5
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Interim Recommendations for Use of Bivalent mRNA COVID-19 Vaccines for Persons Aged ≥6 Months — United States, April 2023

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Throughout the national public health emergency declared in response to the COVID-19 pandemic, CDC, guided by the Advisory Committee on Immunization Practices (ACIP), has offered evidence-based recommendations for the use of COVID-19 vaccines in U.S. populations after each regulatory action by the Food and Drug Administration (FDA). During August 2022–April 2023, FDA amended its Emergency Use Authorizations (EUAs) to authorize the use of a single, age-appropriate, bivalent COVID-19 vaccine dose (i.e., containing components from the ancestral and Omicron BA.4/BA.5 strains in equal amounts) for all persons aged ≥6 years, use of bivalent COVID-19 vaccine doses for children aged 6 months–5 years, and additional bivalent doses for immunocompromised persons and adults aged ≥65 years ( 1 ). ACIP voted in September 2022 on the use of the bivalent vaccine, and CDC made recommendations after the September vote and subsequently, through April 2023, with input from ACIP. This transition to a single bivalent COVID-19 vaccine dose for most persons, with additional doses for persons at increased risk for severe disease, facilitates implementation of simpler, more flexible recommendations. Three COVID-19 vaccines are currently available for use in the United States and recommended by ACIP: 1) the bivalent mRNA Pfizer-BioNTech COVID-19 vaccine, 2) the bivalent mRNA Moderna COVID-19 vaccine, and 3) the monovalent adjuvanted, protein subunit-based Novavax COVID-19 vaccine.* As of August 31, 2022, monovalent mRNA vaccines based on the ancestral SARS-CoV-2 strain are no longer authorized for use in the United States ( 1 ). Since June 2020, ACIP has convened 35 public meetings to review data relevant to the potential use of COVID-19 vaccines. † The ACIP COVID-19 Vaccine Work Group, comprising experts in adult and pediatric medicine, infectious diseases, vaccinology, vaccine safety, public health, and ethics, has met weekly to review COVID-19 surveillance data, evidence for immunogenicity, efficacy, postauthorization effectiveness, safety of COVID-19 vaccines, and implementation considerations. To assess the evidence for benefits and harms associated with use of bivalent vaccines, and to guide deliberations, ACIP used the Evidence to Recommendations (EtR) Framework. § Within this framework, ACIP considered the importance of COVID-19 as a public health problem, including during the Omicron-predominant era, as well as issues of resource use, benefits and harms, patients’ values and preferences, acceptability, feasibility, and equity related to use of the vaccines. ACIP held three public meetings on September 1, 2022, February 24, 2023, and April 19, 2023, to discuss bivalent vaccine policy using the EtR Framework. ACIP voted on adult bivalent doses on September 1, 2022. Authorization for bivalent vaccines was subsequently extended to additional age groups, and CDC updated recommendations, guided by February 24, 2023, and April 19, 2023, input from ACIP (Box). To better protect against the Omicron variant, which emerged in November 2021, ACIP recommended a dose of bivalent mRNA vaccine (containing mRNA encoding the spike protein from both the ancestral SARS-CoV-2 and Omicron BA.4/BA.5 SARS-CoV-2 variants) in September 2022 ( 2 ). Among persons who had only received monovalent COVID-19 vaccines, bivalent COVID-19 vaccines have provided additional protection against infection and COVID-19–associated hospitalization; however, that protection might wane over time ( 3 ). From September 2022 to March 2023, vaccine effectiveness (VE) against emergency department and urgent care visits by adults aged 18–64 years waned from 53% (95% CI = 48%–58%) at 7–59 days after receipt of a bivalent dose to 42% (95% CI = 35%–47%) at 60–119 days. Protection against hospitalization among adults aged 18–64 years without an immunocompromising condition waned from 68% (95% CI = 53%–79%) at 7–59 days to 27% (95% CI = 2%–46%) at 60–119 days ( 3 ). BOX Timeline of COVID-19 bivalent vaccine authorizations, by Food and Drug Administration and CDC vaccine recommendations — United States, August 2022–April 2023 August and September 2022 FDA authorizes and CDC recommends, with a single ACIP vote, 1) a single dose of Pfizer-BioNTech bivalent vaccine for persons aged ≥12 years 2 or more months after receipt of a primary series or previous monovalent booster dose and 2) a single dose of Moderna bivalent vaccine for adults aged ≥18 years 2 or more months after receipt of a primary series or previous monovalent booster dose. October 2022 FDA authorizes and CDC recommends, with ACIP input, a single dose of Pfizer-BioNTech bivalent vaccine for children aged 5–11 years 2 or more months after receipt of a primary series or previous monovalent booster dose. December 2022 FDA authorizes and CDC recommends, with ACIP input, a single dose of Moderna bivalent vaccine