A Bivalent Omicron-containing Booster Vaccine Against Covid-19 ============================================================== * Spyros Chalkias * Charles Harper * Keith Vrbicky * Stephen R. Walsh * Brandon Essink * Adam Brosz * Nichole McGhee * Joanne E. Tomassini * Xing Chen * Ying Chang * Andrea Sutherland * David C. Montefiori * Bethany Girard * Darin K. Edwards * Jing Feng * Honghong Zhou * Lindsey R. Baden * Jacqueline M. Miller * Rituparna Das ## ABSTRACT **Background** Updated vaccination strategies against acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern are needed. Interim results of the safety and immunogenicity of the bivalent omicron-containing mRNA-1273.214 booster candidate are presented. **Methods** In this ongoing, phase 2/3 trial, the 50-μg bivalent vaccine mRNA-1273.214 (25-μg each ancestral Wuhan-Hu-1 and omicron B.1.1.529 spike SARS-CoV-2 mRNAs) was compared to the authorized 50-μg mRNA-1273 booster in adults who previously received 2-dose primary series of 100-μg mRNA-1273 and a first booster dose of 50-μg mRNA-1273 at least 3 months prior. Primary objectives were safety and reactogenicity, and immunogenicity of 50-μg mRNA-1273.214 compared with 50-μg mRNA-1273. Immunogenicity data 28 days after the booster dose are presented. **Results** Four hundred thirty-seven and 377 participants received 50-μg of mRNA-1273.214, or mRNA-1273, respectively. Median time between first and second booster doses of mRNA-1273.214 and mRNA-1273 were similar (136 and 134 days, respectively). In participants with no prior SARS-CoV-2 infection, observed omicron neutralizing antibody geometric mean titers (GMTs [95% confidence interval]) after the mRNA-1273.214 and mRNA-1273 booster doses, were 2372.4 (2070.6−2718.2) and 1473.5 (1270.8−1708.4) respectively and the model-based GMT ratio (97.5% confidence interval) was 1.75 (1.49−2.04). All pre-specified non-inferiority (ancestral SARS-CoV-2 with D614G mutation [D614G] GMT ratio; ancestral SARS-CoV-2 [D614G] and omicron seroresponse rates difference) and superiority primary objectives (omicron GMT ratio) for mRNA-1273.214 compared to mRNA-1273 were met. Additionally, mRNA-1273.214 50-μg induced a potent neutralizing antibody response against omicron subvariants BA.4/BA.5 and higher binding antibody responses against alpha, beta, gamma, delta and omicron variants. Safety and reactogenicity profiles were similar and well-tolerated for both vaccines groups. **Conclusion** The bivalent vaccine mRNA-1273.214 50-μg was well-tolerated and elicited a superior neutralizing antibody response against omicron, compared to mRNA-1273 50-μg, and a non-inferior neutralizing antibody response against the ancestral SARS-CoV-2 (D614G), 28 days after immunization, creating a new tool as we respond to emerging SARS-CoV-2 variants. ## INTRODUCTION Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines are safe and effective against coronavirus disease 2019 (Covid-19). The mRNA-1273 vaccine encoding the Wuhan-Hu-1 (ancestral) spike protein of SARS-CoV-2 demonstrated an acceptable safety profile and efficacy of 93.2% against Covid-19 after a median of 5.3 months following the 2-dose 100-μg primary series immunization in the coronavirus efficacy (COVE) trial.1,2 Soon after the beginning of the SARS-CoV-2 pandemic, several variants with spike protein mutations that can confer immunologic escape or enhanced transmissability have emerged, such as the beta (B.1.351) and delta (B.1.617.2) variants. In 2022, omicron (B.1.1.529) and omicron subvariants (BA.2, BA.2.12.1, BA.4, BA.5), the most antigenically-divergent variants to date, outcompeted other variants in the setting of a background of substantial pre-existing population immunity from vaccination, infection or both.3-7 Omicron and omicron subvariants continue to cause significant morbidity and mortality and re-infection with omicron or a subvariant is possible, especially in unvaccinated individuals.8-10 Immunization with an mRNA-1273 50-μg booster dose increases neutralizing antibody responses against variants and booster doses enhance vaccine effectiveness against Covid-19.11-16 Nonetheless, effectiveness of the currently authorized vaccines against omicron is decreased compared to other variants17-20 and consequently, a second booster dose was authorized in the US for immunocompromised individuals and adults >50 years of age