Immunogenicity and Reactogenicity of Vaccine Boosters after Ad26.COV2.S Priming

Roos S G Sablerolles, Wim J R Rietdijk, Abraham Goorhuis, Douwe F Postma, Leo G Visser, Daryl Geers, Katharina S Schmitz, Hannah M Garcia Garrido, Marion P G Koopmans, Virgil A S H Dalm, Neeltje A Kootstra, Anke L W Huckriede, Melvin Lafeber, Debbie van Baarle, Corine H GeurtsvanKessel, Rory D de Vries, P Hugo M van der Kuy, SWITCH Research Group, N Tjon, K van Grafhorst, L P M van Leeuwen, S Bogers, N Nieuwkoop, F de Wilt, L Gommers, S Scherbeijn, A C P Lamoré, A M Harskamp, I Maurer, A F Girigorie, B D Boeser-Nunnink, M M Mangas Ruiz, R Akkerman, M Beukema, J J de Vries-Idema, J Zuidema, J A Vlot, P H Verbeek-Menken, K Suijk-Benschop, J Fehrmann-Naumann, A van Wengen-Stevenhagen, Roos S G Sablerolles, Wim J R Rietdijk, Abraham Goorhuis, Douwe F Postma, Leo G Visser, Daryl Geers, Katharina S Schmitz, Hannah M Garcia Garrido, Marion P G Koopmans, Virgil A S H Dalm, Neeltje A Kootstra, Anke L W Huckriede, Melvin Lafeber, Debbie van Baarle, Corine H GeurtsvanKessel, Rory D de Vries, P Hugo M van der Kuy, SWITCH Research Group, N Tjon, K van Grafhorst, L P M van Leeuwen, S Bogers, N Nieuwkoop, F de Wilt, L Gommers, S Scherbeijn, A C P Lamoré, A M Harskamp, I Maurer, A F Girigorie, B D Boeser-Nunnink, M M Mangas Ruiz, R Akkerman, M Beukema, J J de Vries-Idema, J Zuidema, J A Vlot, P H Verbeek-Menken, K Suijk-Benschop, J Fehrmann-Naumann, A van Wengen-Stevenhagen

Abstract

Background: The Ad26.COV2.S vaccine, which was approved as a single-shot immunization regimen, has been shown to be effective against severe coronavirus disease 2019. However, this vaccine induces lower severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein (S)-specific antibody levels than those induced by messenger RNA (mRNA)-based vaccines. The immunogenicity and reactogenicity of a homologous or heterologous booster in persons who have received an Ad26.COV2.S priming dose are unclear.

Methods: In this single-blind, multicenter, randomized, controlled trial involving health care workers who had received a priming dose of Ad26.COV2.S vaccine, we assessed immunogenicity and reactogenicity 28 days after a homologous or heterologous booster vaccination. The participants were assigned to receive no booster, an Ad26.COV2.S booster, an mRNA-1273 booster, or a BNT162b2 booster. The primary end point was the level of S-specific binding antibodies, and the secondary end points were the levels of neutralizing antibodies, S-specific T-cell responses, and reactogenicity. A post hoc analysis was performed to compare mRNA-1273 boosting with BNT162b2 boosting.

Results: Homologous or heterologous booster vaccination resulted in higher levels of S-specific binding antibodies, neutralizing antibodies, and T-cell responses than a single Ad26.COV2.S vaccination. The increase in binding antibodies was significantly larger with heterologous regimens that included mRNA-based vaccines than with the homologous booster. The mRNA-1273 booster was most immunogenic and was associated with higher reactogenicity than the BNT162b2 and Ad26.COV2.S boosters. Local and systemic reactions were generally mild to moderate in the first 2 days after booster administration.

Conclusions: The Ad26.COV2.S and mRNA boosters had an acceptable safety profile and were immunogenic in health care workers who had received a priming dose of Ad26.COV2.S vaccine. The strongest responses occurred after boosting with mRNA-based vaccines. Boosting with any available vaccine was better than not boosting. (Funded by the Netherlands Organization for Health Research and Development ZonMw; SWITCH ClinicalTrials.gov number, NCT04927936.).

