Inviting an author to review:
Find an author and click ‘Invite to review selected article’ near their name.
Search for authorsSearch for similar articles
15
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: not found
      • Article: not found

      Bivalent omicron (BA.1) booster vaccination against SARS-CoV-2

      discussion
      a , b , a
      The Lancet. Infectious Diseases
      Elsevier Ltd.

      Read this article at

      ScienceOpenPublisherPMC
      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

          A highly virulent novel coronavirus known as SARS-CoV-2 emerged in 2019, posing a challenge to all humankind. 1 The unprecedentedly rapid development and deployment of effective vaccines substantially reduced the burden of COVID-19, and rapid and comprehensive vaccination programmes protected the majority of the population in high-income countries. Unfortunately, neither vaccination with first-generation vaccines based on the original Wuhan strain nor natural infection provides sterilising immunity against SARS-CoV-2. Despite the presence of proofreading-repair activity, the spontaneous mutation rate of SARS-CoV-2 amounts to around 1·3 × 10–6 per base per infection cycle. 2 As a result, many mutants have emerged and spread around the world since 2019. The most important representatives include alpha (B.1.1.7), beta (B.1.351), delta (B.1.617.2), gamma (P1), 3 and omicron (B.1.1.159) variants, along with omicron subvariants BA.1, BA.2, BA.4, and BA.5. 4 Omicron causes predominantly mild disease and is less likely to result in severe disease compared with the delta variant, but its high contagiousness raises serious concern. The protection rate against the omicron BA.1 variant after a previous SARS-CoV-2 infection is estimated at only 36·1%, although protection against the severe disease remains as high as 90·2%. 5 Several studies have found first-generation vaccines to be less effective against omicron than previous SARS-CoV-2 variants.6, 7 Omicron variants can effectively evade neutralising antibodies induced by both natural infection and vaccination in individuals who have received two or three doses of a vaccine. Therefore, the vaccination strategy should consider the emergence of new variants, which mitigates the development of new bivalent vaccines. BNT162b2 and mRNA-1273.214 are two bivalent mRNA vaccines that encode Wuhan-hu-1 and BA.1 spike (S) proteins. These vaccines were shown to elicit more effective neutralising antibodies response against both the original ancestral strain and the omicron variant.8, 9 First clinical trials of the vaccines showed that neutralising antibody response against the omicron variant was superior to that induced by the corresponding first-generation monovalent vaccine that encodes only the ancestRal Wuhan-Hu-1 S protein. In their Lancet Infectious Diseases publication, Hugo van der Kuy and colleagues 10 have reported on the immunogenicity and reactogenicity of these vaccines in individuals primed with adenovirus or first-generation mRNA-based vaccines that encode the Wuhan-Hu-1 spike protein. As a part of the open-label, multicentre, randomised controlled SWITCH ON trial, health-care workers aged 18–65 years were divided into four groups: (1) Ad26.COV2.S prime and BNT162b2 OMI BA.1 boost (Ad/P), (2) mRNA-based prime and BNT162b2 OMI BA.1 boost (mRNA/P), (3) Ad26.COV2.S prime and mRNA-1273.214 boost (Ad/M), and (4) mRNA-based prime and mRNA-1273.214 boost (mRNA/M). The booster vaccinations with either mRNA vaccine triggered a rapid recall of the humoral and cellular immune response within 7 days, regardless of the priming vaccine. The induced antibodies and T cells reacted with both the omicron BA.1 subvariant and the more antigenically distinct BA.5 omicron subvariant. However, these neutralising antibody concentrations were generally lower than those against ancestral SARS-CoV-2. 10 Hugo van der Kuy and colleagues' findings should be translated to other age groups (those younger than 18 years and those older than 65 years) and ethnic backgrounds cautiously due to the objective limitations of the trial. The role of previous infections might also have been underestimated and requires further investigation. A follow-up evaluation of the frequency of SARS-CoV-2 infection among study participants would help shed light on the effectiveness of the performed booster immunisation. Various vaccine platforms are used for priming and are likely to be used as boosters worldwide. Therefore, the effect of a different priming history needs to be considered, and the optimal prime-boost composition should be studied to choose the most efficient one in the future. Although the data presented by van der Kuy and colleagues showed no difference in the effectiveness of mRNA boosters following different priming regimes, 10 this might not be the case for other vaccines administered worldwide. Considering that the omicron variant mostly causes mild disease and is not expected to be the last SARS-CoV-2 variant, the question of whether further booster immunisation should be done or not remains unresolved. Although the paper by van der Kuy and colleagues provides some insights into mRNA vaccine boosters, we believe more studies are needed. © 2023 Flickr – Jernej Furman 2023 We declare no competing interests.