in the midst of omicron waves.21,22 Therefore, SARS-CoV-2 candidate vaccines targeting omicron that can induce more potent and durable as well as broader immune responses are important to enhance protection. We have previously evaluated a modified, bivalent booster vaccine approach23 with a vaccine that contained equal amounts of mRNA that encode the spike protein of the ancestral SARS-CoV-2 and of the beta variant. Immunogenicity results of the beta-containing bivalent vaccine demonstrated increased and more durable neutralizing antibody responses against beta and multiple variants, even against variants (delta, omicron) not contained in the vaccine, compared to mRNA-1273.23 Here we present the interim analysis results 28 days after a 50-μg booster dose of an omicron-containing bivalent candidate, mRNA-1273.214, from an ongoing safety and immunogenicity phase 2/3 study. ## METHODS ### Trial Oversight and Participants This is an open-label, ongoing Phase 2/3 study ([NCT04927065](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT04927065&atom=%2Fmedrxiv%2Fearly%2F2022%2F06%2F25%2F2022.06.24.22276703.atom)) to evaluate the immunogenicity, safety, and reactogenicity of mRNA-1273.214, a bivalent booster vaccine, as compared to the currently-authorized mRNA-1273 vaccine in adult participants. Adults who had previously received a 2-dose primary series of 100-μg mRNA-1273 and a first booster dose (50 μg) in the COVE1,2 or under US emergency use authorization (EUA), were sequentially enrolled. The first cohort received a single second booster of 50-μg of mRNA-1273 (part F, cohort 2) and the second cohort received 50-μg of the bivalent mRNA-1273.214 vaccine which contained equal amounts of ancestral SARS-CoV-2 and omicron spike protein mRNAs (part G). The mRNA-1273 group serves as a non-contemporaneous within-study comparator. Adults with a known history of SARS-CoV-2 infection within 3 months from screening were excluded from the study (inclusion/exclusion criteria are provided in the supplement). The trial is being conducted across 23 US sites, in accordance with the International Council for Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use, Good Clinical Practice guidelines. The central institutional review board approved the protocol and consent forms. All participants provided written informed consent. ### Trial vaccine The mRNA-1273.214 50-μg vaccine contains equal amounts of mRNAs (25 μg of each mRNA sequence) that encode the prefusion stabilized spike glycoproteins of the ancestral SARS-CoV-2 (Wuhan-Hu-1) and the omicron variant (B.1.1.529 [BA.1]). The mRNA-1273 50-μg vaccine (Moderna Covid-19 Vaccine) contains only the mRNA sequence encoding the spike glycoprotein of the ancestral SARS-CoV-2. In both vaccines, mRNAs are encapsulated in lipid nanoparticles (LNPs). The booster doses of mRNA-1273.214 and mRNA-1273 were administered intramuscular at doses of 50-μg of mRNA in a 0.5 mL volume. ### Safety Assessment The primary safety objective was to evaluate the safety and reactogenicity of 50-μg mRNA-1273.214 and of 50-μg mRNA-1273 when administered as a second booster dose (Table S1 and supplementary methods). Reactogenicity included solicited local and systemic adverse reactions (ARs) that occurred ≤7 days after the booster dose as recorded daily by participants. Unsolicited adverse events (AEs) were recorded by study sites for 28 days post-booster administration. Serious adverse events (SAEs), AEs leading to discontinuation from study vaccine and/or participation, medically-attended AEs (MAAEs) and AEs of special interest (AESIs) are to be recorded by the study sites from day 1 through the entire study period (∼12 months). ### Immunogenicity assessment The pre-specified primary immunogenicity objectives were to demonstrate 1) non-inferior neutralizing antibody responses (based on geometric mean titer [GMT] ratio and seroresponse rate [SRR] difference) or superior neutralizing antibody responses (GMT ratio) against omicron, and 2) non-inferior neutralizing antibody responses (based on GMT ratio) against the ancestral SARS-CoV-2 with the D614G mutation (ancestral SARS-CoV-2 [D614G]), 28 days after the mRNA 1273.214 50-μg second booster dose (day 29) compared with the mRNA-1273 50-μg second booster dose (Table S1 and statistical methods). The pre-specified key secondary objective was to demonstrate