Symptom Burden and Immune Dynamics 6 to 18 Months Following Mild Severe Acute Respiratory Syndrome Coronavirus 2 Infection (SARS-CoV-2): A Case-control Study

Elisabeth B Fjelltveit, Bjørn Blomberg, Kanika Kuwelker, Fan Zhou, Therese B Onyango, Karl A Brokstad, Rebecca Elyanow, Ian M Kaplan, Camilla Tøndel, Kristin G I Mohn, Türküler Özgümüş, Rebecca J Cox, Nina Langeland, Bergen COVID-19 Research Group, Geir Bredholt, Lena Hansen, Sarah Larteley Lartey, Anders Madsen, Jan Stefan Olofsson, Sonja Ljostveit, Marianne Sævik, Hanne Søyland, Helene Heitmann Sandnes, Nina Urke Ertesvåg, Juha Vahokoski, Amit Bansal, Håkon Amdam, Tatiana Fomina, Dagrun Waag Linchausen, Synnøve Hauge, Annette Corydon, Silje Sundøy, Elisabeth B Fjelltveit, Bjørn Blomberg, Kanika Kuwelker, Fan Zhou, Therese B Onyango, Karl A Brokstad, Rebecca Elyanow, Ian M Kaplan, Camilla Tøndel, Kristin G I Mohn, Türküler Özgümüş, Rebecca J Cox, Nina Langeland, Bergen COVID-19 Research Group, Geir Bredholt, Lena Hansen, Sarah Larteley Lartey, Anders Madsen, Jan Stefan Olofsson, Sonja Ljostveit, Marianne Sævik, Hanne Søyland, Helene Heitmann Sandnes, Nina Urke Ertesvåg, Juha Vahokoski, Amit Bansal, Håkon Amdam, Tatiana Fomina, Dagrun Waag Linchausen, Synnøve Hauge, Annette Corydon, Silje Sundøy

Abstract

Background: The burden and duration of persistent symptoms after nonsevere coronavirus disease 2019 (COVID-19) remains uncertain. This study aimed to assess postinfection symptom trajectories in home-isolated COVID-19 cases compared with age- and time- matched seronegative controls, and investigate immunological correlates of long COVID.

Methods: A prospective case-control study included home-isolated COVID-19 cases between February 28 and April 4, 2020, and followed for 12 (n = 233) to 18 (n = 149) months, and 189 age-matched severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-naive controls. We collected clinical data at baseline, 6, 12, and 18 months postinfection, and blood samples at 2, 4, 6, and 12 months for analysis of SARS-CoV-2-specific humoral and cellular responses.

Results: Overall, 46% (108/233) had persisting symptoms 12 months after COVID-19. Compared with controls, adult cases had a high risk of fatigue (27% excess risk, sex, and comorbidity adjusted odds ratio [aOR] 5.86; 95% confidence interval [CI], 3.27-10.5), memory problems (21% excess risk; aOR 7.42; CI, 3.51-15.67), concentration problems (20% excess risk; aOR 8.88; 95% CI, 3.88-20.35), and dyspnea (10% excess risk; aOR 2.66; 95% CI, 1.22-5.79). The prevalence of memory problems increased overall from 6 to 18 months (excess risk 11.5%; 95% CI, 1.5-21.5; P = .024) and among women (excess risk 18.7%; 95% CI, 4.4-32.9; P = .010). Longitudinal spike immunoglobulin G was significantly associated with dyspnea at 12 months. The spike-specific clonal CD4+ T-cell receptor β depth was significantly associated with both dyspnea and number of symptoms at 12 months.

Conclusions: This study documents a high burden of persisting symptoms after mild COVID-19 and suggests that infection induced SARS-CoV-2-specific immune responses may influence long-term symptoms.

Keywords: PASC; SARS CoV-2; T-cells; antibodies; long COVID.

