccine-safety-signal-persons-aged-65-years-and-older (Accessed on February 02, 2023).
  292. COVID-19 mRNA bivalent booster vaccine safety: Vaccines and Related Biological Products Advisory Committee meeting. US Food and Drug Administration. Available at: https://www.fda.gov/media/164811/download (Accessed on February 02, 2023).
  293. Yamin D, Yechezkel M, Arbel R, et al. Safety of COVID-19 Monovalent and Bivalent BNT162b2 mRNA Vaccine Boosters for Adults 60 Years and Above: A Large-Scale Retrospective Study. UNPUBLISHED. https://papers.ssrn.com/sol3/papers.cfm?abstract_id=4336133 (Accessed on February 02, 2023).
  294. Forshee, R. Update on original COVID-19 vaccine and COVID-19 vaccine, bivalent effectiveness and safety. US Food and Drug Administration. https://www.fda.gov/media/164815/download (Accessed on February 02, 2023).
  295. Jabagi MJ, Bertrand M, Botton J, et al. Stroke, Myocardial Infarction, and Pulmonary Embolism after Bivalent Booster. N Engl J Med 2023; 388:1431.
  296. Andrews N, Stowe J, Miller E, Ramsay M. BA.1 Bivalent COVID-19 Vaccine Use and Stroke in England. JAMA 2023; 330:184.
  297. 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.
  298. 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).
  299. 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.
  300. Klein NP, Lewis N, Goddard K, et al. Surveillance for Adverse Events After COVID-19 mRNA Vaccination. JAMA 2021; 326:1390.
  301. Shimabukuro TT, Cole M, Su JR. Reports of Anaphylaxis After Receipt of mRNA COVID-19 Vaccines in the US-December 14, 2020-January 18, 2021. JAMA 2021; 325:1101.
  302. Greenhawt M, Abrams EM, Shaker M, et al. The Risk of Allergic Reaction to SARS-CoV-2 Vaccines and Recommended Evaluation and Management: A Systematic Review, Meta-Analysis, GRADE Assessment, and International Consensus Approach. J Allergy Clin Immunol Pract 2021; 9:3546.
  303. Warren CM, Snow TT, Lee AS, et al. Assessment of Allergic and Anaphylactic Reactions to mRNA COVID-19 Vaccines With Confirmatory Testing in a US Regional Health System. JAMA Netw Open 2021; 4:e2125524.
  304. Khalid MB, Frischmeyer-Guerrerio PA. The conundrum of COVID-19 mRNA vaccine-induced anaphylaxis. J Allergy Clin Immunol Glob 2023; 2:1.
  305. Takano T, Hirose M, Yamasaki Y, et al. Investigation of the incidence of immunisation stress-related response following COVID-19 vaccination in healthcare workers. J Infect Chemother 2022; 28:735.
  306. 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.
  307. 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.
  308. Chen F, Zhu S, Dai Z, et al. Effects of COVID-19 and mRNA vaccines on human fertility. Hum Reprod 2021; 37:5.
  309. 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).
  310. 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.
  311. 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.
  312. Lee GM, Romero JR, Bell BP. Postapproval Vaccine Safety Surveillance for COVID-19 Vaccines in the US. JAMA 2020; 324:1937.
  313. 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).
  314. 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.
  315. 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.
  316. Health Resources and Services Administration. Countermeasures Injury Compensation Program. https://www.hrsa.gov/cicp (Accessed on November 20, 2020).
  317. 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).
  318. Communication skills for talking about COVID vaccines https://www.vitaltalk.org/wp-content/uploads/Communication-skills-for-the-COVID-vaccine_v1.2-1.pdf (Accessed on January 21, 2021).
  319. 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).
  320. Lazarus JV, Ratzan SC, Palayew A, et al. A global survey of potential acceptance of a COVID-19 vaccine. Nat Med 2021; 27:225.
  321. Lun P, Ning K, Wang Y, et al. COVID-19 Vaccination Willingness and Reasons for Vaccine Refusal. JAMA Netw Open 2023; 6:e2337909.
  322. 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.
  323. 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.
  324. 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.
  325. 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.
  326. 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.
  327. 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.
  328. 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.
  329. Twentyman E. Updates to Interim Clinical Considerations for Use of COVID-19 Vaccines. https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2023-04-19/07-COVID-Twentyman-508.pdf (Accessed on April 20, 2023).
  330. Sinclair AH, Taylor MK, Weitz JS, et al. Reasons for Receiving or Not Receiving Bivalent COVID-19 Booster Vaccinations Among Adults - United States, November 1-December 10, 2022. MMWR Morb Mortal Wkly Rep 2023; 72:73.
Topic 129849 Version 184.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