non-inferiority (based on SRR difference) against the ancestral SARS-CoV-2 (D614G) 28 days after the mRNA-1273.214 50-μg second booster dose compared with the mRNA-1273 50-μg second booster dose. Neutralizing antibody geometric mean titers at inhibitory dilutions 50% (ID50) were assessed in validated SARS-CoV-2 spike-pseudotyped lentivirus neutralization assays. Titers were generated against pseudoviruses containing the SARS-CoV-2 full-length spike proteins for the ancestral SARS-CoV-2 (D614G) or the omicron (B.1.1.529 [BA.1]) variant and against a pseudovirus containing full-length spike protein for omicron subvariants BA.4 and BA.5 (referred to as BA.4/BA.5 because of identical spike sequences between BA.4 and BA.5) were assessed using a research-grade pseudovirus assay. Geometric mean (GM) levels were also assessed in an anti-spike protein binding IgG antibody (bAb) assay (Meso Scale Discovery [MSD]) against the ancestral SARS-CoV-2 (D614G), gamma (P.1), alpha (B.1.1.7), beta (B.1.351), delta [B.1.617.2; AY.4], and omicron (B.1.1.529; BA.1) variants (PPD, part of Thermo Fisher Scientific Vaccines Laboratory Services, Richmond, Virginia). Immunogenicity assays are further described in the supplement. An exploratory objective was to assess incidences of symptomatic and asymptomatic SARS-CoV-2 infection in both groups (Table S1 and Supplementary Methods). Symptomatic infection was evaluated using the primary case definition in the COVE study1,2 as well as a secondary case definition based on the Centers for Disease Control and Prevention (CDC) criteria.24 Asymptomatic SARS-CoV-2 infection was defined as a positive reverse-transcriptase polymerase chain reaction (RT-PCR) test or a positive serologic test for anti-nucleocapsid antibody after a negative test at the time of enrollment, in the absence of symptoms. ### Statistical analysis Safety was evaluated in the safety set consisting of all participants who received the 50-μg mRNA-1273.214 or 50-μg mRNA-1273 second booster doses. Solicited adverse reactions were assessed in the solicited safety set (Supplementary Methods). The numbers and percentages of participants with any solicited local and systemic adverse reactions occurring within 7 days post-boost are provided. Unsolicited AEs, SAEs, severe AEs, MAAEs, AESIs and AEs leading to study discontinuation are also summarized. The primary immunogenicity objectives were assessed in the per-protocol set for immunogenicity–SARS-CoV-2-negative set, also referred to as primary analysis set, consisting of all participants who received the booster dose, had antibody data available at the pre-booster and day 29 visits with no major protocol deviations and had no evidence of SARS-CoV-2 infection pre-booster. For the pre-specified primary objectives, a hierarchical testing was pre-specified. Superiority of the antibody response of the second booster dose of 50-μg mRNA-1273.214 compared to the second booster dose of 50-μg mRNA-1273 based on the GMR against omicron was to be tested after the following were demonstrated: 1) non-inferiority of the antibody response of the second booster dose of 50-μg mRNA-1273.214 compared with the second booster dose of 50-μg mRNA-1273 based on the GMR against omicron; 2) non-inferiority of the antibody response of the second dose of 50-μg mRNA-1273.214 compared to the second booster dose of 50-μg mRNA-1273 against omicron based on the difference in SRR; and 3) non-inferiority of the antibody response of the second booster dose of 50-μg mRNA-1273.214 compared to the second booster dose of 50-μg mRNA-1273 based on GMR against the ancestral SARS-CoV-2 (D614G) (Fig. S1 and supplementary methods). The key secondary endpoint of non-inferiority of the antibody response of the second booster dose of 50-μg mRNA-1273.214 compared to the second booster dose of 50-μg of mRNA-1273 against the ancestral SARS-CoV-2 (D614G) based on the difference in SRR, was to be tested if all endpoints for the primary objective were met. The immunogenicity objectives were to be tested 28 days after the booster dose (day 29) and 90 days after the booster dose (day 91) with a two-sided alpha of 0.025 respectively allocated at each one of the two time points. Non-inferiority is considered met when the lower bound of the 97.5% confidence interval (CI) of GMR is ≥0.67 and the SRR-difference is >-10%. Superiority is considered