Conflict of interest statement

Potential conflicts of interest. R.C. declares receipt of a research grant to the University of Bergen from Trond Mohn Stiftelse (TMS2020TMT05) and from the Helse-Vest to Haukeland University Hospital (F11628). N.L. declares receipt of a research grant from the Regional Health Authority of Western Norway and reports roles as Member of Faculty Board, NTNU, Trondheim, Norway, and as Member of the Board, Stavanger University Hospital, Stavanger, Norway. K.M. declares receipt of research grant from Helse-Vest (F12626). R.E. and I.M.K. declare employment and equity ownership with Adaptive Biotechnologies. The remaining authors: No reported conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest.

The Author(s) 2022. Published by Oxford University Press on behalf of Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Study population. Inclusion of SARS-CoV-2 cases (left) and control group (right). Eligible participants tested for SARS-CoV-2 infection by RT-PCR at Bergen Municipality Emergency Clinic (BMEC) were recruited between February 28 and April 4, 2020. Only 1 case (the first most symptomatic) from each household was tested because of the limited testing capacity; thus, individuals living with COVID-19-positive study participants were included as household contacts. If household contact had positive SARS-CoV-2 serology (RBD and spike-IgG ELISA) within 2 months after recruitment, they were registered as cases. Seronegative household contact without a history of COVID-19 symptoms were included as controls. Additional controls were recruited amongst individuals who were prioritized for vaccination, either because of their age, comorbidity, or occupation. At the time of symptom recording, all controls were confirmed seronegative. COVID-19, coronavirus disease 2019; ELISA, enzyme-linked immunosorbent assay; LTF,lost to follow-up; RBD, receptor-binding domain; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.
Figure 2.
Figure 2.
Age-stratified symptom prevalence at 12 months after infection. Bar plot representing the proportion of cases reporting 11 key symptoms at 12 months’ follow-up. The cases reported a mean of 1.4 symptoms overall. The age group 0 to 15 years old (n = 13) is not shown because of absence of symptoms. The light gray area in the bar charts represents the overall proportion with any of the 11 symptoms in the current age group. The colored areas represent the proportion with the specified symptoms.
Figure 3.
Figure 3.
Longitudinal symptom changes up to 18 months after infection. Dumbbell charts present longitudinal data on development of 11 specified symptoms in a subcohort of patients followed for 18 months (n = 148, 1 patient was excluded because of missing data on all symptoms at 6 months). (Left panel) The overall symptom change from 6 to 12 months, (Middle panel) the overall symptom change from 6 to 18 months, and (Right panel) the symptom change in men (n = 73) and women (n = 75) from 6 to 18 months.
Figure 4.
Figure 4.
Kinetics of the spike IgG antibody response in relation to symptoms at 6 and 12 months. The relationship between longitudinal antibody titers and (A) persistent dyspnea versus no dyspnea, (B) 3 or more symptoms at 12 months vs no symptoms, and (C) persistent fatigue vs no fatigue. The generalized estimating equation (GEE) coefficients with 95% confidence intervals (CI) are adjusted for age, sex, comorbidity, and time of measurement. All cases who had been vaccinated against SARS-CoV-2 during the follow-up period (n = 20) were excluded from the analysis of immunological parameters at 12 months. IgG, immunoglobulin G; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.

