At which of the following time intervals should booster vaccinations be scheduled?

Booster COVID-19 vaccination for recommended groups who have already completed their primary course with any COVID-19 vaccine type

Frequently asked questions for vaccinators and health professionals 

First Booster Information

Second Booster Information

Additional resources 

  • National Immunisation Advisory Committee recommendations 
  • National Immunisation Advisory Committee Immunisation Guidelines for Ireland, Chapter 5a COVID-19
  • Clinical Guidance for COVID-19 Vaccination
  • Go back to FAQ page
This page was updated on 19 March 2022

On this page:

  • Dosing Schedules
  • Additional Primary Series Doses
    • Immunocompetent Populations
    • Immunocompromised Populations
  • Booster Doses
  • Mixed Products
  • Timing/Intervals
  • Coadministration
  • Resources
  • Multimedia

This page undergoes regular review and was last comprehensively reviewed on November 1, 2022. Some sections may reflect more recent updates.

Dosing Schedules

The four COVID-19 vaccines available for use in the U.S. follow various dosing schedules. All four vaccines have been demonstrated to be highly effective against severe outcomes related to COVID-19 in clinical trials and postauthorization observational studies. However, given concerns about waning immunity and the emergence of novel variants, there is interest in understanding the potential role of alternative dosing strategies for these vaccines to augment or prolong their protective effect, including (but not limited to) administration of additional doses of each vaccine, schedules containing different products (“mix and match”) and modified dosing intervals.

Although individuals may receive the second dose in the primary series at extended intervals, shorter intervals are still recommended for moderately to severely immunocompromised individuals, adults aged 65 and older and others who need rapid protection. The very small risk of myocarditis, especially for males aged 12-39 years, may be further reduced with an extended interval between primary doses.

Additional Primary Series Doses

Immunocompromised Populations

FDA has authorized and CDC recommends an additional dose of COVID-19 mRNA vaccine as part of the primary series for certain immunocompromised populations.

Key primary studies that have evaluated the effect of additional doses of COVID-19 vaccines as part of a primary series in immunocompromised populations include:

mRNA vaccines

  • In a randomized controlled trial of 120 solid organ transplant recipients, subjects who had previously completed a two-dose series of the Moderna COVID-19 vaccine were randomized 1:1 to receive a third dose of vaccine or saline placebo 2 months after the second dose of the primary vaccine series. Immunogenicity data were available from 117 subjects; prior to the study intervention, only 11.7% and 8.8% of the treatment and placebo groups respectively had an antireceptor binding domain antibody concentration above a prespecified threshold of 100 U/mL. At 1 month after the study intervention, 55% (33 of 60) recipients of a third dose of Moderna COVID-19 vaccine achieved that concentration, compared with only 18% (10 of 57) in the placebo group. Additionally, after the study intervention, the median percentage virus neutralization was 71% in the vaccine group compared with only 13% in the placebo group (Hall, August 2021). In a secondary analysis of this trial, the authors also found that a third dose of vaccine augmented neutralizing antibody responses against the Alpha, Beta and Delta variants (Kumar, November 2021).
  • In a prospective study of 101 solid organ transplant recipients (78 kidney, 12 liver, eight lung or heart and three pancreas) in France, the investigators evaluated immune responses to a third dose of the Pfizer-BioNTech COVID-19 vaccine administered approximately 2 months (61±1 day) after the second dose. The proportion of individuals with detectable anti-SARS-CoV-2 antibodies was 0% at baseline, 4% after dose one and 40% after dose two — this increased to 68% after dose three. Among the individuals who were seropositive after dose two, antibody titers also increased after dose three (from 36±12 to 2676±350). Finally, of the 59 individuals who were seronegative after dose two, 44% (N=26) became seropositive after the third dose (Kamar, June 2021). The same group of investigators reported similar findings in an expanded cohort of 396 solid organ transplant recipients, which included data from the 101 described previously (Del Bello, July 2021).
  • In a prospective study of 159 kidney transplant recipients in France, the investigators evaluated immune responses to a third dose of the Moderna COVID-19 vaccine administered approximately 2 months (median 51 days) after the second dose. The patients selected for this study had to have an antibody response below the cutoff for positivity at baseline (i.e., after two prior doses of Moderna vaccine). Nearly half (N=78, or 49%) of patients developed an antibody response above the cutoff for positivity after a third dose of vaccine. Compared with patients who had had no response to prior doses of vaccine, those with a weak response demonstrated a more robust response to a third dose (27.4% vs. 81.3%) (Benotmane, July 2021).
  • In a non-peer-reviewed observational study of 43 patients with lymphoid malignancies (13 with chronic lymphocytic leukemia, 14 with non-Hodgkin's lymphoma and 16 with multiple myeloma) who had received a third dose of Pfizer-BioNTech COVID-19 vaccine in France, the investigators noted that 18 (41.8%) had no baseline antibody response to prior doses of vaccine. None of these patients developed an antibody response after a third dose. Of note, 14 (77.8%) of these individuals had previously received B-cell-depleting therapy. The third dose of vaccine did elicit higher antibody titers in the majority (23 of 25) of individuals who had at least some baseline antibody response after two doses of vaccine (Re, July 2021 - preprint, not peer-reviewed).
  • In an observational study of 30 solid organ transplant recipients who received a third dose of a COVID-19 vaccine in the United States, 15 individuals (10 kidney, three liver, one heart and one pancreas) who had previously completed a two-dose series of an mRNA vaccine received a third dose of an mRNA vaccine (six received the same mRNA vaccine product, and nine received the other mRNA vaccine product). Four of these individuals had a positive anti-SARS-CoV-2 antibody concentration at baseline, and nine individuals (60%) had a positive result after a third dose (including the four who were seropositive at baseline) (Werbel, June 2021). A follow-up study using the same cohort found that sera from some solid organ transplant recipients who received a third dose of an mRNA COVID-19 vaccine demonstrated greater pseudoneutralization against SARS-CoV-2 variants of concern, though this activity was still lower than that of sera from healthy control individuals who had received only two doses of vaccine (Karaba, August 2021 – preprint, not peer-reviewed).

