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

Maheshi N Ramasamy, Angela M Minassian, Katie J Ewer, Amy L Flaxman, Pedro M Folegatti, Daniel R Owens, Merryn Voysey, Parvinder K Aley, Brian Angus, Gavin Babbage, Sandra Belij-Rammerstorfer, Lisa Berry, Sagida Bibi, Mustapha Bittaye, Katrina Cathie, Harry Chappell, Sue Charlton, Paola Cicconi, Elizabeth A Clutterbuck, Rachel Colin-Jones, Christina Dold, Katherine R W Emary, Sofiya Fedosyuk, Michelle Fuskova, Diane Gbesemete, Catherine Green, Bassam Hallis, Mimi M Hou, Daniel Jenkin, Carina C D Joe, Elizabeth J Kelly, Simon Kerridge, Alison M Lawrie, Alice Lelliott, May N Lwin, Rebecca Makinson, Natalie G Marchevsky, Yama Mujadidi, Alasdair P S Munro, Mihaela Pacurar, Emma Plested, Jade Rand, Thomas Rawlinson, Sarah Rhead, Hannah Robinson, Adam J Ritchie, Amy L Ross-Russell, Stephen Saich, Nisha Singh, Catherine C Smith, Matthew D Snape, Rinn Song, Richard Tarrant, Yrene Themistocleous, Kelly M Thomas, Tonya L Villafana, Sarah C Warren, Marion E E Watson, Alexander D Douglas, Adrian V S Hill, Teresa Lambe, Sarah C Gilbert, Saul N Faust, Andrew J Pollard, Oxford COVID Vaccine Trial Group

Abstract

Background: Older adults (aged ≥70 years) are at increased risk of severe disease and death if they develop COVID-19 and are therefore a priority for immunisation should an efficacious vaccine be developed. Immunogenicity of vaccines is often worse in older adults as a result of immunosenescence. We have reported the immunogenicity of a novel chimpanzee adenovirus-vectored vaccine, ChAdOx1 nCoV-19 (AZD1222), in young adults, and now describe the safety and immunogenicity of this vaccine in a wider range of participants, including adults aged 70 years and older.

Methods: In this report of the phase 2 component of a single-blind, randomised, controlled, phase 2/3 trial (COV002), healthy adults aged 18 years and older were enrolled at two UK clinical research facilities, in an age-escalation manner, into 18-55 years, 56-69 years, and 70 years and older immunogenicity subgroups. Participants were eligible if they did not have severe or uncontrolled medical comorbidities or a high frailty score (if aged ≥65 years). First, participants were recruited to a low-dose cohort, and within each age group, participants were randomly assigned to receive either intramuscular ChAdOx1 nCoV-19 (2·2 × 1010 virus particles) or a control vaccine, MenACWY, using block randomisation and stratified by age and dose group and study site, using the following ratios: in the 18-55 years group, 1:1 to either two doses of ChAdOx1 nCoV-19 or two doses of MenACWY; in the 56-69 years group, 3:1:3:1 to one dose of ChAdOx1 nCoV-19, one dose of MenACWY, two doses of ChAdOx1 nCoV-19, or two doses of MenACWY; and in the 70 years and older, 5:1:5:1 to one dose of ChAdOx1 nCoV-19, one dose of MenACWY, two doses of ChAdOx1 nCoV-19, or two doses of MenACWY. Prime-booster regimens were given 28 days apart. Participants were then recruited to the standard-dose cohort (3·5-6·5 × 1010 virus particles of ChAdOx1 nCoV-19) and the same randomisation procedures were followed, except the 18-55 years group was assigned in a 5:1 ratio to two doses of ChAdOx1 nCoV-19 or two doses of MenACWY. Participants and investigators, but not staff administering the vaccine, were masked to vaccine allocation. The specific objectives of this report were to assess the safety and humoral and cellular immunogenicity of a single-dose and two-dose schedule in adults older than 55 years. Humoral responses at baseline and after each vaccination until 1 year after the booster were assessed using an in-house standardised ELISA, a multiplex immunoassay, and a live severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) microneutralisation assay (MNA80). Cellular responses were assessed using an ex-vivo IFN-γ enzyme-linked immunospot assay. The coprimary outcomes of the trial were efficacy, as measured by the number of cases of symptomatic, virologically confirmed COVID-19, and safety, as measured by the occurrence of serious adverse events. Analyses were by group allocation in participants who received the vaccine. Here, we report the preliminary findings on safety, reactogenicity, and cellular and humoral immune responses. This study is ongoing and is registered with ClinicalTrials.gov, NCT04400838, and ISRCTN, 15281137.