Copyright © 2022 Massachusetts Medical Society.

Figures

Figure 1. Screening, Randomization, and Analysis.
Figure 1. Screening, Randomization, and Analysis.
Between trial enrollment and randomization, 236 health care workers were excluded, including 78 who did not meet the inclusion criteria, 18 who declined to participate after reading the patient information form, and 140 who could not adhere to the strict schedule of the trial or did not reply to the screening questionnaire. After randomization, the numbers of participants who were lost to follow-up did not differ significantly among the groups. SARS-CoV-2 denotes severe acute respiratory syndrome coronavirus 2.
Figure 2. SARS-CoV-2 S–Specific Immune Responses.
Figure 2. SARS-CoV-2 S–Specific Immune Responses.
Panel A shows levels of SARS-CoV-2 spike protein (S)–specific IgG antibodies at baseline (before booster vaccination) and after booster vaccination in the four groups. The lower limit of detection (LLoD) was 4.81 binding antibody units (BAU) per milliliter. The cutoff value for response was 33.8 BAU per milliliter (horizontal line). Panel B shows the per-participant factor changes that were calculated by dividing the after-booster response by the before-booster response for S-specific binding antibodies. The dashed line indicates a factor change of 1 (no increase or decrease). Panel C shows the levels of neutralizing antibodies at baseline (before booster vaccination) and after booster vaccination, as assessed with a plaque reduction neutralization test with a 50% cutoff (PRNT50) in the four groups. The LLoD was 7.7 IU per milliliter. The cutoff value for response was 28.6 IU per milliliter (corresponding to a serum dilution of 1:40; horizontal line). Panel D shows the per-participant factor changes that were calculated by dividing the after-booster response by the before-booster response for neutralizing antibodies. The dashed line indicates a factor change of 1 (no increase or decrease). Panel E shows interferon-γ levels in plasma after stimulation of whole blood with a peptide pool spanning the S protein at baseline (before booster) and after booster vaccination in the four groups. The LLoD was 0.01 IU per milliliter. The cutoff value for response was 0.15 IU per milliliter (horizontal line). Panel F shows per-participant factor changes calculated by dividing the after-booster response by the before-booster response for interferon-γ levels in plasma. The dashed line indicates a factor change of 1 (no increase or decrease). All data are presented in box-and-whisker plots. The whiskers indicate the range, the top and bottom of the boxes indicate the interquartile range, and the horizontal line within each box indicates the median. P values are reported for prespecified primary and secondary end points only (on the basis of Mann–Whitney tests). Comparisons between mRNA-1273 and BNT162b2 boosters (Panels A, C, and E, right panel) and comparisons among the groups on the basis of factor changes (Panels B, D, and F) were performed as post hoc analyses, and estimated effect sizes are reported in Table S4. Each dot in the figure represents an individual participant.
Figure 3. Correlation between S-Specific IgG Antibodies…
Figure 3. Correlation between S-Specific IgG Antibodies and Neutralizing Antibodies.
Panel A shows the correlation between S-specific IgG antibody levels and neutralizing antibody levels for the before-booster data, as assessed with PRNT50 (Spearman’s rank correlation coefficient, 0.82). Linear regression (diagonal lines) on log-transformed data was performed (beta coefficient, 0.90; 95% CI, 0.82 to 0.98). The gray shaded areas indicate the 95% CI of the best-fit line. Panel B shows the correlation between S-specific IgG antibody levels and neutralizing antibody levels for the after-booster data (Spearman’s rank correlation coefficient, 0.93). Linear regression (diagonal lines) on log-transformed data was performed (beta coefficient, 0.72; 95% CI, 0.68 to 0.76). The gray shaded areas indicate the 95% CI of the best-fit line. Each dot in the figure represents an individual participant.
Figure 4. Severity of Systemic and Local…
Figure 4. Severity of Systemic and Local Reactions after Booster Vaccination.
Panel A shows the percentage of participants with systemic reactions (fatigue, chills, fever, nausea, headache, muscle aches, or joint pain), and Panel B shows the percentage of participants with local reactions (redness, swelling, or pain at the injection site) after booster vaccination. These reactions were monitored in the 7 days after the administration of the booster. P values for the between-group differences in the distribution of adverse events are shown in Table S7.