for children aged 6 months–5 years 2 or more months after receipt of a primary series. FDA authorizes and CDC recommends, with ACIP input, a single dose of Pfizer-BioNTech bivalent vaccine for children aged 6 months–4 years as the third dose in a primary series 8 or more weeks after receipt of 2 monovalent doses of Pfizer-BioNTech vaccine. March 2023 FDA authorizes and CDC recommends, with ACIP input, a single dose of Pfizer-BioNTech bivalent vaccine for children aged 6 months–4 years who received 3 monovalent doses of Pfizer-BioNTech as a primary series. April 2023 FDA authorizes and CDC recommends, with ACIP input, a single dose of bivalent vaccine for all persons aged ≥6 years who are unvaccinated or 2 or more months after receipt of a previous monovalent dose. FDA authorizes and CDC recommends, with ACIP input, at the time of initial vaccination (depending on vaccine product) 2 or 3 doses of bivalent vaccine for children aged 6 months–4 years and 1 or 2 doses of bivalent vaccine for children aged 5 years. FDA authorizes and CDC recommends, with ACIP input, that persons aged ≥65 years may receive a single additional bivalent vaccine dose 4 or more months after receipt of their first bivalent dose. FDA authorizes and CDC recommends, with ACIP input, that persons aged ≥6 months who are moderately or severely immunocompromised may receive an optional additional bivalent dose 2 or more months after the most recent bivalent dose and additional bivalent doses as needed. Abbreviations: ACIP = Advisory Committee on Immunization Practices; FDA = Food and Drug Administration. As of May 6, 2023, COVID-19–associated hospitalization rates were highest among adults aged ≥65 years (9.5 per 100,000 persons). ¶ Bivalent booster doses are shown to provide the highest protection against hospitalization among adults, with protection sustained through at least 179 days against critical outcomes, including intensive care unit admission or in-hospital death ( 4 ). However, only 17% of the U.S. population overall and 43.3% of adults aged ≥65 years have received a bivalent dose.** Primary series coverage (i.e., receipt of a complete COVID-19 vaccination series) follows a similar pattern: it is highest among older adults and lowest among young children. Recommendations for Use of Bivalent COVID-19 Vaccines in Persons Aged ≥6 Years Without Immunocompromising Conditions On April 18, 2023, FDA authorized, and on April 20, 2023, CDC recommended a single, age-appropriate bivalent mRNA dose for unvaccinated persons aged ≥6 years without moderate or severe immunocompromise. Previously vaccinated persons without moderate or severe immunocompromise were recommended to receive the vaccine ≥2 months after receipt of any monovalent vaccine dose (Table). TABLE COVID-19 vaccines recommended for persons aged ≥6 months, by immunocompromise status and age group — CDC, United States, April 2023* Immunocompromise status Age group Recommendation Not moderately or severely immunocompromised 6 mos–5 yrs At the time of initial vaccination, depending on vaccine product: 2 or 3 doses of bivalent vaccine for children aged 6 mos–4 yrs (Pfizer-BioNTech); and 1 or 2 doses of bivalent vaccine for children aged 5 yrs (Moderna)† ≥6 yrs A single bivalent dose ≥65 yrs A single bivalent dose and 1 additional, optional, bivalent dose Moderately or severely immunocompromised ≥6 mos A single bivalent dose and additional, optional, bivalent doses as needed * https://www.cdc.gov/vaccines/covid-19/clinical-considerations/interim-considerations-us.html † https://www.cdc.gov/vaccines/covid-19/clinical-considerations/interim-considerations-us.html#not-immunocompromised CDC’s recommendation was based on input from ACIP during public meetings held on February 24, 2023, and April 19, 2023. At these meetings, discussions were guided by clinical trial data demonstrating that bivalent vaccines induce an immune response when administered as a primary series. Immunogenicity data demonstrated that a primary series of an Omicron BA.1-containing bivalent vaccine induced neutralization titers against BA.1 that were approximately 25 times those induced by the original monovalent vaccine †† ( 5 ). The percentage of patients reporting solicited systemic and local reactogenicity after receiving the BA.1-containing vaccine was similar to or less than the percentage reporting these reactions after a receipt of a monovalent vaccine. §§ Unsolicited adverse events generally represented illnesses and commonly reported events during infancy and childhood ( 5 ). CDC recommendations were also guided by ACIP discussions of seroprevalence data indicating that, for most older children, adolescents, and adults, future doses will provide an additional boost after previous infection, previous vaccination, or both. In a March–December 2022 nationwide seroprevalence study conducted among children aged 6 months–17 years, most children and adolescents had evidence of infection-induced immunity; prevalence of infection-induced immunity was highest among persons aged 5–11 and 12–17 years (93%) and lowest among children aged 