Skip to main content
Erschienen in: BMC Medicine 1/2022

Open Access 01.12.2022 | COVID-19 | Correspondence

Sustained seropositivity up to 20.5 months after COVID-19

verfasst von: Carlota Dobaño, Anna Ramírez-Morros, Selena Alonso, Rocío Rubio, Gemma Ruiz-Olalla, Josep Vidal-Alaball, Dídac Macià, Queralt Miró Catalina, Marta Vidal, Aina Fuster Casanovas, Esther Prados de la Torre, Diana Barrios, Alfons Jiménez, Jasmina Zanoncello, Natalia Rodrigo Melero, Carlo Carolis, Luis Izquierdo, Ruth Aguilar, Gemma Moncunill, Anna Ruiz-Comellas

Erschienen in: BMC Medicine | Ausgabe 1/2022

Abstract

This study evaluated the persistence of IgM, IgA, and IgG to SARS-CoV-2 spike and nucleocapsid antigens up to 616 days since the onset of symptoms in a longitudinal cohort of 247 primary health care workers from Barcelona, Spain, followed up since the start of the pandemic. The study also assesses factors affecting antibody levels, including comorbidities and the responses to variants of concern as well as the frequency of reinfections. Despite a gradual and significant decline in antibody levels with time, seropositivity to five SARS-CoV-2 antigens combined was always higher than 90% over the whole study period. In a subset of 23 participants who had not yet been vaccinated by November 2021, seropositivity remained at 95.65% (47.83% IgM, 95.65% IgA, 95.65% IgG). IgG seropositivity against Alpha and Delta predominant variants was comparable to that against the Wuhan variant, while it was lower for Gamma and Beta (minority) variants and for IgA and IgM. Antibody levels at the time point closest to infection were associated with age, smoking, obesity, hospitalization, fever, anosmia/hypogeusia, chest pain, and hypertension in multivariable regression models. Up to 1 year later, just before the massive roll out of vaccination, antibody levels were associated with age, occupation, hospitalization, duration of symptoms, anosmia/hypogeusia, fever, and headache. In addition, tachycardia and cutaneous symptoms associated with slower antibody decay, and oxygen supply with faster antibody decay. Eight reinfections (3.23%) were detected in low responders, which is consistent with a sustained protective role for anti-spike naturally acquired antibodies. Stable persistence of IgG and IgA responses and cross-recognition of the predominant variants circulating in the 2020–2021 period indicate long-lasting and largely variant-transcending humoral immunity in the initial 20.5 months of the pandemic, in the absence of vaccination.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12916-022-02570-3.
Carlota Dobaño and Anna Ramírez-Morros have shared authorship.
Gemma Moncunill and Anna Ruiz-Comella have shared authorship.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
S
Spike
RBD
Receptor-binding domain
HCW
Health care workers
BSA
Bovine serum albumin
COPD
Chronic obstructive pulmonary disease
FL
Full length
PBS
Phosphate buffered saline
MFI
Median fluorescence intensity
PE
Phycoerythrin
RDTs
Rapid diagnostic tests
RT-qPCR
Reverse transcription quantitative polymerase chain reaction
T
Timepoint
VoC
Variants of concern