References

    1. Prevention CfDCa . Post COVID Conditions. Available at: .
    1. Excellence NIfHaC . COVID-19 rapid guideline: managing the long-term effects of COVID-19. Available at: .
    1. Nalbandian A, Sehgal K, Gupta A, et al. . Post-acute COVID-19 syndrome. Nat Med 2021; 27:601–15.
    1. World Health Organization . A clinical case definition of post COVID-19 condition by a Delphi consensus, 6 October 2021 Available at: .
    1. Nanwani-Nanwani K, López-Pérez L, Giménez-Esparza C, et al. . Prevalence of post-intensive care syndrome in mechanically ventilated patients with COVID-19. Scientific Reports 2022; 12:1–11.
    1. Rousseau AF, Minguet P, Colson C, et al. . Post-intensive care syndrome after a critical COVID-19: cohort study from a Belgian follow-up clinic. Ann Intensive Care 2021; 11:118.
    1. Fumagalli C, Zocchi C, Tassetti L, et al. . Factors associated with persistence of symptoms 1 year after COVID-19: a longitudinal, prospective phone-based interview follow-up cohort study. Eur J Int Med 2022; 97:36–41.
    1. Huang L, Yao Q, Gu X, et al. . 1-year outcomes in hospital survivors with COVID-19: a longitudinal cohort study. The Lancet 2021; 398:747–58.
    1. Bellan M, Baricich A, Patrucco F, et al. . Long-term sequelae are highly prevalent one year after hospitalization for severe COVID-19. Sci Rep 2021; 11:1–10.
    1. Zhang X, Wang F, Shen Y, et al. . Symptoms and health outcomes among survivors of COVID-19 infection 1 year after discharge from hospitals in Wuhan, China. JAMA Network open 2021; 4:e2127403-e.
    1. Kim Y, Bitna H, Kim SW, et al. . Post-acute COVID-19 syndrome in patients after 12 months from COVID-19 infection in Korea. BMC Infect Dis 2022; 22:93.
    1. Boscolo-Rizzo P, Guida F, Polesel J, et al. . Sequelae in adults at 12 months after mild-to-moderate coronavirus disease 2019 (COVID-19). Int Forum Allergy Rhinol 2021; 11:1685–8.
    1. Seessle J, Waterboer T, Hippchen T, et al. . Persistent symptoms in adult patients one year after COVID-19: a prospective cohort study. Clin Infect Dis 2021; 16:e0256142.
    1. Huang L, Li X, Gu X, et al. . Health outcomes in people 2 years after surviving hospitalisation with COVID-19: a longitudinal cohort study. Lancet Respir Med 2022; 10:863–76.
    1. Helmsdal G, Hanusson KD, Kristiansen MF, et al. . Long COVID in the long run-23 months follow-up study of persistent symptoms. Open Forum Infect Dis 2022; 9:ofac270. Oxford University Press.
    1. Groff D, Sun A, Ssentongo AE, et al. . Short-term and long-term rates of postacute sequelae of SARS-CoV-2 infection: a systematic review. JAMA Network Open 2021; 4:e2128568-e.
    1. Berg SK, Palm P, Nygaard U, et al. . Long COVID symptoms in SARS-CoV-2-positive children aged 0–14 years and matched controls in Denmark (LongCOVIDKidsDK): a national, cross-sectional study. Lancet Child Adoles Health 2022; 6:614–23.
    1. Havervall S, Rosell A, Phillipson M, et al. . Symptoms and functional impairment assessed 8 months after mild COVID-19 among health care workers. JAMA 2021; 325:2015–6.
    1. Kim Y, Kim S-W, Chang H-H, Kwon KT, Bae S, Hwang S. Post-acute COVID-19 syndrome in patients after 12 months from COVID-19 infection in Korea. BMC Infect Dis 2022; 22:1–12.
    1. Lund LC, Hallas J, Nielsen H, et al. . Post-acute effects of SARS-CoV-2 infection in individuals not requiring hospital admission: a Danish population-based cohort study. Lancet Infect Dis 2021; 21:1373–82.
    1. Xie Y, Bowe B, Al-Aly Z. Burdens of post-acute sequelae of COVID-19 by severity of acute infection, demographics and health status. Nat Commun 2021; 12:6571.
