Risk prediction of covid-19 related death and hospital admission in adults after covid-19 vaccination: national prospective cohort study

Julia Hippisley-Cox, Carol Ac Coupland, Nisha Mehta, Ruth H Keogh, Karla Diaz-Ordaz, Kamlesh Khunti, Ronan A Lyons, Frank Kee, Aziz Sheikh, Shamim Rahman, Jonathan Valabhji, Ewen M Harrison, Peter Sellen, Nazmus Haq, Malcolm G Semple, Peter W M Johnson, Andrew Hayward, Jonathan S Nguyen-Van-Tam, Julia Hippisley-Cox, Carol Ac Coupland, Nisha Mehta, Ruth H Keogh, Karla Diaz-Ordaz, Kamlesh Khunti, Ronan A Lyons, Frank Kee, Aziz Sheikh, Shamim Rahman, Jonathan Valabhji, Ewen M Harrison, Peter Sellen, Nazmus Haq, Malcolm G Semple, Peter W M Johnson, Andrew Hayward, Jonathan S Nguyen-Van-Tam

Abstract

Objectives: To derive and validate risk prediction algorithms to estimate the risk of covid-19 related mortality and hospital admission in UK adults after one or two doses of covid-19 vaccination.

Design: Prospective, population based cohort study using the QResearch database linked to data on covid-19 vaccination, SARS-CoV-2 results, hospital admissions, systemic anticancer treatment, radiotherapy, and the national death and cancer registries.

Settings: Adults aged 19-100 years with one or two doses of covid-19 vaccination between 8 December 2020 and 15 June 2021.

Main outcome measures: Primary outcome was covid-19 related death. Secondary outcome was covid-19 related hospital admission. Outcomes were assessed from 14 days after each vaccination dose. Models were fitted in the derivation cohort to derive risk equations using a range of predictor variables. Performance was evaluated in a separate validation cohort of general practices.

Results: Of 6 952 440 vaccinated patients in the derivation cohort, 5 150 310 (74.1%) had two vaccine doses. Of 2031 covid-19 deaths and 1929 covid-19 hospital admissions, 81 deaths (4.0%) and 71 admissions (3.7%) occurred 14 days or more after the second vaccine dose. The risk algorithms included age, sex, ethnic origin, deprivation, body mass index, a range of comorbidities, and SARS-CoV-2 infection rate. Incidence of covid-19 mortality increased with age and deprivation, male sex, and Indian and Pakistani ethnic origin. Cause specific hazard ratios were highest for patients with Down's syndrome (12.7-fold increase), kidney transplantation (8.1-fold), sickle cell disease (7.7-fold), care home residency (4.1-fold), chemotherapy (4.3-fold), HIV/AIDS (3.3-fold), liver cirrhosis (3.0-fold), neurological conditions (2.6-fold), recent bone marrow transplantation or a solid organ transplantation ever (2.5-fold), dementia (2.2-fold), and Parkinson's disease (2.2-fold). Other conditions with increased risk (ranging from 1.2-fold to 2.0-fold increases) included chronic kidney disease, blood cancer, epilepsy, chronic obstructive pulmonary disease, coronary heart disease, stroke, atrial fibrillation, heart failure, thromboembolism, peripheral vascular disease, and type 2 diabetes. A similar pattern of associations was seen for covid-19 related hospital admissions. No evidence indicated that associations differed after the second dose, although absolute risks were reduced. The risk algorithm explained 74.1% (95% confidence interval 71.1% to 77.0%) of the variation in time to covid-19 death in the validation cohort. Discrimination was high, with a D statistic of 3.46 (95% confidence interval 3.19 to 3.73) and C statistic of 92.5. Performance was similar after each vaccine dose. In the top 5% of patients with the highest predicted covid-19 mortality risk, sensitivity for identifying covid-19 deaths within 70 days was 78.7%.

Conclusion: This population based risk algorithm performed well showing high levels of discrimination for identifying those patients at highest risk of covid-19 related death and hospital admission after vaccination.