Introduction

The maintenance and effectiveness of adaptive immunity directed against SARS-CoV-2 after primary infection are key questions in understanding and controlling the COVID-19 pandemic and any future emerging new coronavirus threat. Despite the global start of vaccination campaigns by the end of 2020, a substantial percentage of the world’s population remains unvaccinated, and their capacity to resist infections relies only on naturally acquired immunity. We have previously shown that 90% of those infected with SARS-CoV-2 remain seropositive 1 year after discharge [1, 2]. To our knowledge, the duration of antibody responses following natural infection has not been assessed beyond 13-20 months to date [310].
SARS-CoV-2 elicits robust humoral immune responses, including production of virus-specific immunoglobulin M (IgM), IgA, and IgG. IgM and IgA isotypes dominate the early effector antibody response to SARS-CoV-2, and IgA greatly contributes to virus neutralization at mucosal sites [11, 12]. In serum, the three isotypes display neutralizing activity, with IgM and IgG1 (predominant subclass of IgG) being the most important contributors [13].
Reinfection and COVID-19 disease rates, including severe cases, may increase if immunity wanes in those who do not get vaccinated. The emergence of SARS-CoV-2 variants of concern (VoC) with high transmissibility and potentially lower susceptibility to antibodies has raised the question of whether antibodies induced by the original Wuhan strain will still protect against reinfections or only against severe COVID-19 [14]. Therefore, data on the long-term persistence and efficacy of the immune response is of vital importance to foresee the evolution of the COVID-19 pandemic especially with more contagious emerging variants like Delta and Omicron [1518]. Data could also be useful to infer the potential duration of vaccine-elicited immunity, which started to be studied a year after the onset of the pandemic.
There is a wide heterogeneity in how individuals respond to SARS-CoV-2 infection in terms of type and potency of immune responses, resulting in diverse viral and clinical presentations and susceptibilities. Systematic reviews and meta-analyses have concluded that men, those over 65 years of age, smokers, and patients with comorbidities such as hypertension, diabetes, cardiovascular disease, cerebrovascular disease, chronic obstructive pulmonary disease (COPD), chronic kidney disease, and cancer, contribute significantly to disease severity and COVID-19 prognostic [1927]. However, thus far, very few studies have assessed the effect of comorbidities on SARS-CoV-2 immune responses, including antibodies that mediate neutralizing protective effector functions [28, 29]. Furthermore, it is also likely that individuals also vary in their capacity to maintain protective antibody responses in time, and the factors determining humoral immune memory are not known.
The main objectives of this study were to evaluate the kinetics of anti-SARS-CoV-2 antibodies over a period of 20.5 months in convalescent unvaccinated individuals from a well-characterized longitudinal cohort of health care workers (HCW) (CoviCatCentral), to assess the effect of clinical and demographic variables on the antibody levels, and to estimate the prevalence of reinfections.

Methods

Study design and subjects

Two hundred forty-seven HCW presenting with COVID-19 in three primary care counties in Barcelona, Spain, were recruited in a prospective cohort from March 2020 [1] and followed up during 2021, with sample collection performed at different time points (T) per individual: T0, July–August 2020; T1, September 2020; T2, October 2020; T3, November 2020; T4, January-February 2021; T5, March–April 2021; T6, May–June 2021; T7, July 2021; and T8, November 2021. Infections were detected by antigen rapid diagnostic tests (RDTs) and quantitative reverse transcription polymerase chain reaction (RT-qPCR) performed on participants with symptoms of COVID-19 or who had been in close contact with someone with SARS-CoV-2 infection. Overall, primary infections occurred between pre-T0 and T4. The effect of baseline characteristics on the anti-SARS-CoV-2 antibody response 1 year after the onset of the pandemic has already been reported [1] except for comorbidities and other risk factors that are addressed here: chronic kidney disease, COPD, asthma, cardiovascular disease, neurological diseases, digestive diseases, autoimmune diseases, cancer, immunosuppression (disease or drug-related), obesity, pregnancy, diabetes mellitus, dyslipidemia, hypertension, depression and/or anxiety, and hypothyroidism. Anti-SARS-CoV-2 serologic testing was performed at nine cross-sectional visits, and data on those not being vaccinated by mid-November 2021 is analyzed here. The later visits included in this analysis were T6 (May-June 2021), T7 (July 2021), and T8 (November 2021). The baseline (T0, July–August 2020) sample was obtained from the SeroCatCentral/VisCat study. T6 (N = 72) included 22 physicians or dentists, 35 nurses, and 15 with other job categories like customer and social services staff, with median (IQR) age of 45 (13) years and 86.3% being women; T7 (N = 39) included 11 physicians/dentists, 21 nurses, and 7 others, with median (IQR) age of 48 (13) years and 87.2% women; T8 (N = 23) included 4 physicians/dentists, 13 nurses, and 6 others, with median (IQR) age of 49 (13) years and 87% women.
The study protocols were approved by the IRB Comitè Ètic d’Investigació Clínica IDIAP Jordi Gol (codes 20/186-PCV, 20/094-PCV and 20/162-PCV), and written informed consent was obtained from participants.