Viral vector vaccines

  • In an observational study of 30 solid organ transplant recipients who received a third dose of a COVID-19 vaccine in the United States, 15 individuals (12 kidney, one kidney-pancreas, one heart and one lung) who had previously completed a two-dose series of an mRNA vaccine received an additional dose of the Johnson & Johnson/Janssen COVID-19 vaccine. Only one of these individuals had a positive antibody response at baseline, and five (33%) had a positive antibody response after the third dose, including the one who was positive at baseline (Werbel, June 2021).

Immunocompetent Populations

There is no current recommendation to offer a third dose of COVID-19 vaccine as part of the primary series to immunocompetent adults. There are limited data on the effect of a third dose used in this context. However, the emergency use authorization of Pfizer-BioNTech vaccine in children 6 months-4 years has a third dose included in the primary series. The third dose was added to the primary series after immunobridging success criteria were not met for this age group with only two primary series vaccine doses.

mRNA vaccines

  • No data in immunocompetent individuals. A discussion of the decision to add a third primary dose to the vaccine series for individuals 6 months-4 years receiving Pfizer-BioNTech can be found here.

Viral vector vaccines

  • In a longitudinal immunogenicity study of the Johnson & Johnson/Janssen COVID-19 vaccine, investigators evaluated antibody and cellular responses 8 months after a first dose of that vaccine, including in 10 healthy individuals who had received a second dose of the vaccine 2 months after the first dose. Neutralizing antibody titers at the 8-month time point did not differ significantly between individuals who had received one or two doses as part of their primary series (Barouch, July 2021).

Booster Doses 

In the U.S., CDC recommends everyone ages 5 years and older receive one updated bivalent mRNA COVID-19 vaccine booster dose after completing their mRNA, viral vector or recombinant subunit primary COVID-19 vaccination series. Children who are 5 years old are only recommended to receive the Pfizer-BioNTech bivalent booster; children 6 years old and older can receive either Pfizer-BioNTech or Moderna bivalent booster. All bivalent boosters should be received at least 2 months after completion after the primary series, or at least 2 months after the last monovalent booster dose.

Bivalent mRNA vaccines contain both a vaccine antigen against the ancestral strain of SARS-CoV-2 as well as an antigen against BA.4/BA.5 Omicron subvariants. These bivalent vaccines have the same total antigen amount as adult monovalent vaccines (i.e., in the U.S., Moderna bivalent vaccine contains 25 mcg of spike protein from ancestral SARS-CoV-2 and 25 mcg of spike protein from Omicron (BA.4/BA.5) SARS-CoV-2, whereas primary series doses each contain 50mcg of spike protein from ancestral SARS-CoV-2; Pfizer bivalent vaccine contains 15 mcg of spike protein from each antigen, whereas primary series doses each contain 30mcg of spike protein from the ancestral strain only). Bivalent boosters should be administered without regard to the number of previous monovalent booster doses received. CDC suggests that where possible, booster vaccines to be received should “match” the vaccines received in the primary series (e.g., individuals receiving Moderna primary series vaccines should, where possible, receive a Moderna bivalent booster). 