6–11 months (63%) ( 6 ). Most adults also had preexisting antibodies against SARS-CoV-2. In a January–March 2022 seroprevalence study of adult blood donors aged ≥16 years, 96% had evidence of immunity from either previous infection, previous vaccination, or both ( 7 ). A rare risk for myocarditis and pericarditis has been identified after receipt of monovalent mRNA COVID-19 vaccines, primarily in adolescent and young adult males. Because data are limited, the risk for myocarditis or pericarditis after receipt of a bivalent dose is not known; however, preliminary estimates suggest that the risk is lower than that observed after a second primary series monovalent dose ( 8 ). Higher rates of myocarditis have also been associated with a shorter interval between doses ( 9 ). Because of the small number of doses administered among adolescent and young adult males, estimating the incidence of myocarditis after a bivalent dose was not possible; however, only a single case of myopericarditis has been observed in the Vaccine Safety Datalink (a postauthorization vaccine safety monitoring system) during the 7 days after receipt of a bivalent dose in a male aged 18–29 years ( 8 ). Recommendations from CDC were also guided by ACIP discussions of the benefits (i.e., reduction in the number of hospitalizations, intensive care unit admissions, and the number of deaths prevented) per 1 million primary series and bivalent vaccine doses administered, stratified by both age group and interval between primary series completion and receipt of a first bivalent dose. Benefits of a primary series and bivalent dose were seen among all age groups and at all intervals; however, the largest observed benefits were among the oldest age groups and those with the longest interval (i.e., ≥11 months) between completion of the primary series and receipt of the bivalent dose ( 10 ). Regular review of safety data, including myocarditis and pericarditis risk after bivalent doses, will continue in national safety surveillance systems. Recommendations for Use of Bivalent COVID-19 Vaccine for Children Aged 6 Months–5 Years During December 2022–April 2023, FDA amended multiple authorizations for bivalent mRNA vaccines for children aged 6 months–5 years. During this period, CDC updated recommendations, with input from ACIP, for children in this age group for use of bivalent doses based on a child’s vaccination history (Table). Among children aged 6 months–4 years, either mRNA vaccine may be used; however, all doses administered to a given child must be from the same manufacturer. Among those receiving Moderna vaccine, ≥2 doses are authorized, including ≥1 bivalent vaccine dose. Among those receiving Pfizer-BioNTech vaccine, ≥3 doses are authorized, including ≥1 bivalent vaccine dose. Based on FDA authorizations, unvaccinated children aged 5 years are authorized to receive 2 doses of Moderna bivalent vaccine (with 4–8 weeks between doses) or 1 dose of Pfizer-BioNTech vaccine. Children aged 5 years who received 1 or 2 doses of monovalent Moderna vaccine are authorized to receive 1 dose of either the bivalent Moderna or Pfizer-BioNTech vaccine. ¶¶ Those who received ≥1 doses of monovalent Pfizer-BioNTech vaccine are authorized to receive ≥1 bivalent Pfizer-BioNTech vaccine doses. CDC recommendations for pediatric immunization were guided by ACIP discussion of studies of bivalent vaccine given as a primary series in children, as well as booster doses among adults, demonstrating that a bivalent dose of either Moderna or Pfizer-BioNTech vaccine broadens the immune response in persons who have received a primary series and a previous monovalent booster dose ( 5 ). Compared with a monovalent booster dose (based on the ancestral SARS-CoV-2 strain), the immune response to Omicron was superior and that to the ancestral strain was noninferior among bivalent vaccine booster dose recipients. Monovalent booster doses of Moderna COVID-19 vaccines were studied in a clinical trial of 145 children aged 17 months–5 years who had received a Moderna primary series 8–13 months previously. Antibody levels after receipt of the monovalent booster dose in a subset of 56 children without previous SARS-CoV-2 infection were four times higher than were levels after the primary series in 294 young adults ( 11 ). Reactogenicity was similar to that observed after receipt of booster doses in other age groups. In a subset of 60 Pfizer-BioNTech pediatric trial participants aged 6 months–4 years who received a single bivalent Pfizer-BioNTech vaccine dose after completion of a 3-dose monovalent primary series, Omicron BA.4/BA.5–specific antibodies were higher compared with those among children who completed the 3-dose primary series of monovalent Pfizer-BioNTech vaccine and did not receive the bivalent booster dose. The bivalent dose was generally well-tolerated, with a lower frequency of postvaccination local and systemic reactions than previously observed in this age group with monovalent doses; no new or concerning safety findings were identified ( 12 ). Additional Bivalent COVID-19 Doses for Adults Aged ≥65 Years and for Persons Aged ≥6 Months Who Are Moderately or Severely Immunocompromised In April 2023, FDA granted an EUA for additional bivalent doses for adults aged ≥65 years and for persons aged ≥6 months with immunocompromise. Adults aged ≥65 years have the option to receive 1 additional bivalent vaccine dose ≥4 months after receipt of the most recent bivalent dose (Table). Persons aged ≥6 months who are moderately or severely immunocompromised have the option to receive ≥1 additional bivalent doses ≥2 months after receipt of the most recent bivalent dose and additional bivalent mRNA doses, as indicated, based on individual circumstances and clinical judgment.