Safety and immunogenicity of a phase 1/2 randomized clinical trial of a quadrivalent, mRNA-based seasonal influenza vaccine (mRNA-1010) in healthy adults: interim analysis

Ivan T Lee, Raffael Nachbagauer, David Ensz, Howard Schwartz, Lizbeth Carmona, Kristi Schaefers, Andrei Avanesov, Daniel Stadlbauer, Carole Henry, Ren Chen, Wenmei Huang, Daniela Ramirez Schrempp, Jintanat Ananworanich, Robert Paris, Ivan T Lee, Raffael Nachbagauer, David Ensz, Howard Schwartz, Lizbeth Carmona, Kristi Schaefers, Andrei Avanesov, Daniel Stadlbauer, Carole Henry, Ren Chen, Wenmei Huang, Daniela Ramirez Schrempp, Jintanat Ananworanich, Robert Paris

Abstract

Despite vaccine availability, influenza remains a substantial global public health concern. Here, we report interim findings on the primary and secondary objectives of the safety, reactogenicity, and humoral immunogenicity of a quadrivalent messenger RNA (mRNA) vaccine against seasonal influenza, mRNA-1010, from the first 2 parts of a 3-part, first-in-human, phase 1/2 clinical trial in healthy adults aged ≥18 years (NCT04956575). In the placebo-controlled Part 1, a single dose of mRNA-1010 (50 µg, 100 µg, or 200 µg) elicited hemagglutination inhibition (HAI) titers against vaccine-matched strains. In the active-comparator-controlled Part 2, mRNA-1010 (25 µg, 50 µg, or 100 µg) elicited higher HAI titers than a standard dose, inactivated seasonal influenza vaccine for influenza A strains and comparable HAI titers for influenza B strains. No safety concerns were identified; solicited adverse reactions were dose-dependent and more frequent after receipt of mRNA-1010 than the active comparator. These interim data support continued development of mRNA-1010.

Conflict of interest statement

I.L., R.N., L.C., K.S., A.A., D.S., C.H., R.C., W.H., D.R.S., J.A., and R.P. are employees of and shareholders in Moderna, Inc. D.E. and H.S. declare no competing interests.

© 2023. The Author(s).