Findings: Between May 30 and Aug 8, 2020, 560 participants were enrolled: 160 aged 18-55 years (100 assigned to ChAdOx1 nCoV-19, 60 assigned to MenACWY), 160 aged 56-69 years (120 assigned to ChAdOx1 nCoV-19: 40 assigned to MenACWY), and 240 aged 70 years and older (200 assigned to ChAdOx1 nCoV-19: 40 assigned to MenACWY). Seven participants did not receive the boost dose of their assigned two-dose regimen, one participant received the incorrect vaccine, and three were excluded from immunogenicity analyses due to incorrectly labelled samples. 280 (50%) of 552 analysable participants were female. Local and systemic reactions were more common in participants given ChAdOx1 nCoV-19 than in those given the control vaccine, and similar in nature to those previously reported (injection-site pain, feeling feverish, muscle ache, headache), but were less common in older adults (aged ≥56 years) than younger adults. In those receiving two standard doses of ChAdOx1 nCoV-19, after the prime vaccination local reactions were reported in 43 (88%) of 49 participants in the 18-55 years group, 22 (73%) of 30 in the 56-69 years group, and 30 (61%) of 49 in the 70 years and older group, and systemic reactions in 42 (86%) participants in the 18-55 years group, 23 (77%) in the 56-69 years group, and 32 (65%) in the 70 years and older group. As of Oct 26, 2020, 13 serious adverse events occurred during the study period, none of which were considered to be related to either study vaccine. In participants who received two doses of vaccine, median anti-spike SARS-CoV-2 IgG responses 28 days after the boost dose were similar across the three age cohorts (standard-dose groups: 18-55 years, 20 713 arbitrary units [AU]/mL [IQR 13 898-33 550], n=39; 56-69 years, 16 170 AU/mL [10 233-40 353], n=26; and ≥70 years 17 561 AU/mL [9705-37 796], n=47; p=0·68). Neutralising antibody titres after a boost dose were similar across all age groups (median MNA80 at day 42 in the standard-dose groups: 18-55 years, 193 [IQR 113-238], n=39; 56-69 years, 144 [119-347], n=20; and ≥70 years, 161 [73-323], n=47; p=0·40). By 14 days after the boost dose, 208 (>99%) of 209 boosted participants had neutralising antibody responses. T-cell responses peaked at day 14 after a single standard dose of ChAdOx1 nCoV-19 (18-55 years: median 1187 spot-forming cells [SFCs] per million peripheral blood mononuclear cells [IQR 841-2428], n=24; 56-69 years: 797 SFCs [383-1817], n=29; and ≥70 years: 977 SFCs [458-1914], n=48).

Interpretation: ChAdOx1 nCoV-19 appears to be better tolerated in older adults than in younger adults and has similar immunogenicity across all age groups after a boost dose. Further assessment of the efficacy of this vaccine is warranted in all age groups and individuals with comorbidities.

Funding: UK Research and Innovation, National Institutes for Health Research (NIHR), Coalition for Epidemic Preparedness Innovations, NIHR Oxford Biomedical Research Centre, Thames Valley and South Midlands NIHR Clinical Research Network, and AstraZeneca.

Copyright © 2020 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY-NC-ND 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Study profile for the low-dose (A) and standard-dose (B) cohorts *One participant excluded from immunogenicity analyses, due to mislabelling of laboratory sample. †Reasons for not receiving boost dose included that the participant moved away or was unavailable for visits, delay in receiving boost dose, or withdrawal of consent.
Figure 2
Figure 2
Solicited local adverse reactions in the 7 days after prime and boost doses of standard-dose vaccine, by age Day 0 is the day of vaccination. Participants shown are those randomly assigned to receive two doses, and data are only shown for participants who received both doses of vaccine.
Figure 3
Figure 3
Solicited systemic adverse reactions in the 7 days after prime and boost doses of standard-dose vaccine, by age Day 0 is the day of vaccination. Feverish is self-reported feeling of feverishness, whereas fever is an objective fever measurement (mild: 38·0 to

Figure 4

SARS-CoV-2 IgG response to the…

Figure 4

SARS-CoV-2 IgG response to the receptor binding domain in the standard-dose groups (A)…

Figure 4
SARS-CoV-2 IgG response to the receptor binding domain in the standard-dose groups (A) and low-dose groups (C) and the spike protein in the standard-dose groups (B) and the low-dose groups (D), by age Datapoints are medians, with whiskers showing the IQRs. Solid lines show participants who were randomly assigned to and received two doses of vaccine and dashed lines indicate participants who were randomly assigned to receive one dose. The vertical black line indicates when participants who received two doses received their boost dose. Data for the control groups are shown in the appendix (p 12)). AU=arbitrary units. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2.