*** The option to receive ≥1 additional bivalent mRNA vaccine doses may be based on the clinical judgment of a health care provider, a person’s risk for severe COVID-19 because of the presence of underlying medial conditions and age, and personal preference and circumstances. CDC made recommendations based on ACIP discussions of VE and clinical epidemiology of COVID-19 among moderately or severely immunocompromised persons and adults aged ≥65 years. Effectiveness of a bivalent vaccine booster dose against hospitalization in adults aged ≥18 years with immunocompromising conditions was 30% (95% CI = 12%–44%) at 7–59 days postvaccination and 31% (95% CI = 4%–50%) at 120–179 days ( 3 ). Among adults aged ≥65 years, waning of absolute VE has been noted after receipt of a bivalent dose. Effectiveness of bivalent vaccines against COVID-19–associated emergency department or urgent care encounters among immunocompetent adults aged ≥65 years declined from 61% (95% CI = 57%–64%) 7–59 days after vaccination to 25% (95% CI = 16%–34%) at 120–179 days ( 3 ). VE against COVID-19–associated hospitalization declined from 64% (95% CI = 59%–69%) 7–59 days after vaccination to 39% (95% CI = 26%–50%) at 120–179 days ( 3 ). Implementation Considerations Before the authorization of a bivalent dose for most persons, 11 mRNA COVID-19 vaccine products were licensed or authorized for use. Authorization of a bivalent dose for most persons reduced the total number of vaccine products to five and eliminated vials that appear similar (i.e., look-alike vials) ( 13 ); these recommendations will thereby simplify implementation for COVID-19 vaccine providers. Reducing the number of products will expand providers’ storage space and, in conjunction with the elimination of look-alike vials, might reduce vaccine administration errors. The transition from a monovalent primary series to a single bivalent dose for most persons, and additional bivalent doses for populations at higher risk for severe disease, allows the COVID-19 vaccination program to progress toward simpler, more flexible, evidence-based recommendations. COVID-19 vaccination remains critical to protecting against serious consequences of COVID-19, and all persons aged ≥6 months should stay up to date with recommended COVID-19 vaccination, including receiving ≥1 bivalent vaccine dose. Before vaccination, providers should provide the EUA Fact Sheet for the vaccine being administered and counsel vaccine recipients about expected systemic and local adverse reactions (reactogenicity). Additional clinical education materials are available, ††† including further clinical considerations. §§§ These interim recommendations and clinical considerations are based on currently available information regarding bivalent COVID-19 vaccine doses and might change as more evidence becomes available. Reporting of Vaccine Adverse Events Adverse events that occur after receipt of any COVID-19 vaccine should be reported to the Vaccine Adverse Event Reporting System (VAERS, https://vaers.hhs.gov or 1-800-822-7967). Vaccination providers are required by FDA to report vaccine administration errors, serious adverse events, cases of myocarditis, cases of pericarditis, cases of multisystem inflammatory syndrome, hospitalization or death, and cases of COVID-19 that result in hospitalization or death after administration of COVID-19 vaccine under EUA. Reporting is encouraged for any clinically significant adverse event even if it is uncertain whether the vaccine caused the event. Summary What is already known about this topic? During August–October 2022, CDC recommended a bivalent COVID-19 mRNA vaccine dose for all persons aged ≥5 years who had received a monovalent primary vaccination series. What is added by this report? During December 2022–April 2023, CDC made recommendations for a single bivalent vaccine dose for most persons aged ≥6 years, bivalent vaccines for children aged 6 months–5 years, and optional additional bivalent booster doses for moderately or severely immunocompromised persons aged ≥6 months and adults aged ≥65 years. What are the implications for public health practice? Transition to a single bivalent COVID-19 vaccine dose for most persons, with additional doses for persons at increased risk for severe disease, facilitates implementation of simpler, more flexible recommendations. All persons aged ≥6 months should receive ≥1 bivalent vaccine dose.