Conflict of interest statement

Competing interests: All authors have completed the ICMJE uniform disclosure form at www.icmje.org/disclosure-of-interest/ and declare: support from the NIHR for the submitted work; JH-C reports grants from NIHR Biomedical Research Centre, Oxford, grants from John Fell Oxford University Press Research Fund, grants from Cancer Research UK, through the Cancer Research UK Oxford Centre, grants from the Oxford Wellcome Institutional Strategic Support Fund and other research councils, during the conduct of the study; JH-C is an unpaid director of QResearch, a not-for-profit organisation that is a partnership between the University of Oxford and EMIS Health, which supplied the QResearch database used for this work; JH-C is a founder and shareholder of ClinRisk and was its medical director until 31 May 2019 (ClinRisk produces open and closed source software to implement clinical risk algorithms (outside this work) into clinical computer systems); JH-C is chair of the NERVTAG risk stratification subgroup and a member of SAGE covid-19 groups and the NHS group advising on prioritisation of use of monoclonal antibodies in covid-19 infection; CACC reports receiving personal fees from ClinRisk outside this work, and is a member of the NERVTAG risk stratification subgroup; KK is supported by the NIHR Applied Research Collaboration-East Midlands and the Leicester BRC and is a member of SAGE; RHK was supported by a UKRI Future Leaders Fellowship (MR/S017968/1); KD-O was supported by a grant from the Alan Turing Institute Health Programme (EP/T001569/1); AS is a member of the Scottish Government Chief Medical Officer’s covid-19 advisory group and a member of AstraZeneca’s thrombotic thrombocytopenic advisory group (both roles are unremunerated); RAL is a member of the Welsh government covid-19 technical advisory group (unrenumerated); MGS reports grants from Department of Health and Social Care NIHR UK, grants from the UK Medical Research Council, grants from Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, during the conduct of the study, and other funds from Integrum Scientific, Greensboro, NC, USA, outside the submitted work; MGS is a member of NERVTAG and attends SAGE covid-19; AH is a member of NERVTAG and the NHS group advising on prioritisation of use of monoclonal antibodies in covid-19 infection; JV is national clinical director for diabetes and obesity at NHS England and Improvement; FK is a member of the Northern Ireland Chief Medical Officer’s pandemic modelling group and strategic intelligence group; JSN-V-T is seconded to the Department of Health and Social Care, England. The views expressed in this manuscript are those of the authors and not necessarily those of Department of Health and Social Care or the UK government.

© Author(s) (or their employer(s)) 2019. Re-use permitted under CC BY. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Fig 1
Fig 1
Use of QCovid3 model showing adjusted cause specific hazard ratios for covid-19 death after vaccination, mutually adjusted and adjusted for fractional polynomial terms for age, body mass index, vaccination dose, and background infection rate at time of vaccination. CKD=chronic kidney disease; HbA1c=glycated haemogoblin
Fig 2
Fig 2
Use of QCovid3 model showing adjusted cause specific hazard ratios for covid-19 hospital admission after vaccination, mutually adjusted and adjusted for fractional polynomial terms for age, body mass index, vaccination dose, and background infection rate at time of vaccination. CKD=chronic kidney disease; HbA1c=glycated haemogoblin
Fig 3
Fig 3
Calibration of the QCovid3 risk model to predict covid-19 related death after vaccination. Data source: QResearch England, 8 December 2020 to 15 June 2021, https://www.qresearch.org/
Fig 4
Fig 4
Calibration of the QCovid3 risk model to predict covid-19 related hospital admission after vaccination. Data source: QResearch England, 8 December 2020 to 15 June 2021, https://www.qresearch.org/