Individuals should only receive one bivalent booster dose. CDC recommends all booster doses be bivalent mRNA vaccines (regardless of the vaccine type received for primary series). An analysis of safety data gathered through the v-safe surveillance system found that booster doses were safe overall and associated with fewer reactogenicity events than second doses of mRNA vaccine (Hause, February 2022). However, individuals aged 18 years and older who are unable to receive a bivalent mRNA booster, or who refuse to receive a bivalent mRNA booster, may receive a Novavax booster starting 6 months after primary series completion.

Evidence

Booster doses augment immune responses (quantitatively or qualitatively) against SARS-CoV-2 (and relevant variants).

Data for augmented immune responses following booster doses come from several studies. In a substudy of the Phase 1 clinical trial of the Pfizer-BioNTech COVID-19 vaccine, 23 participants received a booster dose of the vaccine at approximately 8 months after completion of the primary series. One month after the booster dose, neutralizing antibody titers against both wildtype virus and the Beta (B.1.351) and Delta (B.1.617.2) variants had increased to levels higher than at 1 month after the primary series (Falsey, September 2021). In another study of 97 Israeli healthcare workers aged ≥60 years who received a third dose of the Pfizer-BioNTech COVID-19 vaccine approximately 6-7 months after their primary series, the additional vaccine dose significantly increased anti-spike IgG antibody titers measured 10-19 days after vaccination (Eliakim-Raz, November 2021). 

The two largest studies were multicenter studies that compared the safety and immunogenicity of a variety of different booster vaccines, including heterologous doses. In an interim analysis of a multicenter trial conducted at 10 sites in the U.S., the investigators reported immune responses following heterologous and homologous booster doses among >400 participants who had previously been vaccinated with either two doses of an mRNA COVID-19 vaccine or one dose of the Johnson & Johnson/Janssen COVID-19 vaccine (at least 12 weeks earlier). In this study, a booster dose with any product augmented antibody concentrations, but heterologous boosts (“mixed” regimens) elicited a more robust response than homologous boosts (“matched” regimens) (Atmar, January 2022).

In another large study conducted in the U.K., the investigators reported immune responses following heterologous and homologous boosters among >2800 participants who had previously received either two doses of the Oxford-AstraZeneca or Pfizer-BioNTech COVID-19 vaccine (at least 10-12 weeks earlier). Participants were randomized to receive boosts with the Oxford-AstraZeneca, Pfizer-BioNTech, Moderna or Johnson & Johnson/Janssen COVID-19 vaccines, as well as the Novavax COVID-19 vaccine, the CureVac mRNA COVID-19 vaccine and an inactivated vaccine. All the booster vaccines augmented immune responses following a Pfizer-BioNTech COVID-19 vaccine series, with the two currently authorized mRNA COVID-19 vaccines demonstrating the greatest effect (Munro, December 2021).

Limitations

The durability of these augmented immune responses is unknown. In a longitudinal study of immune responses to SARS-CoV-2 following mRNA COVID-19 vaccination (mostly the Pfizer-BioNTech COVID-19 vaccine), the investigators separately analyzed the kinetics of the immune response in individuals with and without prior SARS-CoV-2 infection. For those with prior infection, the primary vaccine series may already be considered a “booster.” In this group, antibody, memory B cell and T cell responses were augmented by vaccination but had declined nearly back to baseline by 6 months (Goel, October 2021).

Booster doses lead to increased vaccine effectiveness against COVID-19.

Multiple studies have evaluated the clinical effectiveness of a COVID-19 vaccine booster dose. These are mostly observational cohort studies during time periods when either Delta or Omicron were the predominant circulating variants. Together, the data indicate that a booster dose of an mRNA COVID-19 vaccine (i.e., a third dose given several months after a two-dose primary series) provides a significant protective effect against symptomatic COVID-19 (Bar-On, September 2021; Saciuk, November 2021; Patalon, November 2021; Bar-On, December 2021), COVID-19 hospitalization and death compared with two doses of vaccine (Barda, October 2021; Arbel, December 2021; Andrews, January 2022). Preliminary evidence suggests this may also be the case for NVX-CoV2373 when used as a booster after ChAdOx1 or BNT162b2 primary series (Munro, December 2021).