          Related collections

          Most cited references4

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Interim Recommendations from the Advisory Committee on Immunization Practices for the Use of Bivalent Booster Doses of COVID-19 Vaccines — United States, October 2022

            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            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

              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              The Advisory Committee on Immunization Practices’ Recommendation for Use of Moderna COVID-19 Vaccine in Adults Aged ≥18 Years and Considerations for Extended Intervals for Administration of Primary Series Doses of mRNA COVID-19 Vaccines — United States, February 2022

              The mRNA-1273 (Moderna) COVID-19 vaccine is a lipid nanoparticle-encapsulated, nucleoside-modified mRNA vaccine encoding the stabilized prefusion spike glycoprotein of SARS-CoV-2, the virus that causes COVID-19. During December 2020, the vaccine was granted Emergency Use Authorization (EUA) by the Food and Drug Administration (FDA), and the Advisory Committee on Immunization Practices (ACIP) issued an interim recommendation for use among persons aged ≥18 years ( 1 ), which was adopted by CDC. During December 19, 2020–January 30, 2022, approximately 204 million doses of Moderna COVID-19 vaccine were administered in the United States ( 2 ) as a primary series of 2 intramuscular doses (100 μg [0.5 mL] each) 4 weeks apart. On January 31, 2022, FDA approved a Biologics License Application (BLA) for use of the Moderna COVID-19 vaccine (Spikevax, ModernaTX, Inc.) in persons aged ≥18 years ( 3 ). On February 4, 2022, the ACIP COVID-19 Vaccines Work Group conclusions regarding recommendations for the use of the Moderna COVID-19 vaccine were presented to ACIP at a public meeting. The Work Group’s deliberations were based on the Evidence to Recommendation (EtR) Framework,* which incorporates the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) approach † to rank evidence quality. In addition to initial clinical trial data, ACIP considered new information gathered in the 12 months since issuance of the interim recommendations, including additional follow-up time in the clinical trial, real-world vaccine effectiveness studies, and postauthorization vaccine safety monitoring. ACIP also considered comparisons of mRNA vaccine effectiveness and safety in real-world settings when first doses were administered 8 weeks apart instead of the original intervals used in clinical trials (3 weeks for BNT162b2 [Pfizer-BioNTech] COVID-19 vaccine and 4 weeks for Moderna COVID-19 vaccine). Based on this evidence, CDC has provided guidance that an 8-week interval might be optimal for some adolescents and adults. The additional information gathered since the issuance of the interim recommendations increased certainty that the benefits of preventing symptomatic and asymptomatic SARS-CoV-2 infection, hospitalization, and death outweigh vaccine-associated risks of the Moderna COVID-19 vaccine. On February 4, 2022, ACIP modified its interim recommendation to a standard recommendation § for use of the fully licensed Moderna COVID-19 vaccine in persons aged ≥18 years. Recommendations for Use of Moderna COVID-19 Vaccine During June 2020–February 2022, ACIP convened 23 public meetings to review data on the epidemiology of COVID-19 and considerations for use of all COVID-19 vaccines, including the Moderna COVID-19 vaccine ( 4 ). The ACIP COVID-19 Vaccines Work Group, which includes experts in infectious diseases, vaccinology, vaccine safety, public health, and ethics, held meetings each week to review COVID-19 epidemiologic and surveillance data on vaccine efficacy, effectiveness, and safety and implementation considerations. After a systematic review of published and unpublished scientific evidence for benefits and harms ¶ of Moderna COVID-19 vaccination, the Work Group used a modified GRADE approach to assess the certainty of evidence for outcomes related to the vaccine, rated on a scale of type 1 to type 4 (type 1 = high certainty, type 2 = moderate certainty, type 3 = low certainty, and type 4 = very low certainty). Within the EtR Framework, ACIP considered the importance of COVID-19 as a public health problem, benefits and harms (as informed by the GRADE evidence assessment), patients’ values and preferences, issues of resource use, acceptability to stakeholders, feasibility of implementation, and anticipated impact on health equity. Work Group conclusions regarding the evidence for the Moderna COVID-19 vaccine were presented to ACIP at a public meeting on February 4, 2022.** The body of scientific evidence for potential benefits and harms of the Moderna COVID-19 vaccine was guided by one large randomized, double-blind, placebo-controlled Phase III clinical trial ( 5 , 6 ), one Phase II clinical trial ( 7 ), one small Phase I clinical trial ( 8 , 9 ), 26 observational vaccine effectiveness studies, and two postauthorization vaccine safety monitoring systems: the Vaccine Adverse Events Reporting System (VAERS) and the Vaccine Safety Datalink (VSD). VAERS is a national passive surveillance vaccine safety monitoring system managed by CDC and FDA. VSD covers nine participating integrated health care organizations serving approximately 12 million persons and identifies possible adverse events after vaccination, using detailed clinical and demographic data available in near real time from electronic medical records. Updated findings from the ongoing Phase III clinical trial were based