SARS-CoV-2 antibody measurements

Naturally acquired IgM, IgA, and IgG responses to SARS-CoV-2 were quantified by Luminex. The antigen panel included five proteins: the spike full length protein (S) (aa 1-1213 expressed in Expi293 and His tag-purified) produced at the Center for Genomic Regulation (CRG, Barcelona), and its subregion S2 (purchased from SinoBiological), the receptor-binding domain (RBD) kindly donated by the Krammer lab (Mount Sinai, New York), the nucleocapsid (N) full length (FL) protein, and the specific C-terminal (CT) region (both expressed in-house in ISGlobal in E. coli and His tag-purified). In addition, the RBD proteins of four VoC (Alpha, Beta, Gamma and Delta, produced at CRG) were tested in the first and last three visits. Coupling of SARS-CoV-2 proteins to MagPlex® polystyrene 6.5 μm COOH-microspheres (Luminex Corp, Austin, TX, USA) was done as described [1, 30, 31]. Antigen-coupled microspheres were added to a 384-well Clear® flat bottom plate (Greiner Bio-One, Frickenhausen, Germany) in multiplex (2000 microspheres per analyte per well) in a volume of 90 μL of Luminex Buffer (1% BSA, 0.05% Tween 20, 0.05% sodium azide in PBS) using 384 channels Integra Viaflo semi-automatic device (96/384, 384 channel pipette). Two hyperimmune pools (one for IgG, and another one for IgA and IgM) were used as positive controls in each assay plate for QA/QC purposes and were prepared at 2-fold, 8 serial dilutions from 1:12.5. Pre-pandemic samples were used as negative controls to estimate the cutoff of seropositivity. Ten microliters of each dilution of the positive control, negative controls, and test samples (prediluted 1:50 in 96 round-bottom well plates) was added to a 384-well plate using Assist Plus Integra device with 12 channels Voyager pipette. Plasma samples had been previously assessed for optimal sample dilution to avoid saturated responses, tested here at 1:500. To quantify IgM and IgA responses, test samples and controls were pre-treated with anti-human IgG (Gullsorb) at 1:10 dilution, to avoid IgG interferences. Technical blanks consisting of Luminex Buffer and microspheres without samples were added in 4 wells to detect and adjust for non-specific microsphere signals. Plates were incubated for 1 h at room temperature in agitation (Titramax 1000) at 900 rpm and protected from light. Then, the plates were washed three times with 200 μL/well of PBS-T (0.05% Tween 20 in PBS), using BioTek 405 TS (384-well format). Twenty-five microliters of goat anti-human IgG phycoerythrin (PE) (GTIG-001, Moss Bio) diluted 1:400, goat anti-human IgA-PE (GTIA-001, Moss Bio) 1:200, or goat anti-human IgM-PE (GTIM-001, Moss Bio) 1:200 in Luminex Buffer was added to each well and incubated for 30 min. Plates were washed and microspheres resuspended with 80 μL of Luminex Buffer, covered with an adhesive film, and sonicated 20 s on sonicator bath platform, before acquisition on the Flexmap 3D® reader. At least 50 microspheres per analyte per well were acquired, and median fluorescence intensity (MFI) was reported for each analyte. Assay positivity cut-offs specific for each isotype and antigen were calculated as 10 to the mean plus 3 standard deviations of log10-transformed MFI values of 128 pre-pandemic controls (Additional file 1: Fig. S1). Positive serology was defined by being positive for IgG, IgA and/or IgM to any of the SARS-CoV-2 wild type of the antigens tested (NFL, NCT, S, RBD, S2).

Data analysis

We modeled antibody level trajectories over time with linear mixed models (LMM) using linear and quadratic fix effect terms for the time since infection and a random effect intercept to account for the dependency of longitudinal observations coming from the same individual. We repeatedly fitted LMMs changing our outcome of interest, which were the log10(MFI) for the different antigen and antibody isotype pairs. Considering that we modeled the log10(MFI) and that MFI signal is supposed to be relatively linear with antibody levels, negative (or positive) linear trends imply a constant negative (or positive) exponential antibody levels decay (or growth), whereas deviations from a linear trend for the log10(MFI) imply an acceleration or deceleration of the exponential antibody change. Estimated fixed effect regression coefficients and their standard deviations were used for prediction of temporal curves of antibody population averages and their 95% confidence intervals (CI). The associations between baseline determinants, clinical presentations, comorbidities and levels of antibodies were assessed at the time point closest to infection (between 5 and 9 months) and, at a later time point just prior to vaccination, about a year after infection (T4). Both univariable linear regression and stepwise regression models were fit to determine the effects of baseline variables on antibody levels (log10MFI). Multivariable models were selected based on the Akaike and Bayesian information criteria and adjusted r-square parameter. Finally, the formulas of the models were selected specifically at the antibody isotype level. For an easier interpretation of the results, a transformed beta value (%) of the log-linear model was calculated with the formula: ([10^beta]-1)*100, giving the difference (in percentage) in antibody levels when comparing to the reference group for categorical variables or for a one-unit increase for continuous variables. Likewise, a transformed beta value (%) of the log-log model was calculated with the formula: ([10^(beta*log10(1.1))]-1)*100, giving the difference (in percentage) in antibody levels for a 10% increase of the predictor variable, for continuous variables. Finally, we also assessed the association of the same baseline variables with differences in the rate of antibody changes as were estimated in our LMM fits of each antibody isotype kinetics. This association was estimated as a fix effect interaction with the time since symptom onset and was repeatedly estimated for all variables while controlling for a false discovery rate of 5%. Reinfected individuals were not excluded from the analysis of antibody kinetics or from the models to assess the associations of variables with antibody levels or decay. p-values were considered statistically significant at the 5% level. All data collected were managed and analyzed using the R software version 4.1.2.