    1. Blomberg B, Mohn KG, Brokstad KA, et al. . Long COVID in a prospective cohort of home-isolated patients. Nat Med 2021; 27:1607–13.
    1. Havervall S, Jernbom Falk A, Klingstrom J, et al. . SARS-CoV-2 induces a durable and antigen specific humoral immunity after asymptomatic to mild COVID-19 infection. PloS One 2022; 17:e0262169.
    1. Garcia-Abellan J, Padilla S, Fernandez-Gonzalez M, et al. . Antibody response to SARS-CoV-2 is associated with long-term clinical outcome in patients with COVID-19: a longitudinal study. J Clin Immunol 2021; 41:1490–501.
    1. Peluso MJ, Deitchman AN, Torres L, et al. . Long-term SARS-CoV-2-specific immune and inflammatory responses in individuals recovering from COVID-19 with and without post-acute symptoms. Cell Rep 2021; 36:109518.
    1. Phetsouphanh C, Darley DR, Wilson DB, et al. . Immunological dysfunction persists for 8 months following initial mild-to-moderate SARS-CoV-2 infection. Nat Immunol 2022; 23:210–6.
    1. Kuwelker K, Zhou F, Blomberg B, et al. . Attack rates amongst household members of outpatients with confirmed COVID-19 in Bergen, Norway: a case-ascertained study. Lancet Reg Health Eur 2021; 3:100014.
    1. Chalder T, Berelowitz G, Pawlikowska T, et al. . Development of a fatigue scale. J Psychosom Res 1993; 37:147–53.
    1. Trieu MC, Bansal A, Madsen A, et al. . SARS-CoV-2-Specific neutralizing antibody responses in Norwegian health care workers after the first wave of COVID-19 pandemic: a prospective cohort study. J Infect Dis 2021; 223:589–99.
    1. Snyder TM, Gittelman RM, Klinger M, et al. . Magnitude and dynamics of the T-cell response to SARS-CoV-2 infection at both individual and population levels. medRxiv 2020.Version 3. Preprint. NaN NaN [revised 2020 Sep 17]. doi: 10.1101/2020.07.31.20165647
    1. Tan W, Lu Y, Zhang J, et al. . Viral kinetics and antibody responses in patients with COVID-19. medRxiv 2020. doi: 10.1101/2020.03.24.20042382
    1. Taquet M, Dercon Q, Luciano S, Geddes JR, Husain M, Harrison PJ. Incidence, co-occurrence, and evolution of long-COVID features: a 6-month retrospective cohort study of 273,618 survivors of COVID-19. PLoS medicine 2021; 18:e1003773.
    1. Tran V-T, Porcher R, Pane I, Ravaud P. Course of post COVID-19 disease symptoms over time in the ComPaRe long COVID prospective e-cohort. Nat Commun 2022; 13:1812.
    1. Peghin M, Palese A, Venturini M, et al. . Post-COVID-19 symptoms 6 months after acute infection among hospitalized and non-hospitalized patients. Clin Microbiol Infect 2021; 27:1507–13.
    1. Kaplonek P, Fischinger S, Cizmeci D, et al. . mRNA-1273 vaccine-induced antibodies maintain Fc effector functions across SARS-CoV-2 variants of concern. Immunity 2022; 55:355–65 e4.
    1. Chang SE, Feng A, Meng W, et al. . New-onset IgG autoantibodies in hospitalized patients with COVID-19. Nat Commun 2021; 12:5417.
    1. Lucchese G, Flöel A. SARS-CoV-2 and Guillain-Barre syndrome: molecular mimicry with human heat shock proteins as potential pathogenic mechanism. Cell Stress Chaperones 2020; 25:731–5.
    1. Borsche M, Reichel D, Fellbrich A, et al. . Persistent cognitive impairment associated with cerebrospinal fluid anti-SARS-CoV-2 antibodies six months after mild COVID-19. Neurol Res Pract 2021; 3:34.
    1. Apple AC, Oddi A, Peluso MJ, et al. . Risk factors and abnormal cerebrospinal fluid associate with cognitive symptoms after mild COVID-19. Ann Clin Transl Neurol 2022; 9:221–6.
    1. Douaud G, Lee S, Alfaro-Almagro F, et al. . SARS-CoV-2 is associated with changes in brain structure in UK Biobank. Nature 2022; 604:697–707.

Source: PubMed

3
Se inscrever