References

    1. Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med 2020;383:2603-2615.
    1. 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;384:403-416.
    1. Sadoff J, Gray G, Vandebosch A, et al. Safety and efficacy of single-dose Ad26.COV2.S vaccine against Covid-19. N Engl J Med 2021;384:2187-2201.
    1. Voysey M, Clemens SAC, Madhi SA, et al. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet 2021;397:99-111.
    1. Barouch DH, Stephenson KE, Sadoff J, et al. Durable humoral and cellular immune responses 8 months after Ad26. COV2.S vaccination. N Engl J Med 2021;385:951-953.
    1. Sadoff J, Le Gars M, Cardenas V, et al. Durability of antibody responses elicited by a single dose of Ad26.COV2.S and substantial increase following late boosting. August 26, 2021. (). preprint.
    1. van Gils MJ, Lavell AHA, van der Straten K, et al. Four SARS-CoV-2 vaccines induce quantitatively different antibody responses against SARS-CoV-2 variants. September 28, 2021. (). preprint.
    1. Collier A-RY, Yu J, McMahan K, et al. Differential kinetics of immune responses elicited by Covid-19 vaccines. N Engl J Med 2021;385:2010-2012.
    1. Sablerolles RSG, Goorhuis A, GeurtsvanKessel CH, et al. Heterologous Ad26.COV2.S prime and mRNA-based boost COVID-19 vaccination regimens: the SWITCH Trial protocol. Front Immunol 2021;12:753319-753319.
    1. Spencer AJ, McKay PF, Belij-Rammerstorfer S, et al. Heterologous vaccination regimens with self-amplifying RNA and adenoviral COVID vaccines induce robust immune responses in mice. Nat Commun 2021;12:2893-2893.
    1. He Q, Mao Q, An C, et al. Heterologous prime-boost: breaking the protective immune response bottleneck of COVID-19 vaccine candidates. Emerg Microbes Infect 2021;10:629-637.
    1. Duarte-Salles T, Prieto-Alhambra D. Heterologous vaccine regimens against COVID-19. Lancet 2021;398:94-95.
    1. Hacisuleyman E, Hale C, Saito Y, et al. Vaccine breakthrough infections with SARS-CoV-2 variants. N Engl J Med 2021;384:2212-2218.
    1. Krause PR, Fleming TR, Longini IM, et al. SARS-CoV-2 variants and vaccines. N Engl J Med 2021;385:179-186.
    1. Liu X, Shaw RH, Stuart ASV, et al. Safety and immunogenicity of heterologous versus homologous prime-boost schedules with an adenoviral vectored and mRNA COVID-19 vaccine (Com-COV): a single-blind, randomised, non-inferiority trial. Lancet 2021;398:856-869.
    1. Hillus D, Schwarz T, Tober-Lau P, et al. Safety, reactogenicity, and immunogenicity of homologous and heterologous prime-boost immunisation with ChAdOx1 nCoV-19 and BNT162b2: a prospective cohort study. Lancet Respir Med 2021;9:1255-1265.
    1. Borobia AM, Carcas AJ, Pérez-Olmeda M, et al. Immunogenicity and reactogenicity of BNT162b2 booster in ChAdOx1-S-primed participants (CombiVacS): a multicentre, open-label, randomised, controlled, phase 2 trial. Lancet 2021;398:121-130.
    1. Shaw RH, Stuart A, Greenland M, Liu X, Nguyen Van-Tam JS, Snape MD. Heterologous prime-boost COVID-19 vaccination: initial reactogenicity data. Lancet 2021;397:2043-2046.
    1. Atmar RL, Lyke KE, Deming ME, et al. Heterologous SARS-CoV-2 booster vaccinations — preliminary report. October 15, 2021. (). preprint.