Figures

Fig. 1. Participant disposition by study part.
Fig. 1. Participant disposition by study part.
One participant in the mRNA-1010 100-µg group in Part 2 of the study died due to cardiac arrest. This adverse event was considered by the investigator as unrelated to the study vaccination. All randomly assigned participants who received study vaccination were included in the safety population; participants were included in the group based on the actual vaccine received. The immunogenicity per-protocol population included all randomly assigned participants who received vaccination and complied with immunogenicity blood sampling timing to have baseline and ≥1 post-vaccination time point assessment, did not have influenza infection at baseline through Day 29 (as documented by polymerase chain reaction), and had no major protocol deviation that impacted the immune response. mRNA messenger RNA. *There was 1 dosing error in Part 1 (n = 1 participant randomized to mRNA-1010 200 µg but received 100 µg) and 2 dosing errors in Part 2 (n = 1 participant randomized to mRNA-1010 25 µg but received 50 µg; n = 1 participant randomized to mRNA-1010 50 µg but received active comparator).
Fig. 2. Summary of any solicited local…
Fig. 2. Summary of any solicited local and systemic adverse reactions within 7 days after vaccination by age group in each study part.
Percentages of participants in the solicited safety population reporting any solicited adverse reactions in Part 1 (a) or Part 2 (b). In Part 1, number of participants in the placebo group were 20 (18–49 years) and 24 (≥50 years); number of participants in the mRNA-1010 groups were 23 (50 µg), 21 (100 µg), and 23 (200 µg) for 18–49 years, and 22, (50 µg), 25 (100 µg), and 21 (200 µg) for ≥50 years. In Part 2, number of participants in the Afluria group were 21 (18–49 years), 19 (50–64 years), and 13 (≥65 years); number of participants in the mRNA-1010 groups were 60 (25 µg), 57 (50 µg), and 58 (100 µg) for 18–49 years; 58 (25 µg), 58 (50 µg), and 57 (100 µg), for 50–64 years; and 31 (25 µg), 31 (50 µg), and 30 (100 µg) for ≥65 years. mRNA, messenger RNA.
Fig. 3. GMTs and GMFRs of anti-hemagglutinin…
Fig. 3. GMTs and GMFRs of anti-hemagglutinin antibodies for vaccine-matched seasonal influenza strains in adults in Part 1.
Hemagglutination inhibition GMTs with associated 95% CIs against seasonal influenza A strains (A/Wisconsin/588/2019[H1N1]pdm09 and A/Hong Kong/45/2019[H3N2]) or influenza B strains (B/Washington/02/2019 (B/Victoria lineage) and B/Phuket/3073/2013 (B/Yamagata lineage)) are shown at Day 1 (baseline) and Day 29 (28 days after vaccination) among participants a 18–49 years and b ≥50 years of age in the per-protocol population for Part 1. Dots correspond to participant-level titers (18–49 years, n = 19 (placebo), 22 (mRNA-1010 50 µg), 21 (mRNA-1010 100 µg), and 22 (mRNA-1010 200 µg); ≥50 years, n = 22 (placebo), 21 (mRNA-1010 50 µg), 24 (mRNA-1010 100 µg), and 19 (mRNA-1010 200 µg). GMFRs at Day 29 from Day 1 are shown above each Day 29 bar plot. LLOQs were 10 (H1N1, B/Victoria, and B/Yamagata) or 14 (H3N2); ULOQs were 6400 (H1N1, H3N2, and B/Yamagata) or 3200 (B/Victoria). HAI GMTs <LLOQ were replaced by 0.5× LLOQ and values > ULOQ were converted to ULOQ. CI confidence interval, HAI hemagglutination inhibition, GMFR geometric mean fold-rise, GMT geometric mean titer, LLOQ lower limit of quantification, mRNA messenger RNA, ULOQ upper limit of quantification.
Fig. 4. GMTs and GMFRs of anti-hemagglutinin…
Fig. 4. GMTs and GMFRs of anti-hemagglutinin antibodies for vaccine-matched seasonal influenza strains in adults in Part 2.
Hemagglutination inhibition GMTs with associated 95% CIs against seasonal influenza A strains (A/Wisconsin/588/2019[H1N1]pdm09 and A/Cambodia/e0826360/2020[H3N2]) or influenza B strains (B/Washington/02/2019 (B/Victoria lineage) and B/Phuket/3073/2013 (B/Yamagata lineage)) are shown at Day 1 (baseline) and Day 29 (28 days after vaccination) among participants a 18–49 years, b 50–64 years, and c ≥65 years of age in the per-protocol population for Part 2. Dots corresponding to participant-level titers; numbers of participants in the Afluria groups were 20 (18–49 years), 20 (50–64 years), and 12 (≥65 years); numbers of participants in the mRNA-1010 groups were 55 (25 µg), 53 (50 µg), and 58 (100 µg) for 18–49 years; 57 (25 µg), 54 (50 µg), and 54 (100 µg) for 50–64 years; and 31 (25 µg), 31, (50 µg), and 30 (100 µg) for ≥65 years. GMFRs at Day 29 from Day 1 are shown above each Day 29 bar plot. LLOQs were 10 for each influenza strain; ULOQs were 6400 (H1N1 and B/Yamagata), 1280 (H3N2), or 3200 (B/Victoria). HAI GMTs <LLOQ were replaced by 0.5× LLOQ and values >ULOQ were converted to ULOQ. CI confidence interval, HAI hemagglutination inhibition, GMFR geometric mean fold-rise, GMT geometric mean titer, LLOQ lower limit of quantification, mRNA messenger RNA, ULOQ upper limit of quantification.
Fig. 5. Ratios of GMTs of anti-hemagglutinin…
Fig. 5. Ratios of GMTs of anti-hemagglutinin antibodies after vaccination with mRNA-1010 compared with Afluria in adults in Part 2.
Ratios of HAI GMTs with associated 95% CIs against vaccine-matched seasonal influenza strains (A/Wisconsin/588/2019[H1N1]pdm09, A/Cambodia/e0826360/2020[H3N2], B/Washington/02/2019 (B/Victoria lineage), and B/Phuket/3073/2013 (B/Yamagata lineage)) at 28 days after vaccination with mRNA-1010 compared with Afluria are shown for participants a 18–49 years, b 50–64 years, and c ≥65 years of age in the per-protocol population for Part 2. Horizontal dotted line indicates a GMT ratio of 1, which reflects comparable GMTs between mRNA-1010 and Afluria. Numbers of participants in the Alfuria groups were 20 (18–49 years), 20 (50–64 years), and 12 ( ≥65 years); numbers of participants in the mRNA-1010 groups were 55 (25 µg), 53 (50 µg), and 58 (100 µg) for 18–49 years; 57 (25 µg), 54 (50 µg), and 54 (200 µg) for 50–64 years; and 31 (25 µg), 31 (50 µg), and 30 (100 µg) for ≥65 years. CI confidence interval, HAI hemagglutination inhibition, GMT geometric mean titer, LLOQ lower limit of quantification, mRNA messenger RNA, ULOQ upper limit of quantification.