met when the lower bound of the 97.5% CI of GMR is >1. Superiority of the mRNA-1273.214 antibody response against omicron, compared to mRNA-1273, is considered demonstrated if superiority based on GMR is met at days 29 or 91. The day 29 interim analysis results are presented here. The GMTs (95% CI) for mRNA-1273.214 and mRNA-1273 booster doses are calculated by using t-distribution of log-transformed antibody titers. An analysis of covariance (ANCOVA) model was performed to assess the difference in antibody responses between mRNA-1273.214 and mRNA-1273 groups, with antibody titers post-booster as a dependent variable, and a group variable (mRNA-1273.214 and mRNA-1273) as the fixed effect, adjusting for age groups (<65, ≥65 years) and pre-booster antibody titers. The GMTs (95% CI) estimated by the geometric least square mean (GLSM) from the model for each group and the GMR (mRNA-1273.214 compared with mRNA-1273) estimated by the ratio of GLSM from the model (97.5% CIs) are provided. The 97.5% CI for GMR was used to assess the differences in antibody responses between groups. Seroresponse at a participant level is defined as a change from 1). View this table: [Table 2.](http://medrxiv.org/content/early/2022/06/25/2022.06.24.22276703/T2) Table 2. Primary Immunogenicity Analysis of Ancestral SARS-CoV-2 (D614G) and Omicron after 50-μg of mRNA-1273.214 and mRNA-1273 Administered as Second Booster Doses The seroresponse rates (SRRs) (95% CI) against ancestral SARS-COV-2 (D614G) were 100% (98.9−100% and 98.6−100%) 28 days after the mRNA-1273.214 and mRNA-1273 booster doses, respectively, with an SRR difference of 0 meeting the non-inferiority criterion (lower bound of CI >-10%). The omicron SRRs were 100% (98.9−100%) for mRNA-1273.214 and 99.2% (97.2−99.9%) for mRNA-1273 28 days following the booster doses, and the estimated SRR difference (97.5% CI) was 1.5% (−1.1−4.0) meeting the non-inferiority criterion (lower bound of CI >-10%). Therefore, all primary and key secondary immunogenicity endpoints were met based on the pre-specified testing sequence (Fig. S1). All immunogenicity endpoints were also met based on the analysis which included all participants, with and without evidence of SARS-CoV-2 infection pre-booster (Table S5). To evaluate whether the antibody responses were consistent between participants with and without prior SARS-CoV-2 infections, a pre-planned subgroup analysis was performed (Fig. 3 and Fig. S2; Tables S6-S7). In those with evidence of prior SARS-CoV-2 infection, observed GMTs against the ancestral SARS-CoV-2 (D614G) were 9509.7 (7345.9-12310.9) and 7003.5 (5592.6-8770.4) and estimated GMTs were 9891.5 (8732.2-11204.8) and 7776.5 (6813.0-8876.3) for the mRNA-1273.214 and mRNA-1273 groups, respectively with a GMR (95% CI) of 1.27 (1.07-1.51). Against omicron, observed GMTs (95% CI) were 7676.2 (5618.2-10488.1) and 3885.6 (2877.8-5246.4) and estimated GMTs were 7669.2 (6470.7-9089.6) and 4041.5 (3375.1-4839.5) for mRNA-1273.214 and mRNA-1273 with a GMR (95% CI) of 1.90 (1.50-2.40). For both groups, the SRRs were 100% for the ancestral SARS-CoV-2 (D614G) and omicron, and the SRR difference was 0. ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/06/25/2022.06.24.22276703/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2022/06/25/2022.06.24.22276703/F3) Figure 3. Observed Neutralizing Antibody Titers Against Ancestral SARS-CoV-2 (D614G) and Omicron after 50-μg of mRNA-1273.214 and mRNA-1273 Administered as Second Booster Doses. GM = geometric mean; CI = confidence interval. Pseudovirus neutralizing antibody geometric mean titers are from participants with non-missing data at the timepoint. Nine participants in the mRNA-1273 arm were missing pre-booster SARS-CoV-2 status. Antibody values reported as below the lower limit of quantification (LLOQ: 18.5 for ancestral SARS-CoV-2 (D614G); 19.9 for omicron) were replaced by 0.5 x LLOQ. Values greater than the upper limit of quantification (ULOQ: 45,118 for ancestral SARS-CoV-2 (D614G); 15502.7 for omicron) were replaced by the ULOQ if actual values are not available. 