Efficacy of ChAdOx1 nCoV-19 (AZD1222) vaccine against SARS-CoV-2 variant of concern 202012/01 (B.1.1.7): an exploratory analysis of a randomised controlled trial

Katherine R W Emary, Tanya Golubchik, Parvinder K Aley, Cristina V Ariani, Brian Angus, Sagida Bibi, Beth Blane, David Bonsall, Paola Cicconi, Sue Charlton, Elizabeth A Clutterbuck, Andrea M Collins, Tony Cox, Thomas C Darton, Christina Dold, Alexander D Douglas, Christopher J A Duncan, Katie J Ewer, Amy L Flaxman, Saul N Faust, Daniela M Ferreira, Shuo Feng, Adam Finn, Pedro M Folegatti, Michelle Fuskova, Eva Galiza, Anna L Goodman, Catherine M Green, Christopher A Green, Melanie Greenland, Bassam Hallis, Paul T Heath, Jodie Hay, Helen C Hill, Daniel Jenkin, Simon Kerridge, Rajeka Lazarus, Vincenzo Libri, Patrick J Lillie, Catherine Ludden, Natalie G Marchevsky, Angela M Minassian, Alastair C McGregor, Yama F Mujadidi, Daniel J Phillips, Emma Plested, Katrina M Pollock, Hannah Robinson, Andrew Smith, Rinn Song, Matthew D Snape, Rebecca K Sutherland, Emma C Thomson, Mark Toshner, David P J Turner, Johan Vekemans, Tonya L Villafana, Christopher J Williams, Adrian V S Hill, Teresa Lambe, Sarah C Gilbert, Merryn Voysey, Maheshi N Ramasamy, Andrew J Pollard, COVID-19 Genomics UK consortium, AMPHEUS Project, Oxford COVID-19 Vaccine Trial Group, Katherine R W Emary, Tanya Golubchik, Parvinder K Aley, Cristina V Ariani, Brian Angus, Sagida Bibi, Beth Blane, David Bonsall, Paola Cicconi, Sue Charlton, Elizabeth A Clutterbuck, Andrea M Collins, Tony Cox, Thomas C Darton, Christina Dold, Alexander D Douglas, Christopher J A Duncan, Katie J Ewer, Amy L Flaxman, Saul N Faust, Daniela M Ferreira, Shuo Feng, Adam Finn, Pedro M Folegatti, Michelle Fuskova, Eva Galiza, Anna L Goodman, Catherine M Green, Christopher A Green, Melanie Greenland, Bassam Hallis, Paul T Heath, Jodie Hay, Helen C Hill, Daniel Jenkin, Simon Kerridge, Rajeka Lazarus, Vincenzo Libri, Patrick J Lillie, Catherine Ludden, Natalie G Marchevsky, Angela M Minassian, Alastair C McGregor, Yama F Mujadidi, Daniel J Phillips, Emma Plested, Katrina M Pollock, Hannah Robinson, Andrew Smith, Rinn Song, Matthew D Snape, Rebecca K Sutherland, Emma C Thomson, Mark Toshner, David P J Turner, Johan Vekemans, Tonya L Villafana, Christopher J Williams, Adrian V S Hill, Teresa Lambe, Sarah C Gilbert, Merryn Voysey, Maheshi N Ramasamy, Andrew J Pollard, COVID-19 Genomics UK consortium, AMPHEUS Project, Oxford COVID-19 Vaccine Trial Group

Abstract

Background: A new variant of SARS-CoV-2, B.1.1.7, emerged as the dominant cause of COVID-19 disease in the UK from November, 2020. We report a post-hoc analysis of the efficacy of the adenoviral vector vaccine, ChAdOx1 nCoV-19 (AZD1222), against this variant.

Methods: Volunteers (aged ≥18 years) who were enrolled in phase 2/3 vaccine efficacy studies in the UK, and who were randomly assigned (1:1) to receive ChAdOx1 nCoV-19 or a meningococcal conjugate control (MenACWY) vaccine, provided upper airway swabs on a weekly basis and also if they developed symptoms of COVID-19 disease (a cough, a fever of 37·8°C or higher, shortness of breath, anosmia, or ageusia). Swabs were tested by nucleic acid amplification test (NAAT) for SARS-CoV-2 and positive samples were sequenced through the COVID-19 Genomics UK consortium. Neutralising antibody responses were measured using a live-virus microneutralisation assay against the B.1.1.7 lineage and a canonical non-B.1.1.7 lineage (Victoria). The efficacy analysis included symptomatic COVID-19 in seronegative participants with a NAAT positive swab more than 14 days after a second dose of vaccine. Participants were analysed according to vaccine received. Vaccine efficacy was calculated as 1 - relative risk (ChAdOx1 nCoV-19 vs MenACWY groups) derived from a robust Poisson regression model. This study is continuing and is registered with ClinicalTrials.gov, NCT04400838, and ISRCTN, 15281137.