          Related collections

          Most cited references10

          • Record: found
          • Abstract: found
          • Article: not found

          Covid-19 Vaccine Effectiveness against the Omicron (B.1.1.529) Variant

          Background A rapid increase in coronavirus disease 2019 (Covid-19) cases due to the omicron (B.1.1.529) variant of severe acute respiratory syndrome coronavirus 2 in highly vaccinated populations has aroused concerns about the effectiveness of current vaccines. Methods We used a test-negative case–control design to estimate vaccine effectiveness against symptomatic disease caused by the omicron and delta (B.1.617.2) variants in England. Vaccine effectiveness was calculated after primary immunization with two doses of BNT162b2 (Pfizer–BioNTech), ChAdOx1 nCoV-19 (AstraZeneca), or mRNA-1273 (Moderna) vaccine and after a booster dose of BNT162b2, ChAdOx1 nCoV-19, or mRNA-1273. Results Between November 27, 2021, and January 12, 2022, a total of 886,774 eligible persons infected with the omicron variant, 204,154 eligible persons infected with the delta variant, and 1,572,621 eligible test-negative controls were identified. At all time points investigated and for all combinations of primary course and booster vaccines, vaccine effectiveness against symptomatic disease was higher for the delta variant than for the omicron variant. No effect against the omicron variant was noted from 20 weeks after two ChAdOx1 nCoV-19 doses, whereas vaccine effectiveness after two BNT162b2 doses was 65.5% (95% confidence interval [CI], 63.9 to 67.0) at 2 to 4 weeks, dropping to 8.8% (95% CI, 7.0 to 10.5) at 25 or more weeks. Among ChAdOx1 nCoV-19 primary course recipients, vaccine effectiveness increased to 62.4% (95% CI, 61.8 to 63.0) at 2 to 4 weeks after a BNT162b2 booster before decreasing to 39.6% (95% CI, 38.0 to 41.1) at 10 or more weeks. Among BNT162b2 primary course recipients, vaccine effectiveness increased to 67.2% (95% CI, 66.5 to 67.8) at 2 to 4 weeks after a BNT162b2 booster before declining to 45.7% (95% CI, 44.7 to 46.7) at 10 or more weeks. Vaccine effectiveness after a ChAdOx1 nCoV-19 primary course increased to 70.1% (95% CI, 69.5 to 70.7) at 2 to 4 weeks after an mRNA-1273 booster and decreased to 60.9% (95% CI, 59.7 to 62.1) at 5 to 9 weeks. After a BNT162b2 primary course, the mRNA-1273 booster increased vaccine effectiveness to 73.9% (95% CI, 73.1 to 74.6) at 2 to 4 weeks; vaccine effectiveness fell to 64.4% (95% CI, 62.6 to 66.1) at 5 to 9 weeks. Conclusions Primary immunization with two doses of ChAdOx1 nCoV-19 or BNT162b2 vaccine provided limited protection against symptomatic disease caused by the omicron variant. A BNT162b2 or mRNA-1273 booster after either the ChAdOx1 nCoV-19 or BNT162b2 primary course substantially increased protection, but that protection waned over time. (Funded by the U.K. Health Security Agency.)
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Neutralization Escape by SARS-CoV-2 Omicron Subvariants BA.2.12.1, BA.4, and BA.5