The emergence of the Omicron variant and its capacity for immune escape also motivated a number of studies to evaluate the benefits of booster doses. Booster doses of mRNA COVID-19 and the Johnson & Johnson/Janssen COVID-19 vaccines appear to restore in vitro neutralization titers (Nemet, December 2021; Garcia-Beltran, December 2021; Pajon, January 2022; Lyke, January 2022 – preprint, not peer-reviewed) as well as clinical effectiveness (Andrews, December 2021 – preprint, not peer-reviewed; Thompson, January 2022; Johnson, January 2022; Accorsi, January 2022; Gray, December 2021 – preprint, not peer-reviewed; Hui Xuan Tan, February 2022; Ferdinands, February 2022). As an example, in a non-peer-reviewed analysis of SARS-CoV-2 infections in the U.K. following the emergence of the Omicron variant, the authors found that two doses of the Pfizer-BioNTech COVID-19 vaccine demonstrated a vaccine effectiveness against symptomatic COVID-19 (presumed due to Omicron) of 88.0% (95% CI, 65.9-95.8%) 2-9 weeks after dose 2, 48.5% (95% CI, 24.3-65.0%) at 10-14 weeks post dose 2 and 34-37% from 15 weeks post dose 2; 2 weeks after a booster dose with the Pfizer COVID-19 vaccine, vaccine effectiveness had increased to 75.5% (95% CI, 56.1-86.3%) (Andrews, December 2021 – preprint, not peer-reviewed).

Limitations

The durability of this improvement in vaccine effectiveness is poorly characterized. In an analysis of urgent care encounters, emergent department visits and hospitalizations due to COVID-19 in the VISION Network in the U.S., investigators found waning effectiveness of third (booster) doses of mRNA COVID-19 vaccines against all three outcomes after 2 months, during both the Delta and Omicron waves. Notably, booster dose effectiveness waned more significantly against urgent care and emergency department encounters, but showed more durability against hospitalization, remaining greater than 75% for up to 4 months after vaccination during both time periods (Ferdinands, February 2022).

Bivalent booster doses likely provide additional protection against Omicron subvariants of SARS-CoV-2. 

Preliminary safety and efficacy information presented at a Sept. 1, 2022 ACIP meeting suggest that the inclusion of a second SARS-CoV-2 variant antigen in mRNA vaccines broadens the overall antibody response to SARS-CoV-2. In presentations to ACIP, safety data about bivalent vaccines from approximately 1,400 individuals was presented. The information presented concerned bivalent vaccines, all of which included an antigen for the SARS-CoV-2 ancestral strain; some bivalent vaccines also included antigen for the BA.1 Omicron subvariant and others included antigen for BA.4/BA.5 Omicron subvariants. Overall, Omicron-specific bivalent booster vaccines resulted in higher anti-Omicron antibody and higher antibody titers for other SARS-CoV-2 variants. Modeling work presented at this meeting suggested that broad uptake of these updated bivalent vaccines in the early fall of 2022 could prevent a substantial number of hospitalizations. 

Mixed Products

subunit and viral vector COVID-19 vaccines. Interim CDC guidance addresses clinical considerations related to heterologous booster doses, including patient benefit-risk considerations when selecting which booster dose to receive.

Evidence 

Primary series 

There have been few studies evaluating a mixed product primary series with the vaccines currently available in the U.S. The largest such study, the Com-COV2 trial, was conducted in the U.K. and compared the safety and immunogenicity of two doses of the Pfizer-BioNTech and Oxford-AstraZeneca vaccines with heterologous schedules containing one dose of either followed by one dose of either the Moderna or Novavax COVID-19 vaccine (Stuart, December 2021). The safety and immunogenicity of a vaccine series containing two doses of the Pfizer-BioNTech vaccine were similar to a vaccine series containing one dose of Pfizer-BioNTech followed by one dose of Moderna. Antibody and cellular response were slightly higher in the mixed schedule group, which may be attributable to the higher antigen content in the Moderna vaccine rather than any benefit of a mixed schedule per se.