Findings: Participants in efficacy cohorts were recruited between May 31 and Nov 13, 2020, and received booster doses between Aug 3 and Dec 30, 2020. Of 8534 participants in the primary efficacy cohort, 6636 (78%) were aged 18-55 years and 5065 (59%) were female. Between Oct 1, 2020, and Jan 14, 2021, 520 participants developed SARS-CoV-2 infection. 1466 NAAT positive nose and throat swabs were collected from these participants during the trial. Of these, 401 swabs from 311 participants were successfully sequenced. Laboratory virus neutralisation activity by vaccine-induced antibodies was lower against the B.1.1.7 variant than against the Victoria lineage (geometric mean ratio 8·9, 95% CI 7·2-11·0). Clinical vaccine efficacy against symptomatic NAAT positive infection was 70·4% (95% CI 43·6-84·5) for B.1.1.7 and 81·5% (67·9-89·4) for non-B.1.1.7 lineages.

Interpretation: ChAdOx1 nCoV-19 showed reduced neutralisation activity against the B.1.1.7 variant compared with a non-B.1.1.7 variant in vitro, but the vaccine showed efficacy against the B.1.1.7 variant of SARS-CoV-2.

Funding: UK Research and Innovation, National Institute 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.

Conflict of interest statement

Declaration of interests Oxford University has entered into a partnership with AstraZeneca for further development of ChAdOx1 nCoV-19. AstraZeneca reviewed the data from the study and the final manuscript before submission but the authors retained editorial control. SCG is cofounder of Vaccitech (collaborators in the early development of this vaccine candidate) and is named as an inventor on a patent covering use of ChAdOx1-vectored vaccines (PCT/GB2012/000467) and a patent application covering this SARS-CoV-2 vaccine. TL is named as an inventor on a patent application covering this SARS-CoV-2 vaccine and was a consultant to Vaccitech. PMF is a consultant to Vaccitech. AJP is chair of the UK Department of Health and Social Care Joint Committee on Vaccination and Immunisation but does not participate in policy advice on coronavirus vaccines, and is a member of the WHO Strategic Advisory Group of Experts. AJP and SNF are NIHR senior investigators. AVSH is a cofounder of and consultant to Vaccitech and is named as an inventor on a patent covering design and use of ChAdOx1-vectored vaccines (PCT/GB2012/000467). MDS reports grants from Janssen, GlaxoSmithKline, Medimmune, Novavax, and MCM Vaccine, and grants and non-financial support from Pfizer outside of the submitted work. CMG reports personal fees from the Duke Human Vaccine Institute outside of the submitted work. ADD reports grants and personal fees from AstraZeneca outside of the submitted work. SNF reports grants from Janssen and Valneva outside of the submitted work. TLV and JV are employees of AstraZeneca. All other authors declare no competing interests.

Copyright © 2021 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Flow diagram of swabs included in the analysis
Figure 2
Figure 2
Consensus phylogeny of SARS-CoV-2 genomes identified in this study Clades are coloured by variant lineage and tips are coloured by vaccine allocation. Only genomes with at least 40% coverage are included (n=247). Lineages were assigned by Pangolin version 2.1.7 (lineages version 2021–02–12).
Figure 3
Figure 3
Weekly and cumulative number of B.1.1.7 and non-B.1.1.7 isolates identified in the UK trial between Oct 1, 2020, and Jan 14, 2021 Bars show overall weekly case counts (left axis) and lines show cumulative case counts (right axis).
Figure 4
Figure 4
Minimum Ct values across all NAAT positive swabs Ct values from positive NAATs performed at Lighthouse Laboratories using a ThermoFisher TaqPath three-gene assay. Each datapoint represents one participant. For each participant, the minimum of the Ct value for the N gene and ORF1ab gene was taken for each NAAT positive swab, and the minimum across all swabs for the same participant was calculated as a proxy for maximum viral load. Low Ct values are associated with a higher viral load. The midlines of the boxes show medians and the outer bounds of the boxes show IQRs. Error bars show the most extreme point within 1.5 × IQR above or below the 75th or 25th percentile. Ct=cycle threshold. NAAT=nucleic acid amplification test.
Figure 5
Figure 5
Length of the NAAT-positive period per participant Three primary symptomatic participants who received ChAdOx1 nCoV-19 remained NAAT positive for 8, 9, and 11 weeks, respectively, and one primary symptomatic participant who received the control vaccine remained NAAT positive for 11 weeks; these cases are not shown in the figure. NAAT=nucleic acid amplification test.
Figure 6
Figure 6
Live-virus microneutralisation antibody titres of sera against B.1.1.7 and a canonical non-B.1.1.7 strain (Victoria) The geometric mean titre is 58 (95% CI 44–77) for B.1.1.7 and 517 (424–631) for Victoria. The geometric mean ratio (Victoria vs B.1.1.7) is 8·9 (95% CI 7·2–11·0). The midlines of the boxes show medians and the outer bounds of the boxes show IQRs. Error bars show the most extreme point within 1·5 × IQR above or below the 75th or 25th percentile. Lines connect samples from the same participant collected at the same trial timepoint (n=49). ND50=titre at which 50% virus neutralisation is achieved.