            To the Editor: In recent months, multiple lineages of the omicron (B.1.1.529) variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have emerged, 1 with subvariants BA.1 and BA.2 showing substantial escape from neutralizing antibodies. 2-5 Subvariant BA.2.12.1 is now the dominant strain in the United States, and BA.4 and BA.5 are dominant in South Africa (Figure 1A). Subvariants BA.4 and BA.5 have identical sequences of the spike protein. We evaluated neutralizing antibody titers against the reference WA1/2020 isolate of SARS-CoV-2 along with omicron subvariants BA.1, BA.2, BA.2.12.1, and BA.4 or BA.5 in 27 participants who had been vaccinated and boosted with messenger RNA vaccine BNT162b2 (Pfizer–BioNTech) and in 27 participants who had been infected with the BA.1 or BA.2 subvariant a median of 29 days earlier (range, 2 to 113) (Tables S1 and S2 in the Supplementary Appendix, available with the full text of this letter at NEJM.org). In the vaccine cohort, participants were excluded if they had a history of SARS-CoV-2 infection or a positive result on nucleocapsid serologic analysis or if they had received another vaccine against coronavirus disease 2019 (Covid-19) or an immunosuppressive medication. Six months after the initial two BNT162b2 immunizations, the median neutralizing antibody pseudovirus titer was 124 against WA1/2020 but less than 20 against all the tested omicron subvariants (Figure 1B). Two weeks after administration of the booster dose, the median neutralizing antibody titer increased substantially, to 5783 against the WA1/2020 isolate, 900 against the BA.1 subvariant, 829 against the BA.2 subvariant, 410 against the BA.2.12.1 subvariant, and 275 against the BA.4 or BA.5 subvariant. These data show that as compared with the response against the WA1/2020 isolate, the neutralizing antibody titer was lower by a factor of 6.4 against BA.1, by a factor of 7.0 against BA.2, by a factor of 14.1 against BA.2.12.1, and by a factor of 21.0 against BA.4 or BA.5. In addition, as compared with the median neutralizing antibody titer against the BA.1 subvariant, the median titer was lower by a factor of 2.2 against the BA.2.12.1 subvariant and by a factor of 3.3 against the BA.4 or BA.5 subvariant. Among the participants who had been infected with the BA.1 or BA.2 subvariant of omicron, all but one had been vaccinated against Covid-19. Because of the variation in sampling after the onset of infection, some samples may not reflect peak neutralizing antibody titers (Table S2). Among the participants with a history of Covid-19, the median neutralizing antibody titer was 11,050 against the WA1/2020 isolate, 1740 against the BA.1 subvariant, 1910 against the BA.2 subvariant, 1150 against the BA.2.12.1 subvariant, and 590 against the BA.4 or BA.5 subvariant (Figure 1C). These data show that as compared with the WA1/2020 isolate, the median neutralizing antibody titer was lower by a factor of 6.4 against BA.1, by a factor of 5.8 against BA.2, by a factor of 9.6 against BA.2.12.1, and by a factor of 18.7 against BA.4 or BA.5. In addition, as compared with the median titers against the BA.1 subvariant, the median titer was lower by a factor of 1.5 against the BA.2.12.1 subvariant and by a factor of 2.9 against the BA.4 or BA.5 subvariant. These data show that the BA.2.12.1, BA.4, and BA.5 subvariants substantially escape neutralizing antibodies induced by both vaccination and infection. Moreover, neutralizing antibody titers against the BA.4 or BA.5 subvariant and (to a lesser extent) against the BA.2.12.1 subvariant were lower than titers against the BA.1 and BA.2 subvariants, which suggests that the SARS-CoV-2 omicron variant has continued to evolve with increasing neutralization escape. These findings provide immunologic context for the current surges caused by the BA.2.12.1, BA.4, and BA.5 subvariants in populations with high frequencies of vaccination and BA.1 or BA.2 infection.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A Bivalent Omicron-Containing Booster Vaccine against Covid-19