Figure 5

Neutralising antibody titres measured using…

Figure 5

Neutralising antibody titres measured using a live SARS-CoV-2 microneutralisation assay (MNA 80 )…

Figure 5
Neutralising antibody titres measured using a live SARS-CoV-2 microneutralisation assay (MNA80) after prime and boost doses of vaccine in standard-dose groups (A) and low-dose groups (B), by age Datapoints are medians, with whiskers showing the IQR. Solid lines show participants who were randomly assigned to and received two doses of vaccine and dashed lines indicate participants who were randomly assigned to receive one dose. Horizontal dotted lines show upper and lower limits of assay (values outside this range set to 640 beyond the upper limit and 5 beyond the lower limit). Data for the control groups are shown in the appendix (p 14)). To normalise data across assay runs, a reference sample was included in all assay runs and test samples normalised to this value by generating log10 ratios. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2.

Figure 6

IFN-γ ELISpot response to peptides…

Figure 6

IFN-γ ELISpot response to peptides spanning the SARS-CoV-2 spike insert after prime and…

Figure 6
IFN-γ ELISpot response to peptides spanning the SARS-CoV-2 spike insert after prime and boost doses of vaccine for all participants who were given two doses of vaccine, by age group and vaccine dose ELISpot data were unavailable for the 18–55 years low-dose group because PBMCs were not collected in this group. Datapoints are medians, with whiskers showing the IQR. The lower limit of detection is 48 SFCs per million PBMCs (horizontal dotted line). Day 42 samples are from participants who received the boost dose at day 28 (vertical dotted line). Data for both one-dose and two-dose groups, with numbers analysed at each timepoint, are in the appendix (p 15)). ELISpot=enzyme-linked immunospot. PBMC=peripheral blood mononuclear cells. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2. SFC=spot-forming cells.

Figure 7

Anti-ChAdOx1 vector neutralising titres after…

Figure 7

Anti-ChAdOx1 vector neutralising titres after prime and boost doses of vaccine, by age…