on 30,420 enrolled participants contributing approximately 11,000 person-years of data, with a median follow-up of 5 months during September 4, 2020–March 26, 2021 (ending with the date placebo recipients were offered crossover to receive study vaccine). Pooled effectiveness estimates were calculated when multiple observational studies reported data on a specific outcome; the study periods for the observational studies included in the pooled estimates ranged from 1 to 10 months (median = 5 months). The estimated efficacy of the Moderna COVID-19 vaccine in the Phase III clinical trial was based on outcomes that occurred ≥14 days after receipt of the second dose. The demographic characteristics of participants, including age and race ( 5 ), have remained consistent since initial enrollment. Efficacy in preventing symptomatic, laboratory-confirmed COVID-19 in persons aged ≥18 years without evidence of previous SARS-CoV-2 infection was 92.7% (Table 1). One hospitalization occurred among the vaccinated group and 24 hospitalizations among the placebo group, yielding an estimated vaccine efficacy of 95.9% against COVID-19–associated hospitalization. No COVID-19–associated deaths occurred among study participants in the vaccinated group, and three occurred in the placebo group resulting in a vaccine efficacy of 100% against COVID-19–associated deaths. Efficacy in preventing asymptomatic SARS-CoV-2 infection was 57.4%. Observational data were available for all beneficial outcomes assessed: the pooled vaccine effectiveness estimates were 89.2% for prevention of symptomatic, laboratory-confirmed COVID-19 (11 studies); 94.8% against COVID-19–associated hospitalizations (15 studies), 93.8% against COVID-19–associated death (five studies), and 69.8% against asymptomatic SARS-CoV-2 infection (three studies). Most of the follow-up time occurred before B.1.1.529 (Omicron) became the predominant circulating SARS-CoV-2 variant. From the GRADE evidence assessment, the level of certainty for the benefits of Moderna COVID-19 vaccination among persons aged ≥18 years was type 1 (high certainty) for the prevention of symptomatic SARS-CoV-2 infection, type 1 (high certainty) for the prevention of asymptomatic SARS-CoV-2 infection, type 2 (moderate certainty) for prevention of COVID-19–associated hospitalization, and type 2 (moderate certainty) for the prevention of COVID-19–associated death. TABLE 1 Summary of the certainty of evidence of potential benefits of Moderna COVID-19 vaccination — United States, February 2022 Potential benefit Clinical trial evidence Observational evidence GRADE evidence certainty† No. of studies Vaccine efficacy (95% CI) No. of studies Pooled vaccine effectiveness* (95% CI) Prevention of symptomatic, laboratory-confirmed COVID-19§ 1 92.7 (90.4–94.4) 11 89.2 (82.0–93.6) 1 Prevention of COVID-19–associated hospitalization§ 1 95.9 (69.5–99.4) 15 94.8 (93.1–96.1) 2 Prevention of COVID-19–associated death 1 100 (NE–100) 5 93.8 (91.5–95.4) 2 Prevention of asymptomatic SARS-CoV-2 infection 1 57.4 (50.1–63.6) 3 69.8 (60.9–76.7) 1 Abbreviations: GRADE = Grading of Recommendations, Assessment, Development and Evaluation; NE = not evaluable. * Vaccine effectiveness estimates were pooled to provide an overall estimate across studies for the purposes of GRADE review. † GRADE evidence certainty: 1 = high certainty, 2 = moderate certainty, 3 = low certainty, 4 = very low certainty. § Considered a critical outcome in GRADE. https://www.cdc.gov/vaccines/acip/recs/grade/about-grade.html In the Phase III clinical trial, severe local and systemic adverse reactions (i.e., reactogenicity) in the 7 days after vaccination (grade 3 or higher, †† defined as adverse reactions interfering with daily activity) were more likely to occur among vaccine recipients (21.3%) than placebo recipients (4.5%) (relative risk = 5.03; 95% CI = 4.65–5.45) (Table 2). Among vaccine recipients, the most common grade 3 symptoms were fatigue, headache, joint pain, muscle pain, and injection-site pain. Overall, reactions categorized as grade 3 or higher were more likely to be reported after the second dose than after the first dose. The frequency of serious adverse events §§ was 1.7% among vaccine recipients and 1.9% among placebo recipients. Based on data from VAERS and VSD, two rare but clinically serious adverse events after vaccination were detected: anaphylaxis and myocarditis or myopericarditis. ¶¶ Based on VSD data, 5.1 cases of anaphylaxis per 1 million doses of Moderna COVID-19 vaccine administered among persons aged ≥18 years were observed ( 10 ). Myocarditis or pericarditis were more common among vaccine recipients who were younger and male, and occurred more frequently after the second vaccine dose; 65.7 cases per 1 million doses of Moderna COVID-19 vaccine administered were observed from analysis of VSD chart-reviewed myocarditis and myopericarditis cases that met CDC case definitions ( 11 ) among men aged 18–39 years after dose 2 and occurring within a 0–7-day risk interval after vaccination. Although VAERS data are subject to the limitations of a passive surveillance system,*** the elevated number of observed versus expected myocarditis and myopericarditis cases during the 0–7-day risk interval after receipt of the second Moderna vaccine dose is generally consistent with the findings from VSD. The level of certainty from the GRADE evidence assessment regarding potential harms after vaccination was type 2 (moderate certainty) for serious adverse events and type 1 (high certainty) for reactogenicity. GRADE was last completed for Moderna COVID-19 