The Com-COV2 trial and other studies outside the U.S. have also evaluated mixed schedules containing one dose of an mRNA vaccine and one dose of the Oxford-AstraZeneca vaccine (which is based on a similar technology to the Johnson & Johnson/Janssen COVID-19 vaccine), though the time between the first and second dose of vaccine in these studies has been variable. Most of these studies have concluded that a two-dose schedule that includes both vaccines, in either order, generates a robust antibody and cellular response, compared with a single dose of either vaccine. Furthermore, in the studies where a heterologous and homologous (i.e., containing two doses of the same vaccine product) schedule were directly compared, the safety profile and immune responses with both schedules appeared to be similar (Borobia, June 2021; Shaw, May 2021; Liu, August 2021; Ostadgavahi, May 2021; Hillus, November 2021; Schmidt, September 2021; Tenbusch, July 2021; Dimeglio, August 2021). In some studies, a heterologous schedule containing one Oxford-AstraZeneca and one mRNA COVID-19 vaccine dose elicited a more robust cellular response and higher neutralizing antibody titers against SARS-CoV-2 variants than a homologous schedule containing two doses of the Oxford-AstraZeneca vaccine (Barros-Martins, July 2021; Kaku, February 2022).

Boosters 

Some studies suggest that heterologous boosters may be superior to homologous boosters. Per CDC guidance, mRNA COVID-19 boosters are preferred over the Johnson & Johnson/Janssen COVID-19 vaccine. Data to support this recommendation come from studies on immunogenicity (Atmar, January 2022; Munro, December 2021; Sablerolles, January 2022) and clinical effectiveness (Mayr, February 2022; Hui Xuan Tan, February 2022).

Timing/Intervals

The interval between doses of two-dose COVID-19 vaccines may impact their immunogenicity and clinical effectiveness. However, to date there are limited data for this strategy; therefore, alternative schedules are not currently recommended.

Key primary studies that have evaluated the effect of alternate COVID-19 vaccine schedules are summarized below.

mRNA vaccine interval

There is accumulating evidence that a longer interval between the two doses of mRNA COVID-19 vaccines may confer improved immunogenicity and potentially even clinical effectiveness. Studies in the U.K. and Canada have demonstrated that a longer time period (ranging from 6-16 weeks depending on the study) between the two doses of both the Pfizer-BioNTech and Moderna COVID-19 vaccines is associated with increased antibody and cellular responses (Parry, August 2021; Grunau, November 2021; Grunau, December 2021; Payne, November 2021). A separate analysis in the U.K. of 750 adults aged 50-79 years confirmed these findings and further demonstrated that an interval of >45 days between dose one and two of the Pfizer-BioNTech COVID-19 vaccine was associated with improved vaccine effectiveness (over a follow-up period that preceded emergence of the Delta variant) against SARS-CoV-2 infection (Amirthalingam, December 2021).

Viral vector vaccine interval

In an exploratory analysis of a Phase 3 randomized controlled trial of the Oxford-AstraZeneca COVID-19 vaccine, investigators evaluated the impact of variable timing of the second dose of vaccine. In this analysis, vaccine efficacy against primary symptomatic COVID-19 (starting 14 days after the second dose) was higher with longer dose intervals. Vaccine efficacy was 55.1% (95% CI, 33.0-69.9) when the interval between the two doses was less than 6 weeks and 81.3% (60.3-91.2) when the interval was more than 12 weeks (Voysey, February 2021).

Coadministration (with non-COVID-19 vaccines)

Per CDC recommendations, COVID-19 vaccines can be administered without regard to timing of other vaccines. Of note, the effect of coadministration or closely spaced administration of COVID-19 and non-COVID-19 vaccines on immunogenicity and reactogenicity has not yet been well characterized.

Individuals receiving an orthopoxvirus vaccine (JYNNEOS or ACAM2000) may consider waiting 4 weeks before receiving a COVID-19 vaccine due to a risk of myocarditis/pericarditis associated with orthopoxvirus vaccines. If an orthopoxvirus vaccine is being administered in the context of prophylaxis, administration should not be delayed because of recent receipt of a COVID-19 vaccine.

Resources

How many seconds are counted when assessing a child's respirations?

Count the respirations for 30 seconds and double that number to determine the respiratory rate. Remember that respiratory rates that are too fast or too slow for the child's development prevent adequate oxygenation.

Which of the following is the appropriate area for measuring the head circumference?

Head circumference or OFC [occipital frontal circumference] is measured over the most prominent part on the back of the head (occiput) and just above the eyebrows (supraorbital ridges). This can be translated to mean the largest circumference of the head.

What is the only vaccine that is contraindicated in healthy infants under 6 months of age?

ACIP recommends that yellow fever vaccine never be given to infants aged <6 months.

Which muscle is the preferred site for injections on infants to 2 years of age?

For infants and younger children receiving more than two injections in a single limb, the thigh is the preferred site because of the greater muscle mass.