              Abstract Background The safety and immunogenicity of the bivalent omicron-containing mRNA-1273.214 booster vaccine are not known. Methods In this ongoing, phase 2–3 study, we compared the 50-μg bivalent vaccine mRNA-1273.214 (25 μg each of ancestral Wuhan-Hu-1 and omicron B.1.1.529 [BA.1] spike messenger RNAs) with the previously authorized 50-μg mRNA-1273 booster. We administered mRNA-1273.214 or mRNA-1273 as a second booster in adults who had previously received a two-dose (100-μg) primary series and first booster (50-μg) dose of mRNA-1273 (≥3 months earlier). The primary objectives were to assess the safety, reactogenicity, and immunogenicity of mRNA-1273.214 at 28 days after the booster dose. Results Interim results are presented. Sequential groups of participants received 50 μg of mRNA-1273.214 (437 participants) or mRNA-1273 (377 participants) as a second booster dose. The median time between the first and second boosters was similar for mRNA-1273.214 (136 days) and mRNA-1273 (134 days). In participants with no previous severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the geometric mean titers of neutralizing antibodies against the omicron BA.1 variant were 2372.4 (95% confidence interval [CI], 2070.6 to 2718.2) after receipt of the mRNA-1273.214 booster and 1473.5 (95% CI, 1270.8 to 1708.4) after receipt of the mRNA-1273 booster. In addition, 50-μg mRNA-1273.214 and 50-μg mRNA-1273 elicited geometric mean titers of 727.4 (95% CI, 632.8 to 836.1) and 492.1 (95% CI, 431.1 to 561.9), respectively, against omicron BA.4 and BA.5 (BA.4/5), and the mRNA-1273.214 booster also elicited higher binding antibody responses against multiple other variants (alpha, beta, gamma, and delta) than the mRNA-1273 booster. Safety and reactogenicity were similar with the two booster vaccines. Vaccine effectiveness was not assessed in this study; in an exploratory analysis, SARS-CoV-2 infection occurred in 11 participants after the mRNA-1273.214 booster and in 9 participants after the mRNA-1273 booster. Conclusions The bivalent omicron-containing vaccine mRNA-1273.214 elicited neutralizing antibody responses against omicron that were superior to those with mRNA-1273, without evident safety concerns. (Funded by Moderna; ClinicalTrials.gov number, NCT04927065.)
                Bookmark

                Author and article information

                Journal
                Lancet Infect Dis
                Lancet Infect Dis
                The Lancet. Infectious Diseases
                Elsevier Ltd.
                1473-3099
                1474-4457
                21 April 2023
                21 April 2023
                Affiliations
                [a ]Smorodintsev Research Institute of Influenza, Saint Petersburg, Russia
                [b ]Institute of Biomedical Systems and Biotechnologies, Peter the Great St Petersburg Polytechnic University, Saint Petersburg, 195251, Russia
                Article
                S1473-3099(23)00189-5
                10.1016/S1473-3099(23)00189-5
                10119556
                c108e245-cc60-4686-80e1-4d94ff6b5575
                © 2023 Elsevier Ltd. All rights reserved.

                Since January 2020 Elsevier has created a COVID-19 resource centre with free information in English and Mandarin on the novel coronavirus COVID-19. The COVID-19 resource centre is hosted on Elsevier Connect, the company's public news and information website. Elsevier hereby grants permission to make all its COVID-19-related research that is available on the COVID-19 resource centre - including this research content - immediately available in PubMed Central and other publicly funded repositories, such as the WHO COVID database with rights for unrestricted research re-use and analyses in any form or by any means with acknowledgement of the original source. These permissions are granted for free by Elsevier for as long as the COVID-19 resource centre remains active.

                History
                Categories
                Comment

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article