primary vaccination in December 2020 ( 1 ); since that time, additional data became available on all prespecified outcomes of interest, resulting in a higher level of certainty in the estimates for the benefit of vaccination in prevention of asymptomatic infection and death (the GRADE evidence profile is available at https://www.cdc.gov/vaccines/acip/recs/grade/bla-covid-19-moderna-vaccine.html). Overall, the benefits for the Moderna COVID-19 vaccine outweigh any observed vaccine-associated risks (Table 1) (Table 2). TABLE 2 Summary of the certainty of evidence of potential harms of Moderna COVID-19 vaccination — United States, February 2022 Characteristic Clinical trial evidence Observational evidence GRADE evidence certainty* No. of studies Relative risk (95% CI) No. of studies No. of cases per 1 million doses Potential harms, pooled data Reactogenicity 2 5.03 (4.65–5.45) 0 — † 1 Serious adverse events§ 2 0.92 (0.78–1.08) 0 —¶ 2 Potential harms by data source VSD Anaphylaxis, persons ≥18 yrs NA NA 1 5.1** 3 Myocarditis, sex and age group, yrs Men, 18–39 NA NA 1 65.7†† 3 Women, 18–39 NA NA 1 6.2†† 3 VAERS Myocarditis, sex and age group, yrs Men, 18–24 NA NA 1 40.0§§ 3 Women, 18–24 NA NA 1 5.5§§ Men, 25–29 NA NA 1 18.3¶¶ Women, 25–29 NA NA 1 5.8¶¶ Men, 30–39 NA NA 1 8.4*** Women, 30–39 NA NA 1 0.6*** Abbreviations: GRADE = Grading of Recommendations, Assessment, Development and Evaluation; NA = not applicable; RR = relative risk; VAERS = Vaccine Adverse Event Reporting System; VSD = Vaccine Safety Datalink. * GRADE evidence certainty is ranked as follows: 1 = high certainty, 2 = moderate certainty, 3 = low certainty, 4 = very low certainty. † Observational evidence did not include a measure of reactogenicity. § Considered a critical outcome in GRADE. https://www.cdc.gov/vaccines/acip/recs/grade/about-grade.html ¶ Observational evidence did not include an aggregate measure of serious adverse events. Data on specific serious adverse events identified through postauthorization safety surveillance were reviewed. Increased risk for myocarditis and anaphylaxis were observed in VAERS and VSD. ** Based on VSD chart reviewed cases of anaphylaxis, in persons aged ≥18 years, occurring in a 0–1-day risk interval after vaccination (RR = 5.1; 95% CI = 3.3–7.6). †† Based on VSD chart-reviewed cases of myocarditis and pericarditis that met CDC case definitions among persons aged 18–39 years after dose 2, occurring in a 0–7-day risk interval after vaccination. §§ Based on VAERS chart-reviewed cases of myocarditis that met CDC case definitions among men and women aged 18–24 years, days 0–7 after dose 2. ¶¶ Based on VAERS chart-reviewed cases of myocarditis that met CDC case definitions among men and women aged 25–29 years, days 0–7 after dose 2. *** Based on VAERS chart-reviewed cases of myocarditis that met CDC case definitions among men and women aged 30–39 years, days 0–7 after dose 2. Data reviewed within the EtR Framework support the use of the Moderna COVID-19 vaccine. The Work Group concluded that COVID-19 remains an important public health problem and that the desirable effects of disease prevention through vaccination with Moderna COVID-19 vaccine in persons aged ≥18 years are large and outweigh the potential harms. With 204 million doses of Moderna COVID-19 vaccine administered to date ( 2 ), the Work Group determined that the vaccine is acceptable to vaccine providers and that implementation of vaccination is feasible. The Work Group also acknowledged that vaccine-eligible persons aged ≥18 years probably considered the desirable effects of vaccination to be favorable compared with the undesirable effects; however, there is likely important variability in vaccine acceptance within this age group, especially among those who are currently unvaccinated. Despite having recommendations for the Moderna COVID-19 vaccine for >1 year, data indicate vaccine coverage varies by geography, race/ethnicity, sexual orientation, and gender identity ( 12 – 14 ). Because these disparities remained even after the Pfizer COVID-19 vaccine had received standard authorization, the Work Group concluded that changing from an interim to a standard ACIP recommendation alone for the Moderna COVID-19 vaccine would probably have minimal impact on health equity (the evidence used to inform the EtR is available at https://www.cdc.gov/vaccines/acip/recs/grade/bla-covid-19-moderna-etr.html). Interval Between Primary mRNA COVID-19 Vaccination Series Doses In addition to data presented to guide the recommendation for use of the Moderna COVID-19 vaccine, data were also presented to ACIP regarding the optimal interval between the first and second dose of a Moderna or Pfizer-BioNTech mRNA primary vaccination series. mRNA COVID-19 vaccines are safe and effective at the authorized interval between the first and second doses (4 weeks for Moderna vaccine; 3 weeks for Pfizer-BioNTech vaccine), but an extended interval might be considered for some populations. An elevated risk for myocarditis and myopericarditis among mRNA COVID-19 vaccine recipients has been observed, particularly in adolescent and young adult males ( 11 , 15 ). Several studies in adolescents and adults have indicated the small risk for myocarditis associated with mRNA COVID-19 vaccines might be reduced ( 16 ) and peak antibody responses and vaccine effectiveness might be increased ( 17 – 20 ) with an interval longer than 4 weeks between the 2 primary series doses. In a population-based cohort study in Ontario, Canada, rates of myocarditis among persons aged ≥18 years were lower with an extended interval (>4 to <8 weeks and ≥8 weeks) compared with the shorter interval (3–4 weeks) between the first and second doses of a primary series for both Moderna and Pfizer-BioNTech vaccines ( 16 ). In several studies, neutralizing antibody titers were higher after an extended interval between doses in a primary mRNA vaccine series (range = 6–14 weeks), compared with a standard interval of 3–4 weeks ( 17 – 20 ). Vaccine effectiveness against infection and hospitalization was higher with an extended (6–8-week) interval than with a standard (3–4-week) interval ( 19 ). ††† Based on this evidence presented to ACIP, CDC has provided guidance that an 8-week interval might be optimal for some adolescents and adults, especially for males aged 12–39 years. Additional primary series interval considerations are available at https://www.cdc.gov/vaccines/covid-19/clinical-considerations/covid-19-vaccines-us.html. After a year of use under an FDA-issued EUA and ACIP interim recommendation, the Moderna COVID-19 vaccine received full FDA approval and is recommended by ACIP for use in persons aged ≥18 years in the United States. Spikevax, the trade name of the Moderna COVID-19 vaccine, has the same formulation and can be used interchangeably with the Moderna COVID-19 vaccine used under EUA without presenting any safety or effectiveness concerns. ACIP considered new information beyond what was available at the time of the interim recommendation, including an additional 3 months of follow-up of the Phase III clinical trial participants, 26 observational vaccine effectiveness studies involving large populations of vaccinated persons, and two postauthorization safety monitoring systems with data from millions of vaccinated persons in the United States. The additional information increased certainty that the benefits of Moderna COVID-19 vaccine outweigh vaccine-associated risks. The Moderna COVID-19 vaccine continues to have FDA authorization and interim ACIP recommendations for a booster dose ( 21 ), as well as an additional dose in persons aged ≥18 years with moderate to severe immunocompromise ( 22 ). For an mRNA primary series, an 8-week interval between first and second doses might be optimal for some persons aged ≥12 years, especially males aged 12–39 years. Before vaccination, a fact sheet ( 23 ) or vaccine information sheet should be provided to recipients. Providers should counsel Moderna COVID-19 vaccine recipients about expected systemic and local reactogenicity. Additional clinical considerations for COVID-19 vaccine administration are available at https://www.cdc.gov/vaccines/covid-19/clinical-considerations/covid-19-vaccines-us.html Reporting of Vaccine Adverse Events Providers are required to report adverse events that occur after receipt of any COVID-19 vaccine to VAERS ( 23 ) (https://vaers.hhs.gov/index.html or 1–800–822–7967). Any person who administers or receives a COVID-19 vaccine is encouraged to report any clinically significant adverse event, regardless of whether it is clear that a vaccine caused the adverse event. In addition, all COVID-19 vaccine recipients are encouraged to enroll in v-safe, a CDC voluntary smartphone-based online tool that uses text messaging and online surveys to conduct periodic health check-ins after vaccination. CDC’s v-safe (https://www.cdc.gov/vsafe ) call center follows up on reports to v-safe that include possible medically significant health events to collect additional information for completion of a VAERS report. Summary What is already known about this topic? On January 31, 2022, the Food and Drug Administration (FDA) granted full approval to the Moderna COVID-19 vaccine for persons aged ≥18 years. What is added by this report? On February 4, 2022, after a systematic review of the evidence, the Advisory Committee on Immunization Practices issued a standard recommendation for use of the Moderna COVID-19 vaccine in persons aged ≥18 years. CDC provided guidance that an 8-week interval between primary series doses of mRNA vaccines might be optimal for some persons. What are the implications for public health practice? Use of the FDA-approved Moderna COVID-19 vaccine is recommended for persons aged ≥18 years; benefits of the prevention of infection and associated hospitalization or death outweigh vaccine-associated risks.
                Bookmark

                Author and article information

                Journal
                MMWR Morb Mortal Wkly Rep
                MMWR Morb Mortal Wkly Rep
                WR
                Morbidity and Mortality Weekly Report
                Centers for Disease Control and Prevention
                0149-2195
                1545-861X
                16 June 2023
                16 June 2023
                : 72
                : 24
                : 657-662
                Affiliations
                National Center for Immunization and Respiratory Diseases, CDC; Institute for Health Research, Kaiser Permanente Colorado, Denver, Colorado.
                Author notes
                Corresponding author: Danielle L. Moulia, wwe8@ 123456cdc.gov .
                Article
                mm7224a3
                10.15585/mmwr.mm7224a3
                10328466
                37319020
                18831d19-879a-4ad0-a9b5-fb46996f0ddf

                All material in the MMWR Series is in the public domain and may be used and reprinted without permission; citation as to source, however, is appreciated.

                History
                Categories
                Full Report

                Comments

                Comment on this article