References

    1. Polack FP, Thomas SJ, Kitchin N. 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. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med. 2021;384:403–416.
    1. Voysey M, Clemens SAC, Madhi SA. 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. Novavax Novavax COVID-19 vaccine demonstrates 89.3% efficacy in UK phase 3 trial. Jan 28, 2021.
    1. Janssen Johnson & Johnson announces single-shot Janssen COVID-19 vaccine candidate met primary endpoints in interim analysis of its phase 3 ENSEMBLE trial. Jan 29, 2021.
    1. Logunov DY, Dolzhikova IV, Shcheblyakov DV. Safety and efficacy of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine: an interim analysis of a randomised controlled phase 3 trial in Russia. Lancet. 2021;397:671–681.
    1. Faria NR, Morales Claro I, Candido D. Genomic characterisation of an emergent SARS-CoV-2 lineage in Manaus: preliminary findings. January, 2021.
    1. Tegally H, Wilkinson E, Giovanetti M. Emergence and rapid spread of a new severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) lineage with multiple spike mutations in South Africa. medRxiv. 2020 doi: 10.1101/2020.12.21.20248640. published online Dec 22. (preprint).
    1. Rambaut A, Loman N, Pybus O. Preliminary genomic characterisation of an emergent SARS-CoV-2 lineage in the UK defined by a novel set of spike mutations. December, 2020.
    1. Starr TN, Greaney AJ, Hilton SK. Deep mutational scanning of SARS-CoV-2 receptor binding domain reveals constraints on folding and ACE2 binding. Cell. 2020;182:1295. 310.e20.
    1. Kemp SA, Datir RP, Collier DA. Recurrent emergence and transmission of a SARS-CoV-2 spike deletion ΔH69/V70. bioRxiv. 2020 doi: 10.1101/2020.12.14.422555. published online Dec 15. (preprint).
    1. Bal A, Destras G, Gaymard A. Two-step strategy for the identification of SARS-CoV-2 variant of concern 202012/01 and other variants with spike deletion H69-V70, France, August to December 2020. medRxiv. 2021 doi: 10.1101/2020.11.10.20228528. published online Jan 11. (preprint).
    1. Public Health England Investigation of novel SARS-CoV-2 variant: variant of concern 202012/01. Technical briefing 5. Jan 14, 2021.
    1. Volz E, Mishra S, Chand M. Transmission of SARS-CoV-2 lineage B.1.1.7 in England: insights from linking epidemiological and genetic data. medRxiv. 2021 doi: 10.1101/2020.12.30.20249034. published online Jan 4. (preprint).
    1. Public Health England Investigation of novel SARS-CoV-2 variant: variant of concern 202012/01. Technical briefing 4. Jan 8, 2021.
    1. Kidd M, Richter A, Best A. S-variant SARS-CoV-2 is associated with significantly higher viral loads in samples tested by ThermoFisher TaqPath RT-QPCR. medRxiv. 2020 doi: 10.1101/2020.12.24.20248834. published online Dec 27. (preprint).
    1. Golubchik T, Lythgoe KA, Hall M. Early analysis of a potential link between viral load and the N501Y mutation in the SARS-COV-2 spike protein. medRxiv. 2021 doi: 10.1101/2021.01.12.20249080. published online Jan 15. (preprint).
    1. Leung K, Shum MH, Leung GM, Lam TT, Wu JT. Early transmissibility assessment of the N501Y mutant strains of SARS-CoV-2 in the United Kingdom, October to November 2020. Euro Surveill. 2021;26
    1. New and Emerging Respiratory Virus Threats Advisory Group NERVTAG paper on COVID-19 variant of concern B.1.1.7. Jan 22, 2021.