    1. Food and Drug Administration. Toxicity grading scale for healthy adults and adolescent volunteers enrolled in preventive vaccine clinical trials. Guidance for industry. September 2007. ().
    1. Mahmoud SA, Ganesan S, Bissar S, Zamil I, Zaher WA. Evaluation of serological tests for detecting SARS-CoV-2 antibodies: implementation in assessing post vaccination status. April 30, 2021. (). preprint.
    1. Leuzinger K, Osthoff M, Dräger S, et al. Comparing immunoassays for SARS-CoV-2 antibody detection in patients with and without laboratory-confirmed SARS-CoV-2 infection. J Clin Microbiol 2021;59:e0138121-e0138121.
    1. Sanders J-SF, Bemelman FJ, Messchendorp AL, et al. The RECOVAC Immune-Response Study: the immunogenicity, tolerability and safety of COVID-19 vaccination in patients with chronic kidney disease, on dialysis, or living with a kidney transplant. Transplantation 2021. November 19 (Epub ahead of print).
    1. Goel RR, Painter MM, Apostolidis SA, et al. mRNA vaccines induce durable immune memory to SARS-CoV-2 and variants of concern. Science 2021;374:abm0829-abm0829.
    1. Krammer F. A correlate of protection for SARS-CoV-2 vaccines is urgently needed. Nat Med 2021;27:1147-1148.
    1. 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;27:1205-1211.
    1. Mlcochova P, Kemp SA, Dhar MS, et al. SARS-CoV-2 B.1.617.2 Delta variant replication and immune evasion. Nature 2021;599:114-119.
    1. Planas D, Veyer D, Baidaliuk A, et al. Reduced sensitivity of SARS-CoV-2 variant Delta to antibody neutralization. Nature 2021;596:276-280.
    1. Alter G, Yu J, Liu J, et al. Immunogenicity of Ad26.COV2.S vaccine against SARS-CoV-2 variants in humans. Nature 2021;596:268-272.
    1. Tan AT, Lim JM, Le Bert N, et al. Rapid measurement of SARS-CoV-2 spike T cells in whole blood from vaccinated and naturally infected individuals. J Clin Invest 2021;131:131-131.
    1. Geers D, Shamier MC, Bogers S, et al. SARS-CoV-2 variants of concern partially escape humoral but not T-cell responses in COVID-19 convalescent donors and vaccinees. Sci Immunol 2021;6:eabj1750-eabj1750.
    1. GeurtsvanKessel CH, Geers D, Schmitz KS, et al. Divergent SARS CoV-2 Omicron-specific T- and B-cell responses in COVID-19 vaccine recipients. December 27, 2021. (). preprint.
    1. Anderson EJ, Rouphael NG, Widge AT, et al. Safety and immunogenicity of SARS-CoV-2 mRNA-1273 vaccine in older adults. N Engl J Med 2020;383:2427-2438.
    1. Walsh EE, Frenck RW Jr, Falsey AR, et al. Safety and immunogenicity of two RNA-based Covid-19 vaccine candidates. N Engl J Med 2020;383:2439-2450.
    1. Ramasamy MN, Minassian AM, Ewer KJ, et al. Safety and immunogenicity of ChAdOx1 nCoV-19 vaccine administered in a prime-boost regimen in young and old adults (COV002): a single-blind, randomised, controlled, phase 2/3 trial. Lancet 2021;396:1979-1993.
    1. Voysey M, Costa Clemens SA, Madhi SA, et al. Single-dose administration and the influence of the timing of the booster dose on immunogenicity and efficacy of ChAdOx1 nCoV-19 (AZD1222) vaccine: a pooled analysis of four randomised trials. Lancet 2021;397:881-891.
    1. Stephenson KE, Le Gars M, Sadoff J, et al. Immunogenicity of the Ad26.COV2.S vaccine for COVID-19. JAMA 2021;325:1535-1544.

Source: PubMed

3
Tilaa