95% CIs were calculated based on the t-distribution of the log-transformed values or the difference in the log-transformed values for GM value and GM fold-rise, respectively, then back transformed to the original scale for presentation. Data for observed nAb GMTs by prior infection status are provided in Table S7. Given the emergence of omicron subvariants, we also assessed the neutralizing antibody response 28 days after the 50-μg mRNA-1273.214 booster dose against the omicron BA.4 and BA.5 subvariants which have additional spike mutations compared to BA.1. The observed GMTs, 28 days following the booster dose, were 727.4 (632.8-836.1) and the fold-rise in titers, compared to the pre-booster level of 115.6 (98.5-135.6) was 6.3 (5.7-6.9) in participants without evidence of prior SARS-CoV-2 infection pre-booster (Fig. S3 and Table S8). In the subgroup of participants with prior SARS-CoV-2 infection, the GMTs against the omicron subvariants was 2337.4 (1825.5-2992.9) and the fold-rise from the pre-booster level of 719.5 (531.6, 973.9) was 3.2 (2.8-3.8). In all participants regardless of prior SARS-CoV-2 infection, GMTs were 172.7 (147.5-202.3) pre-booster and rose to 940.6 (826.3-1070.6) with a fold-rise of 5.4 (5.0-5.9). Differences between the two vaccines based on binding antibody titers were also assessed for ancestral SARS-CoV-2 and omicron, as well as other variants of concern. In participants without evidence of prior SARS-CoV-2 infection, the binding antibody GMTs (95% CI) were higher (nominal alpha of 0.05) following mRNA-1273.214 than mRNA-1273 booster doses with GMRs (95% CI) of 1.19 (1.10-1.28) for alpha, 1.15 (1.07-1.25) for beta, 1.19 (1.10-1.28) for gamma and 1.11 (1.03-1.19) for delta (Fig. S4 and Table S9). Similar GMRs between the mRNA-1273.214 and mRNA-1273 vaccine groups were observed in the analysis of all participants regardless of prior SARS-CoV-2 infection (Table S10). The observed binding antibody titers for the ancestral SARS-CoV-2 and omicron are summarized in Table S11. ### Incidence of SARS-CoV-2 infections In participants with no prior evidence of SARS-SoV-2 infection, starting 14 days after vaccination, SARS-CoV-2 infections, regardless of the presence of symptoms, occurred in 11 (3.2%) and 5 (1.9%) participants in the mRNA-1273.214 and mRNA-1273 groups, respectively. Of those 6 (1.8%) and 4 (1.5%) infections were asymptomatic, and there were 5 (1.5%) and 1 (0.4%) Covid-19 events (symptomatic infections) in the mRNA-1273.214 and mRNA-1273 groups, respectively. There were no emergency room visits or hospitalizations due to Covid-19. ## DISCUSSION Our results indicate that a 50-μg dose of the bivalent omicron-containing mRNA-1273.214 vaccine had a safety and reactogenicity profile that was similar to the prototype mRNA-1273 (50-μg) booster vaccine when administered as second booster doses. The frequency of the adverse reactions after a second booster dose of 50-μg mRNA-1273.214 was similar to or lower than that of a first booster dose of 50-μg mRNA-1273 and of the second dose of the 100-μg mRNA-1273 primary series as previously reported.11,15 Overall, the reactogenicity profile of mRNA-1273.214 is reassuring, with three prior doses administered and with an interval of at least 3 months from the prior dose. We have also previously evaluated the safety and reactogenicity of another bivalent candidate, mRNA-1273.211, which had similar safety and reactogenicity to the mRNA-1273.214 vaccine, with a reassuring safety profile through 6 months post-vaccination.23 Neutralizing antibody responses have been used as a surrogate to assess effectiveness of Covid-19 vaccines.25,26 The 50-μg mRNA-1273.214 booster vaccine elicited a superior neutralizing antibody response against omicron, compared to 50-μg mRNA-1273, 28 days after the booster dose. The magnitude of the difference in the omicron neutralizing antibody response between the two vaccines (GMR 1.75 [97.5% CI, 1.49−2.04]), exceeds the recommended superiority criteria.27 The neutralizing antibody response against the ancestral SARS-CoV-2 (D614G) was also higher with mRNA-1273.214, compared to mRNA-1273, indicating no decrement in the ancestral SARS-CoV-2 (D614G) antibody responses after the booster dose of the bivalent vaccine. Neutralizing antibody responses were consistently higher with mRNA-1273.214, compared to mRNA-1273, in participants with and without evidence of prior SARS-CoV-2 infection. Given the epidemiological data suggestive of decreasing vaccine effectiveness against infection with omicron and that break-through infections can occur in vaccinated individuals, including those with prior SARS-CoV-2 