    1. Hall V, Foulkes S, Charlett A. Do antibody positive healthcare workers have lower SARS-CoV-2 infection rates than antibody negative healthcare workers? Large multi-centre prospective cohort study (the SIREN study), England: June to November 2020. medRxiv. 2021 doi: 10.1101/2021.01.13.21249642. published online Jan 15. (preprint).
    1. Harrington D, Kele B, Pereira S. Confirmed reinfection with SARS-CoV-2 variant VOC-202012/01. Clin Infect Dis. 2021 doi: 10.1093/cid/ciab014. published online Jan 9.
    1. Ju B, Zhang Q, Ge J. Human neutralizing antibodies elicited by SARS-CoV-2 infection. Nature. 2020;584:115–119.
    1. Haynes WA, Kamath K, Lucas C, Shon J, Iwasaki A. Impact of B.1.1.7 variant mutations on antibody recognition of linear SARS-CoV-2 epitopes. medRxiv. 2021 doi: 10.1101/2021.01.06.20248960. published online Jan 8. (preprint).
    1. Hu J, Peng P, Wang K. Emerging SARS-CoV-2 variants reduce neutralization sensitivity to convalescent sera and monoclonal antibodies. bioRxiv. 2021 doi: 10.1101/2021.01.22.427749. published online Jan 22. (preprint).
    1. Collier DA, Meng B, Ferreira IATM. Impact of SARS-CoV-2 B.1.1.7 spike variant on neutralisation potency of sera from individuals vaccinated with Pfizer vaccine BNT162b2. medRxiv. 2021 doi: 10.1101/2021.01.19.21249840. published online Jan 20. (preprint).
    1. Muik A, Wallisch A-K, Sänger B. Neutralization of SARS-CoV-2 lineage B.1.1.7 pseudovirus by BNT162b2 vaccine-elicited human sera. bioRxiv. 2021 doi: 10.1101/2021.01.18.426984. published online Jan 19. (preprint).
    1. Xie X, Zou J, Fontes-Garfias CR. Neutralization of N501Y mutant SARS-CoV-2 by BNT162b2 vaccine-elicited sera. bioRxiv. 2021 doi: 10.1101/2021.01.07.425740. published online Jan 7. (preprint).
    1. Shen X, Tang H, McDanal C. SARS-CoV-2 variant B.1.1.7 is susceptible to neutralizing antibodies elicited by ancestral spike vaccines. bioRxiv. 2021 doi: 10.1101/2021.01.27.428516. published online Jan 29. (preprint).
    1. Barrett JR, Belij-Rammerstorfer S, Dold C. Phase 1/2 trial of SARS-CoV-2 vaccine ChAdOx1 nCoV-19 with a booster dose induces multifunctional antibody responses. Nat Med. 2021;27:279–288.
    1. Sahin U, Muik A, Derhovanessian E. Concurrent human antibody and TH1 type T-cell responses elicited by a COVID-19 RNA vaccine. medRxiv. 2020 doi: 10.1101/2020.07.17.20140533. published online July 20. (preprint).
    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;383:2427–2438.
    1. Ewer KJ, Barrett JR, Belij-Rammerstorfer S. T cell and antibody responses induced by a single dose of ChAdOx1 nCoV-19 (AZD1222) vaccine in a phase 1/2 clinical trial. Nat Med. 2021;27:270–278.
    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. Ramasamy MN, Minassian AM, Ewer KJ. 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. Lythgoe KA, Hall M, Ferretti L. Within-host genomics of SARS-CoV-2. bioRxiv. 2020 doi: 10.1101/2020.05.28.118992. published online Dec 10. (preprint).
    1. Bonsall D, Golubchik T, de Cesare M. A comprehensive genomics solution for HIV surveillance and clinical monitoring in low-income settings. J Clin Microbiol. 2020;58:e00382–e00420.
    1. Wymant C, Blanquart F, Golubchik T. Easy and accurate reconstruction of whole HIV genomes from short-read sequence data with shiver. Virus Evol. 2018;4
    1. Katoh K, Misawa K, Kuma K, Miyata T. MAFFT: a novel method for rapid multiple sequence alignment based on fast Fourier transform. Nucleic Acids Res. 2002;30:3059–3066.
    1. Nguyen L-T, Schmidt HA, von Haeseler A, Minh BQ. IQ-TREE: a fast and effective stochastic algorithm for estimating maximum-likelihood phylogenies. Mol Biol Evol. 2015;32:268–274.