References

    1. Clift AK, Coupland CAC, Keogh RH, et al. . Living risk prediction algorithm (QCOVID) for risk of hospital admission and mortality from coronavirus 19 in adults: national derivation and validation cohort study. BMJ 2020;371:m3731. 10.1136/bmj.m3731.
    1. Nafilyan V, Humberstone B, Mehta N, et al. . An external validation of the QCovid risk prediction algorithm for risk of mortality from COVID-19 in adults: a national validation cohort study in England. Lancet Digit Health 2021;3:e425-33. 10.1016/S2589-7500(21)00080-7.
    1. NHS Digital. Coronavirus Shielded Patient List open data set, England 2021. .
    1. Voysey M, Clemens SAC, Madhi SA, et al. Oxford COVID Vaccine Trial Group . 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. Polack FP, Thomas SJ, Kitchin N, et al. C4591001 Clinical Trial Group . Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N Engl J Med 2020;383:2603-15. 10.1056/NEJMoa2034577.
    1. Baden LR, El Sahly HM, Essink B, et al. COVE Study Group . Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N Engl J Med 2021;384:403-16. 10.1056/NEJMoa2035389.
    1. Lim SH, Campbell N, Johnson M, et al. . Antibody responses after SARS-CoV-2 vaccination in patients with lymphoma. Lancet Haematol 2021;8:e542-4. 10.1016/S2352-3026(21)00199-X.
    1. Clift AK, Coupland CAC, Keogh RH, Hemingway H, Hippisley-Cox J. COVID-19 Mortality Risk in Down Syndrome: Results From a Cohort Study of 8 Million Adults. Ann Intern Med 2021;174:572-6.
    1. Hippisley-Cox J, Young D, Coupland C, et al. . Risk of severe COVID-19 disease with ACE inhibitors and angiotensin receptor blockers: cohort study including 8.3 million people. Heart 2020;106:1503-11. 10.1136/heartjnl-2020-317393.
    1. Hippisley-Cox J, Coupland C. Risk of myocardial infarction in patients taking cyclo-oxygenase-2 inhibitors or conventional non-steroidal anti-inflammatory drugs: population based nested case-control analysis. BMJ 2005;330:1366-9. 10.1136/bmj.330.7504.1366.
    1. UK Government. COVID-19 deaths in the UK. 2921 2021.
    1. Hippisley-Cox J, Clift AK, Coupland CAC, et al. Protocol for the development and evaluation of a tool for predicting risk of short-term adverse outcomes due to COVID-19 in the general UK population. medRxiv 2020:2020.06.28.20141986-2020.06.28.86. 10.1101/2020.06.28.20141986
    1. Gao M, Piernas C, Astbury NM, et al. . Associations between body-mass index and COVID-19 severity in 6·9 million people in England: a prospective, community-based, cohort study. Lancet Diabetes Endocrinol 2021;9:350-9. 10.1016/S2213-8587(21)00089-9.
    1. Townsend P, Davidson N. The Black report. Penguin, 1982.
    1. UK Government. Coronar (COVID-19) in the UK downloaded data 2021. .
    1. Dafni U. Landmark analysis at the 25-year landmark point. Circ Cardiovasc Qual Outcomes 2011;4:363-71. 10.1161/CIRCOUTCOMES.110.957951.
    1. Little RJA, Rubin DB. Statistical analysis with missing data/Roderick J.A. Little, Donald B. Rubin. 2nd ed 2002. Wiley, c, 200210.1002/9781119013563.
    1. Kalbfleisch J, Prentice R. The Statistical Analysis of Failure Time Data. 2002:114-810.1002/9781118032985.
    1. Wolbers M, Blanche P, Koller MT, Witteman JC, Gerds TA. Concordance for prognostic models with competing risks. Biostatistics 2014;15:526-39. 10.1093/biostatistics/kxt059.
    1. Royston P. Explained variation for survival models. Stata J 2006;6:1-1410.1177/1536867X0600600105.
    1. Coviello V, Boggess M. Cumulative incidence estimation in the presence of competing risks. Stata J 2004;4:103-1210.1177/1536867X0400400201.
    1. Collins GS, Reitsma JB, Altman DG, Moons KG. Transparent Reporting of a multivariable prediction model for Individual Prognosis or Diagnosis (TRIPOD): the TRIPOD statement. Ann Intern Med 2015;162:55-63. 10.7326/M14-0697.
    1. Wall EC, Wu M, Harvey R, et al. . Neutralising antibody activity against SARS-CoV-2 VOCs B.1.617.2 and B.1.351 by BNT162b2 vaccination. Lancet 2021;397:2331-3. 10.1016/S0140-6736(21)01290-3.