infection,8-10,17,28 it is important to be able to boost immune responses in this group. The mechanisms of an increased antibody response with vaccines that contain both the ancestral SARS-CoV-2-spike protein sequence as well as that of an antigenically-divergent variant could include the generation of new memory immune responses but are yet to be elucidated.29 The ability to cross-react with multiple variants is highly desirable given the continuous evolution of SARS-CoV-2 and the emergence of escape variants. The results of the binding antibody responses indicate higher responses with the bivalent omicron-containing vaccine, compared to the prototype mRNA-1273, even when the variants are not contained in the vaccine. We had previously reported that a bivalent booster vaccine containing the ancestral SARS-CoV-2 and an antigenically-divergent variant can induce antibodies which cross-neutralize and cross-react with multiple variants and consistently elicit higher antibody responses compared to mRNA-1273.23 mRNA-1273.214 elicited potent neutralizing antibodies responses against the omicron BA.4 and BA.5 subvariants and the increase (fold-rise) in neutralizing antibody titers was consistent with the increase in titers against omicron BA.1, regardless of prior SARS-CoV-2 infection. In addition, the longevity of the neutralizing antibody response is important and we previously observed a more durable response against multiple variants, 6 months after immunization with the beta-containing bivalent vaccine, compared to mRNA-1273.23 We will continue to monitor the persistence of the antibody responses elicited by mRNA-1273.214 in the ongoing clinical study. There are limitations to the study, including that it was not randomized. While the enrollment dates of the booster groups were within weeks of each other, representing similar epidemiological environments of circulating variants, we did not ascertain variant sequences. Additionally, the temporal distances between primary vaccination and first and second booster doses were similar between the two groups, despite the sequential study design. The study only assessed humoral immune responses; thus, future work is needed to characterize the potential contribution of cellular responses to protection. We present data from day 28 post-boost given the public health importance of these data; longer term follow-up is underway to characterize the antibody persistence and, although the study was not designed to evaluate vaccine effectiveness, to describe infection rates post-boost. In conclusion, the omicron-containing bivalent vaccine mRNA-1273.214 had a safety and reactogenicity profile similar to that of the current booster vaccine mRNA-1273 when administered at the 50-μg dose. mRNA-1273.214 elicited a superior neutralizing antibody response against omicron, compared to mRNA-1273, and potent neutralizing antibody responses against the BA.4 and BA.5 omicron subvariants 28 days after immunization. Antibody responses were also higher against the ancestral SARS-CoV-2 (D614G) and multiple additional variants. These results are consistent with the evaluation of our first, beta-containing, bivalent vaccine which induced enhanced and durable antibody responses against multiple SARS-CoV-2 variants23 and bivalent vaccines can be a new tool as we respond to emerging variants. ## Supporting information Supplementary Appendix [[supplements/276703_file02.pdf]](pending:yes) CONSORT Checklist [[supplements/276703_file03.pdf]](pending:yes) ## Data Availability As the trial is ongoing, access to patient-level data and supporting clinical documents with qualified external researchers may be available upon request and subject to review once the trial is complete. ## AUTHOR CONTRIBUTIONS SC, JF, JMM, RD and HZ contributed to the design of the study and oversight. SC, SRW, AB, CH, and NMcG contributed to data collection. BG and DCM were responsible for immunogenicity assays. SC, JF, HZ, RD, JMM, LRB and JET contributed to data analysis and/or interpretation of the data. SC, JF, LRB and JET contributed to drafting the manuscript. All authors critically reviewed and provided input to manuscript drafts and approved the final version for submission to the journal. ## DECLARATION OF INTEREST SRW has conducted clinical trials funded by NIAID/NIH, Moderna, Inc., Janssen Vaccines, and Sanofi Pasteur; BE, AB and