Figure 7
Anti-ChAdOx1 vector neutralising titres after prime and boost doses of vaccine, by age and vaccine dose, and the correlation between pre-boost dose anti-ChAdOx1 neutralising antibodies and 28 days after boost dose antibody and T-cell responses (A) Anti-ChAdOx1 neutralising antibody titres in participants who received ChAdOx1 nCoV-19 vaccine by age and dose: datapoints are medians, with whiskers showing the IQR. Values below the limit of detection were assigned a value of 1. (B) Anti-ChAdOx1 neutralising antibody titre immediately before boost dose of vaccine versus standardised IgG ELISA against SARS-CoV-2 spike 28 days after the boost dose of vaccine with linear regression of logged values (p=0·037). (C) Anti-ChAdOx1 neutralising antibody titres immediately before boost dose of vaccine versus SARS-CoV-2 spike specific T cells measured by IFN-γ ELISpot on day 14 after the boost dose of vaccine with linear regression of logged values (p=0·22). In B and C, each datapoint is one participant and the solid line shows the linear regression, with the shaded area showing the 95% CI from an unadjusted linear regression of anti-vector neutralisation titres against logged ELISA (in B) or ELISpot (in C) response. Data were unavailable at day 56 for the 56–69 years standard-dose group. ELISpot=enzyme-linked immunospot. PBMC=peripheral blood mononuclear cells. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2. SFC=spot-forming cells.
All figures (7)
Figure 4
Figure 4
SARS-CoV-2 IgG response to the receptor binding domain in the standard-dose groups (A) and low-dose groups (C) and the spike protein in the standard-dose groups (B) and the low-dose groups (D), by age Datapoints are medians, with whiskers showing the IQRs. Solid lines show participants who were randomly assigned to and received two doses of vaccine and dashed lines indicate participants who were randomly assigned to receive one dose. The vertical black line indicates when participants who received two doses received their boost dose. Data for the control groups are shown in the appendix (p 12)). AU=arbitrary units. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2.
Figure 5
Figure 5
Neutralising antibody titres measured using a live SARS-CoV-2 microneutralisation assay (MNA80) after prime and boost doses of vaccine in standard-dose groups (A) and low-dose groups (B), by age Datapoints are medians, with whiskers showing the IQR. Solid lines show participants who were randomly assigned to and received two doses of vaccine and dashed lines indicate participants who were randomly assigned to receive one dose. Horizontal dotted lines show upper and lower limits of assay (values outside this range set to 640 beyond the upper limit and 5 beyond the lower limit). Data for the control groups are shown in the appendix (p 14)). To normalise data across assay runs, a reference sample was included in all assay runs and test samples normalised to this value by generating log10 ratios. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2.
Figure 6
Figure 6
IFN-γ ELISpot response to peptides spanning the SARS-CoV-2 spike insert after prime and boost doses of vaccine for all participants who were given two doses of vaccine, by age group and vaccine dose ELISpot data were unavailable for the 18–55 years low-dose group because PBMCs were not collected in this group. Datapoints are medians, with whiskers showing the IQR. The lower limit of detection is 48 SFCs per million PBMCs (horizontal dotted line). Day 42 samples are from participants who received the boost dose at day 28 (vertical dotted line). Data for both one-dose and two-dose groups, with numbers analysed at each timepoint, are in the appendix (p 15)). ELISpot=enzyme-linked immunospot. PBMC=peripheral blood mononuclear cells. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2. SFC=spot-forming cells.
Figure 7
Figure 7
Anti-ChAdOx1 vector neutralising titres after prime and boost doses of vaccine, by age and vaccine dose, and the correlation between pre-boost dose anti-ChAdOx1 neutralising antibodies and 28 days after boost dose antibody and T-cell responses (A) Anti-ChAdOx1 neutralising antibody titres in participants who received ChAdOx1 nCoV-19 vaccine by age and dose: datapoints are medians, with whiskers showing the IQR. Values below the limit of detection were assigned a value of 1. (B) Anti-ChAdOx1 neutralising antibody titre immediately before boost dose of vaccine versus standardised IgG ELISA against SARS-CoV-2 spike 28 days after the boost dose of vaccine with linear regression of logged values (p=0·037). (C) Anti-ChAdOx1 neutralising antibody titres immediately before boost dose of vaccine versus SARS-CoV-2 spike specific T cells measured by IFN-γ ELISpot on day 14 after the boost dose of vaccine with linear regression of logged values (p=0·22). In B and C, each datapoint is one participant and the solid line shows the linear regression, with the shaded area showing the 95% CI from an unadjusted linear regression of anti-vector neutralisation titres against logged ELISA (in B) or ELISpot (in C) response. Data were unavailable at day 56 for the 56–69 years standard-dose group. ELISpot=enzyme-linked immunospot. PBMC=peripheral blood mononuclear cells. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2. SFC=spot-forming cells.