    1. Wibmer CK, Ayres F, Hermanus T. SARS-CoV-2 501Y.V2 escapes neutralization by South African COVID-19 donor plasma. bioRxiv. 2021 doi: 10.1101/2021.01.18.427166. published online Jan 19. (preprint).
    1. Wang P, Liu L, Iketani S. Increased resistance of SARS-CoV-2 variants B.1.351 and B.1.1.7 to antibody neutralization. bioRxiv. 2021 doi: 10.1101/2021.01.25.428137. published online Jan 26. (preprint).
    1. Wu K, Werner AP, Moliva JI. mRNA-1273 vaccine induces neutralizing antibodies against spike mutants from global SARS-CoV-2 variants. bioRxiv. 2021 doi: 10.1101/2021.01.25.427948. published online Jan 25. (preprint).
    1. Edara VV, Floyd K, Lai L. Infection and mRNA-1273 vaccine antibodies neutralize SARS-CoV-2 UK variant. medRxiv. 2021 doi: 10.1101/2021.02.02.21250799. published online Feb 5. (preprint).
    1. Supasa P, Zhou D, Dejnirattisai W. Reduced neutralization of SARS-CoV-2 B.1.1.7 variant by convalescent and vaccine sera. Cell. 2021 doi: 10.1016/j.cell.2021.02.033. published online Feb 18.
    1. WHO WHO R&D Blueprint: an international randomised trial of candidate vaccines against COVID-19. April 19, 2020.
    1. Voysey M, Costa Clemens SA, Madhi SA. 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. Lopez Bernal J, Andrews A, Gower C. Early effectiveness of COVID-19 vaccination with BNT162b2 mRNA vaccine and ChAdOx1 adenovirus vector vaccine on symptomatic disease, hospitalisations and mortality in older adults in England. medRxiv. 2021 doi: 10.1101/2021.03.01.21252652. published online March 2. (preprint).
    1. Tada T, Dcosta BM, Samanovic-Golden M. Neutralization of viruses with European, South African, and United States SARS-CoV-2 variant spike proteins by convalescent sera and BNT162b2 mRNA vaccine-elicited antibodies. bioRxiv. 2021 doi: 10.1101/2021.02.05.430003. published online Feb 7. (preprint).
    1. Madhi SA, Baillie VL, Cutland CL. Safety and efficacy of the ChAdOx1 nCoV-19 (AZD1222) Covid-19 vaccine against the B.1.351 variant in South Africa. medRxiv. 2021 doi: 10.1101/2021.02.10.21251247. published online Feb 12. (preprint).
    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. Mercado NB, Zahn R, Wegmann F. Single-shot Ad26 vaccine protects against SARS-CoV-2 in rhesus macaques. Nature. 2020;586:583–588.
    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. Mayers C, Baker K. Impact of false-positives and false-negatives in the UK's COVID-19 RT-PCR testing programme. June 3, 2020.
    1. Singanayagam A, Patel M, Charlett A. Duration of infectiousness and correlation with RT-PCR cycle threshold values in cases of COVID-19, England, January to May 2020. Euro Surveill. 2020;25
    1. Bullard J, Dust K, Funk D. Predicting infectious severe acute respiratory syndrome coronavirus 2 from diagnostic samples. Clin Infect Dis. 2020;71:2663–2666.
    1. van Kampen JJA, van de Vijver DAMC, Fraaij PLA. Duration and key determinants of infectious virus shedding in hospitalized patients with coronavirus disease-2019 (COVID-19) Nat Commun. 2021;12:267.
    1. Walsh KA, Spillane S, Comber L. The duration of infectiousness of individuals infected with SARS-CoV-2. J Infect. 2020;81:847–856.
    1. Qiu X, Nergiz AI, Maraolo AE, Bogoch II, Low N, Cevik M. Defining the role of asymptomatic and pre-symptomatic SARS-CoV-2 transmission—a living systematic review. Clin Microbiol Infect. 2021 doi: 10.1016/j.cmi.2021.01.011. published online Jan 20.

Source: PubMed

3
Subscribe