References

    1. Cowling BJ, et al. Comparative immunogenicity of several enhanced influenza vaccine options for older adults: a randomized, controlled trial. Clin. Infect. Dis. 2020;71:1704–1714. doi: 10.1093/cid/ciz1034.
    1. World Health Organization. Vaccines against influenza: WHO position paper—May 2022. Weekly Epidemiological Record.19, 185–208 (2022).
    1. Yamayoshi S, Kawaoka Y. Current and future influenza vaccines. Nat. Med. 2019;25:212–220. doi: 10.1038/s41591-018-0340-z.
    1. Centers for Disease Control and Prevention. Flu Vaccination Coverage, United States, 2020–21 Influenza Season. (2021).
    1. Luliano AD, et al. Estimates of global seasonal influenza-associated respiratory mortality: a modelling study. Lancet. 2018;391:1285–1300. doi: 10.1016/S0140-6736(17)33293-2.
    1. Lowen AC, Steel J. Roles of humidity and temperature in shaping influenza seasonality. J. Virol. 2014;88:7692–7695. doi: 10.1128/JVI.03544-13.
    1. Walker TA, et al. Risk of severe influenza among adults with chronic medical conditions. J. Infect. Dis. 2020;221:183–190. doi: 10.1093/infdis/jiz570.
    1. Centers for Disease Control and Prevention. Flu & people 65 years and older. (2022).
    1. Paget J, et al. Global mortality associated with seasonal influenza epidemics: new burden estimates and predictors from the GLaMOR Project. J. Glob. Health. 2019;9:020421. doi: 10.7189/jogh.09.020421.
    1. McElhaney JE, et al. The immune response to influenza in older humans: beyond immune senescence. Immun. Ageing. 2020;17:10. doi: 10.1186/s12979-020-00181-1.
    1. Centers for Disease Control and Prevention. Types of influenza viruses. (2021).
    1. Chivukula S, et al. Development of multivalent mRNA vaccine candidates for seasonal or pandemic influenza. NPJ Vaccines. 2021;6:153. doi: 10.1038/s41541-021-00420-6.
    1. Nachbagauer R, Palese P. Is a universal influenza virus vaccine possible. Annu. Rev. Med. 2020;71:315–327. doi: 10.1146/annurev-med-120617-041310.
    1. Centers for Disease Control and Prevention. Vaccine effectiveness: how well do flu vaccines work? (2022).
    1. Chung JR, et al. Interim estimates of 2021-22 seasonal influenza vaccine effectiveness—United States, February 2022. MMWR Morb. Mortal. Wkly. Rep. 2022;71:365–370. doi: 10.15585/mmwr.mm7110a1.
    1. Zost SJ, et al. Contemporary H3N2 influenza viruses have a glycosylation site that alters binding of antibodies elicited by egg-adapted vaccine strains. Proc. Natl Acad. Sci. USA. 2017;114:12578–12583. doi: 10.1073/pnas.1712377114.
    1. Takada K, et al. A humanized MDCK cell line for the efficient isolation and propagation of human influenza viruses. Nat. Microbiol. 2019;4:1268–1273. doi: 10.1038/s41564-019-0433-6.
    1. Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines—a new era in vaccinology. Nat. Rev. Drug Discov. 2018;17:261–279. doi: 10.1038/nrd.2017.243.
    1. Hou X, Zaks T, Langer R, Dong Y. Lipid nanoparticles for mRNA delivery. Nat. Rev. Mater. 2021;6:1078–1094. doi: 10.1038/s41578-021-00358-0.
    1. United States Food and Drug Administration. Package Insert - SPIKEVAX. (2022).
    1. Baden LR, et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N. Engl. J. Med. 2021;384:403–416. doi: 10.1056/NEJMoa2035389.
    1. Bruxvoort, K. J. et al. Real-world effectiveness of the mRNA-1273 vaccine against COVID-19: interim results from a prospective observational cohort study. Lancet Reg. Health Am. 100134, 10.1016/j.lana.2021.100134 (2021).
    1. Bruxvoort KJ, et al. Effectiveness of mRNA-1273 against delta, mu, and other emerging variants of SARS-CoV-2: test negative case-control study. BMJ. 2021;375:e068848. doi: 10.1136/bmj-2021-068848.
    1. Tseng HF, et al. Effectiveness of mRNA-1273 against SARS-CoV-2 Omicron and Delta variants. Nat. Med. 2022;28:1063–1071. doi: 10.1038/s41591-022-01753-y.
    1. Cox RJ. Correlates of protection to influenza virus, where do we go from here. Hum. Vaccine Immunother. 2013;9:405–408. doi: 10.4161/hv.22908.
    1. Dolgin E. mRNA flu shots move into trials. Nat. Rev. Drug Discov. 2021;20:801–803. doi: 10.1038/d41573-021-00176-7.
    1. Raymond DD, et al. Influenza immunization elicits antibodies specific for an egg-adapted vaccine strain. Nat. Med. 2016;22:1465–1469. doi: 10.1038/nm.4223.
    1. Lederer K, et al. SARS-CoV-2 mRNA vaccines foster potent antigen-specific germinal center responses associated with neutralizing antibody generation. Immunity. 2020;53:1281–1295.e1285. doi: 10.1016/j.immuni.2020.11.009.
    1. Turner JS, et al. SARS-CoV-2 mRNA vaccines induce persistent human germinal centre responses. Nature. 2021;596:109–113. doi: 10.1038/s41586-021-03738-2.
    1. Goel RR, et al. mRNA vaccines induce durable immune memory to SARS-CoV-2 and variants of concern. Science. 2021;374:abm0829. doi: 10.1126/science.abm0829.
    1. Mateus J, et al. Low-dose mRNA-1273 COVID-19 vaccine generates durable memory enhanced by cross-reactive T cells. Science. 2021;374:eabj9853. doi: 10.1126/science.abj9853.

Source: PubMed

3
S'abonner