References

    1. WHO . World Health Organization; Geneva: Nov 13, 2020. Coronavirus disease (COVID-19) dashboard.
    1. Wu Z, McGoogan JM. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China: summary of a report of 72 314 cases From the Chinese Center for Disease Control and Prevention. JAMA. 2020;323:1239–1242.
    1. WHO . World Health Organization; Geneva: Nov 12, 2020. Draft landscape of COVID-19 vaccine candidates.
    1. WHO . World Health Organization; Geneva: April 29, 2020. WHO target product profiles for COVID-19 vaccines: version 3.
    1. Joint Committee on Vaccination and Immunisation Priority groups for coronavirus (COVID-19) vaccination: advice from the JCVI, 25 September 2020. UK Government. Sept 25, 2020.
    1. US Centers for Disease Control and Prevention The COVID-19 vaccination program interim operational guidance for jurisdictions playbook. Nov 5, 2020.
    1. Aw D, Silva AB, Palmer DB. Immunosenescence: emerging challenges for an ageing population. Immunology. 2007;120:435–446.
    1. Peng Y, Mentzer AJ, Liu G. Broad and strong memory CD4+ and CD8+ T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nat Immunol. 2020;21:1336–1345.
    1. Anderson EJ, Rouphael NG, Widge AT. Safety and immunogenicity of SARS-CoV-2 mRNA-1273 vaccine in older adults. N Engl J Med. 2020 doi: 10.1056/NEJMoa2028436. published online Sept 29.
    1. Walsh EE, Frenck RW, Jr, Falsey AR. Safety and immunogenicity of two RNA-based COVID-19 vaccine candidates. N Engl J Med. 2020 doi: 10.1056/NEJMoa2027906. published online Oct 14.
    1. Zhu FC, Guan XH, Li YH. Immunogenicity and safety of a recombinant adenovirus type-5-vectored COVID-19 vaccine in healthy adults aged 18 years or older: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet. 2020;396:479–488.
    1. Logunov DY, Dolzhikova IV, Zubkova OV. Safety and immunogenicity of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: two open, non-randomised phase 1/2 studies from Russia. Lancet. 2020;396:887–897.
    1. Xia S, Zhang Y, Wang Y. Safety and immunogenicity of an inactivated SARS-CoV-2 vaccine, BBIBP-CorV: a randomised, double-blind, placebo-controlled, phase 1/2 trial. Lancet Infect Dis. 2020 doi: 10.1016/S1473-3099(20)30831-8. published online Oct 15.
    1. Xia S, Duan K, Zhang Y. Effect of an inactivated vaccine against SARS-CoV-2 on safety and immunogenicity outcomes: interim analysis of 2 randomized clinical trials. JAMA. 2020;324:951–960.
    1. Keech C, Albert G, Cho I. Phase 1-2 Trial of a SARS-CoV-2 recombinant spike protein nanoparticle vaccine. N Engl J Med. 2020 doi: 10.1056/NEJMoa2026920. published online Sept 2.
    1. Milligan ID, Gibani MM, Sewell R. Safety and immunogenicity of novel adenovirus type 26- and modified vaccinia Ankara-vectored Ebola vaccines: a randomized clinical trial. JAMA. 2016;315:1610–1623.
    1. van Doremalen N, Lambe T, Spencer A. ChAdOx1 nCoV-19 vaccine prevents SARS-CoV-2 pneumonia in rhesus macaques. Nature. 2020;586:578–582.
    1. Folegatti PM, Ewer KJ, Aley PK. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet. 2020;396:467–478.
    1. Rockwood K, Song X, MacKnight C. A global clinical measure of fitness and frailty in elderly people. CMAJ. 2005;173:489–495.
    1. Nimenrix: summary of medicinal product characteristics. Oct 28, 2020.
    1. Dicks MD, Spencer AJ, Edwards NJ. A novel chimpanzee adenovirus vector with low human seroprevalence: improved systems for vector derivation and comparative immunogenicity. PLoS One. 2012;7
    1. Coughlan L, Sridhar S, Payne R. Heterologous two-dose vaccination with simian adenovirus and poxvirus vectors elicits long-lasting cellular immunity to influenza virus A in healthy adults. EBioMedicine. 2018;29:146–154.
    1. Tapia MD, Sow SO, Mbaye KD. Safety, reactogenicity, and immunogenicity of a chimpanzee adenovirus vectored Ebola vaccine in children in Africa: a randomised, observer-blind, placebo-controlled, phase 2 trial. Lancet Infect Dis. 2020;20:719–730.
    1. Wilkie M, Satti I, Minhinnick A. A phase I trial evaluating the safety and immunogenicity of a candidate tuberculosis vaccination regimen, ChAdOx1 85A prime - MVA85A boost in healthy UK adults. Vaccine. 2020;38:779–789.
    1. Ewer K, Rampling T, Venkatraman N. A monovalent chimpanzee adenovirus Ebola vaccine boosted with MVA. N Engl J Med. 2016;374:1635–1646.
    1. Lambert PH, Ambrosino DM, Andersen SR. Consensus summary report for CEPI/BC March 12-13, 2020 meeting: assessment of risk of disease enhancement with COVID-19 vaccines. Vaccine. 2020;38:4783–4791.
    1. Goodwin K, Viboud C, Simonsen L. Antibody response to influenza vaccination in the elderly: a quantitative review. Vaccine. 2006;24:1159–1169.
    1. Denis F, Mounier M, Hessel L. Hepatitis-B vaccination in the elderly. J Infect Dis. 1984;149
    1. Williams K, Bastian AR, Feldman RA. Phase 1 safety and immunogenicity study of a respiratory syncytial virus vaccine with an adenovirus 26 vector encoding prefusion F (Ad26.RSV.preF) in adults aged ≥60 years. J Infect Dis. 2020;222:979–988.
    1. Green CA, Sande CJ, Scarselli E. Novel genetically-modified chimpanzee adenovirus and MVA-vectored respiratory syncytial virus vaccine safely boosts humoral and cellular immunity in healthy older adults. J Infect. 2019;78:382–392.

Source: PubMed

3
Sottoscrivi