KV have nothing to disclose; DCM reports funding from Moderna, Inc. for pseudovirus neutralization assays performed in the study; LRB is a co-primary principal investigator of the COVE trial funded by NIAID and conducted in conjunction with Moderna, Inc. SC, NMcG, XC, YC, AS, BG, DKE, JF, HZ, JMM and RD are employees of Moderna, Inc. and may hold stock/stock options in the company. JET is a Moderna consultant. ## FUNDING This study was funded by Moderna, Inc., Cambridge, Massachusetts, USA. ## DATA SHARING STATEMENT As the trial is ongoing, access to patient-level data and supporting clinical documents with qualified external researchers may be available upon request and subject to review once the trial is complete. ## ACKNOWLEGEMENTS We thank the participants in the trial and the members of the mRNA-1273 trial team (listed in the Supplementary Appendix) for their dedication and contributions to the trial, the Immune Assay Team at Duke University Medical Center, Durham, NC for PsVNA analyses, and Frank J. Dutko, Ph.D., (Moderna consultant) for writing and editorial support. * Received June 24, 2022. * Revision received June 24, 2022. * Accepted June 25, 2022. * © 2022, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial 4.0 International), CC BY-NC 4.0, as described at [http://creativecommons.org/licenses/by-nc/4.0/](http://creativecommons.org/licenses/by-nc/4.0/) ## References 1. 1.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;385:1774–85. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F06%2F25%2F2022.06.24.22276703.atom) 2. 2.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;384:403–16. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2035389&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F06%2F25%2F2022.06.24.22276703.atom) 3. 3.Cao Y, Yisimayi A, Jian F, et al. BA.2.12.1, BA.4 and BA.5 escape antibodies elicited by Omicron infection. bioRxiv 2022. (Accessed June 24, 2022, at [https://doi.org/10.1101/2022.04.30.489997](https://doi.org/10.1101/2022.04.30.489997) 4. 4.CoVariants. Overview of Variants in Countries. 2022. (Accessed June 24, 2022, at [https://covariants.org/per-country](https://covariants.org/per-country).) 5. 5.CoVariants. Overview of Variants/Mutations. 2022. (Accessed June 24, 2022, at [https://covariants.org/variants](https://covariants.org/variants).) 6. 6.Centers for Disease Control and Prevention. COVID Data Tracker: Variant Proportions. 2022. (Accessed June 24, 2022, at [https://covid.cdc.gov/covid-data-tracker/#variant-proportions](https://covid.cdc.gov/covid-data-tracker/#variant-proportions).) 7. 7.Investigation of SARS-CoV-2 variants: technical briefings. 2022. (Accessed June 24, 2022, at [https://www.gov.uk/government/publications/investigation-of-sars-cov-2-variants-technical-briefings](https://www.gov.uk/government/publications/investigation-of-sars-cov-2-variants-technical-briefings).) 8. 8.Pulliam JRC, van Schalkwyk C, Govender N, et al. Increased risk of SARS-CoV-2 reinfection associated with emergence of Omicron in South Africa. Science 2022; (Accessed June 24, 2022, at [https://www.science.org/doi/10.1126/science.abn4947](https://www.science.org/doi/10.1126/science.abn4947).) 9. 9.Suryawanshi RK, Chen IP, Ma T, et al. Limited cross-variant immunity from SARS-CoV-2 Omicron without vaccination. Nature 2022. (Accessed June 24, 2022, at [https://www.nature.com/articles/s41586-022-04865-0](https://www.nature.com/articles/s41586-022-04865-0).) 10. 10.Tang J, Novak T, Hecker J, et al. Cross-reactive immunity against the SARS-CoV-2 Omicron variant is low in pediatric patients with prior COVID-19 or MIS-C. Nat Commun 2022;13:2979; (Accessed June 24, 2022, at [https://doi.org/10.1038/s41467-022-30649-1](https://doi.org/10.1038/s41467-022-30649-1).) 11. 11.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;385:2485–7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1101/2021.09.17.21263624&link_type=DOI) 12. 12.Girard B, Tomassini JE, Deng W, et al. mRNA-1273 Vaccine-elicited Neutralization of SARS-CoV-2 Omicron in Adolescents and Children. medRxiv 2022; (Accessed June 24, 2022, at [https://doi.org/10.1101/2022.01.24.22269666](https://doi.org/10.1101/2022.01.24.22269666).) 13. 13.Wu K, Werner AP, Koch M, et al. Serum Neutralizing Activity Elicited by mRNA-1273 Vaccine. N Engl J Med 2021;384:1468–70. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMc2102179&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33730471&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F06%2F25%2F2022.06.24.22276703.atom) 14. 14.Pegu A, O’Connell SE, Schmidt SD, et al. Durability of mRNA-1273 vaccine-induced antibodies against SARS-CoV-2 variants. Science 2021;373:1372–7. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F06%2F25%2F2022.06.24.22276703.atom) 15. 15.Chu L, Vrbicky K, Montefiori D, et al. Immune response to SARS-CoV-2 after a booster of mRNA-1273: an open-label phase 2 trial. Nature Medicine 2022;28:1042–9. 16. 16.Pajon R, Doria-Rose NA, Shen X, et al. SARS-CoV-2 Omicron Variant Neutralization after mRNA-1273 Booster Vaccination. N Engl J Med 2022;386:1088–91. 17. 17.Tseng HF, Ackerson BK, Luo Y, et al. Effectiveness of mRNA-1273 against SARS-CoV-2 omicron and delta variants. Nat Med 2022;28:1063–71. 18. 18.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;71:139–45. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.15585/mmwr.mm7104e3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=35085224&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F06%2F25%2F2022.06.24.22276703.atom) 19. 19.Prasad N, Derado G, Nanduri SA, et al. Effectiveness of a COVID-19 Additional Primary or Booster Vaccine Dose in Preventing SARS-CoV-2 Infection Among Nursing Home Residents During Widespread Circulation of the Omicron Variant - United States, February 14-March 27, 2022. MMWR Morb Mortal Wkly Rep 2022;71:633–7. 20. 20.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;71:255–63. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.15585/mmwr.mm7107e2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=35176007&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F06%2F25%2F2022.06.24.22276703.atom) 21. 21.Centers for Disease Control and Prevention. COVID-19 Vaccines for People who are Moderately or Severely Immunocompromised. (Accessed June 24, 2022, at [https://www.cdc.gov/coronavirus/2019-ncov/vaccines/recommendations/immuno.html?s_cid=11710:immunocompromised%20covid%20booster%20dose:sem.ga:p:RG:GM:gen:PTN.Grants:FY22](https://www.cdc.gov/coronavirus/2019-ncov/vaccines/recommendations/immuno.html?s_cid=11710:immunocompromised%20covid%20booster%20dose:sem.ga:p:RG:GM:gen:PTN.Grants:FY22).) 22. 22.Centers for Disease Control and Prevention. Stay Up to Date with Your COVID-19 Vaccines. 2022. (Accessed June 24, 2022, at [https://www.cdc.gov/coronavirus/2019-ncov/vaccines/stay-up-to-date.html](https://www.cdc.gov/coronavirus/2019-ncov/vaccines/stay-up-to-date.html).) 23. 23.Chalkias S, Eder F, Essink B, et al. Safety, Immunogenicity and Antibody Persistence of a Bivalent Beta-Containing Booster Vaccine. Research Square 2022. (Accessed June 24, 2022, at [https://doi.org/10.21203/rs.3.rs-1555201/v1](https://doi.org/10.21203/rs.3.rs-1555201/v1) 24. 24.Centers for Disease Control and Prevention. Symptoms of COVID-19. (Accessed June, 24, 2022, at [https://www.cdc.gov/coronavirus/2019-ncov/symptoms-testing/symptoms.html](https://www.cdc.gov/coronavirus/2019-ncov/symptoms-testing/symptoms.html).) 25. 25.Gilbert PB, Montefiori DC, McDermott AB, et al. Immune correlates analysis of the mRNA-1273 COVID-19 vaccine efficacy clinical trial. Science 2022;375:43–50. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F06%2F25%2F2022.06.24.22276703.atom) 26. 26.Khoury DS, Cromer D, Reynaldi A, et al. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med 2021;27:1205–11. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-021-01377-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F06%2F25%2F2022.06.24.22276703.atom) 27. 27.U.S. Food and Drug Administration. Emergency Use Authorization for Vaccines to Prevent COVID-19, Guidance for Industry 2022. (Accessed June, 24, 2022, at [https://www.fda.gov/media/142749/download](https://www.fda.gov/media/142749/download).) 28. 28.Carazo S, Skowronski DM, Brisson M, et al. Protection against Omicron re-infection conferred by prior heterologous SARS-CoV-2 infection, with and without mRNA vaccination. medRxiv 2022. (Accessed June, 24, 2022, at [https://doi.org/10.1101/2022.04.29.22274455](https://doi.org/10.1101/2022.04.29.22274455).) 29. 29.Gaebler C, Wang Z, Lorenzi JCC, et al. Evolution of antibody immunity to SARS-CoV-2. Nature 2021;591:639–44. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F06%2F25%2F2022.06.24.22276703.atom)