    1. Hoes J, Boef AGC, Knol MJ, et al. . Socioeconomic Status Is Associated With Antibody Levels Against Vaccine Preventable Diseases in the Netherlands. Front Public Health 2018;6:209. 10.3389/fpubh.2018.00209.
    1. Zimmermann P, Curtis N. Factors That Influence the Immune Response to Vaccination. Clin Microbiol Rev 2019;32:e00084-18. 10.1128/CMR.00084-18.
    1. Baker RJ, Mark PB, Patel RK, Stevens KK, Palmer N. Renal association clinical practice guideline in post-operative care in the kidney transplant recipient. BMC Nephrol 2017;18:174. 10.1186/s12882-017-0553-2.
    1. Garthwaite E, Reddy V, Douthwaite S, Lines S, Tyerman K, Eccles J. Clinical practice guideline management of blood borne viruses within the haemodialysis unit. BMC Nephrol 2019;20:388. 10.1186/s12882-019-1529-1.
    1. Stevens CE, Alter HJ, Taylor PE, Zang EA, Harley EJ, Szmuness W. Hepatitis B vaccine in patients receiving hemodialysis. Immunogenicity and efficacy. N Engl J Med 1984;311:496-501. 10.1056/NEJM198408233110803.
    1. Cavdar C, Sayan M, Sifil A, et al. . The comparison of antibody response to influenza vaccination in continuous ambulatory peritoneal dialysis, hemodialysis and renal transplantation patients. Scand J Urol Nephrol 2003;37:71-6. 10.1080/00365590310008749.
    1. Remschmidt C, Wichmann O, Harder T. Influenza vaccination in patients with end-stage renal disease: systematic review and assessment of quality of evidence related to vaccine efficacy, effectiveness, and safety. BMC Med 2014;12:244-44. 10.1186/s12916-014-0244-9.
    1. Kennedy NA, Lin S, Goodhand JR, et al. . Infliximab is associated with attenuated immunogenicity to BNT162b2 and ChAdOx1 nCoV-19 SARS-CoV-2 vaccines. medRxiv 2021:2021.03.25.21254335. 10.1101/2021.03.25.21254335
    1. Katikireddi SV, Lal S, Carrol ED, et al. . Unequal impact of the COVID-19 crisis on minority ethnic groups: a framework for understanding and addressing inequalities. J Epidemiol Community Health 2021;75:970-4. 10.1136/jech-2020-216061.
    1. Sattar N, McInnes IB, McMurray JJV. Obesity Is a Risk Factor for Severe COVID-19 Infection: Multiple Potential Mechanisms. Circulation 2020;142:4-6. 10.1161/CIRCULATIONAHA.120.047659.
    1. Pike MM, Decker PA, Larson NB, et al. . Improvement in Cardiovascular Risk Prediction with Electronic Health Records. J Cardiovasc Transl Res 2016;9:214-22. 10.1007/s12265-016-9687-z.
    1. Kengne AP, Beulens JW, Peelen LM, et al. . Non-invasive risk scores for prediction of type 2 diabetes (EPIC-InterAct): a validation of existing models. Lancet Diabetes Endocrinol 2014;2:19-29. 10.1016/S2213-8587(13)70103-7.
    1. Hippisley-Cox J, Coupland C, Brindle P. Development and validation of QRISK3 risk prediction algorithms to estimate future risk of cardiovascular disease: prospective cohort study. BMJ 2017;357:j2099. 10.1136/bmj.j2099.
    1. Hippisley-Cox J, Coupland C. Development and validation of QDiabetes-2018 risk prediction algorithm to estimate future risk of type 2 diabetes: cohort study. BMJ 2017;359:j5019. 10.1136/bmj.j5019.
    1. Hippisley-Cox J, Coupland C. Development and validation of QMortality risk prediction algorithm to estimate short term risk of death and assess frailty: cohort study. BMJ 2017;358:j4208. 10.1136/bmj.j4208.
    1. Patone M, Thomas KRH, et al. . Mortality and Critical Care Unit admission associated with the SARS-CoV-2 Variant of Concern 202012/01 in England: an observational cohort study. Lancet Infect Dis 2021; [forthcoming] 10.1016/S1473-3099(21)00318-2.
    1. Collins GS, Altman DG. An independent external validation and evaluation of QRISK cardiovascular risk prediction: a prospective open cohort study. BMJ 2009;339:b2584. 10.1136/bmj.b2584.
    1. Collins GS, Altman DG. An independent and external validation of QRISK2 cardiovascular disease risk score: a prospective open cohort study. BMJ 2010;340:c2442. 10.1136/bmj.c2442.
    1. Collins GS, Altman DG. Predicting the 10 year risk of cardiovascular disease in the United Kingdom: independent and external validation of an updated version of QRISK2. BMJ 2012;344:e4181. 10.1136/bmj.e4181.

Source: PubMed

3
Se inscrever