Results and discussion

Of the total 247 HCW with past COVID-19 disease included in the cohort, Table 1 shows the number of non-vaccinated participants tested serologically per visit (T0–T8), involving 809 plasma samples and 15,267 antibody-antigen pair measurements overall. Among them, SARS-CoV-2 seropositivity combining all Ig isotypes and antigens was > 95% up to November 2021 (N = 23). The highest seropositivity was for IgG (~96%), especially for anti-S and anti-RBD responses, and IgA (~96%), mainly for anti-S responses. Seropositivity for IgM was ~48%, mainly for anti-RBD responses. Compared to July 2021, IgG levels remained stable and IgA and IgM seropositivity was increased in November 2021, probably due to an increase in asymptomatic infections, coinciding with the start of the sixth wave in Catalonia. This increase can be observed in the trajectory plots between T7 and T8 in Fig. 1, and it is especially evident for IgM to RBD and IgA to RBD and NFL. The kinetics of antibody levels up to 616 days since symptoms onset are shown in Fig. 1. The decay was more pronounced for anti-N than anti-S IgGs, with a remarkable sustain of S and S2 antibodies, less so for RBD. Overall, there was a slight but significant increase in IgA levels to S and S2 with time as observed by the predicted positive change in levels (Fig. 1), in contrast to the gradual decrease in antibody levels to the other antigens. Consistently, multivariable models at T4 had negative beta coefficients for all except IgA to S antigens that did not significantly wane with days since symptoms onset (Additional file 1: Table S1). Anti-S IgA unexpected rise might be related to sub-patent re-exposures resolved at the mucosal compartment. Thus, antibody kinetics after natural infection appeared to be more stably sustained than that after COVID-19 vaccination, which has been reported by vaccine manufacturers to decline more pronouncedly by 6–9 months [3234].
Table 1
SARS-CoV-2 seropositivity (overall, by isotype, and by isotype-antigen pair) in a cohort of pre-exposed non-vaccinated health care workers over 2020 and 2021
 
T0 (n = 127)
T1 (n = 122)
T2 (n = 118)
T3 (n = 52)
T4 (n = 151)
T5 (n = 105)
T6 (n = 72)
T7 (n = 39)
T8 (n = 23)
n
%
n
%
n
%
n
%
n
%
n
%
n
%
n
%
n
%
Overall seropositivity
118
92.91%
111
90.98%
109
92.37%
49
94.23%
145
96.03%
105
100%
71
98.61%
38
97.44%
22
95.65%
IgM seropositivity
81
63.78%
65
53.28%
54
45.76%
22
42.31%
76
50%
48
45.71%
11
15.28%
4
10.26%
11
47.83%
 IgM N CT
5
3.94%
3
2.46%
3
2.54%
3
5.77%
6
3.97%
2
1.90%
1
1.39%
0
0%
1
4.35%
 IgM N FL
5
3.94%
2
1.64%
1
0.85%
0
0%
2
1.32%
1
0.95%
1
1.39%
0
0%
1
4.35%
 IgM RBD
70
55.12%
54
44.26%
44
37.29%
16
30.77%
57
37.75%
41
39.05%
11
15.28%
3
7.69%
11
47.83%
 IgM S
35
27.56%
38
31.15%
32
27.12%
16
30.77%
45
29.80%
28
26.67%
3
4.17%
1
2.56%
2
8.70%
 IgM S2
22
17.32%
21
17.21%
20
16.95%
10
19.23%
34
22.52%
17
16.19%
1
1.39%
1
2.56%
0
0%
IgA seropositivity
112
88.19%
100
81.97%
90
76.27%
43
82.69%
134
88.74%
94
89.52%
57
79.17%
30
76.92%
22
95.65%
 IgA N CT
12
9.45%
29
23.77%
24
20.34%
19
36.54%
56
37.09%
12
11.43%
2
2.78%
1
2.56%
1
4.35%
 IgA N FL
33
25.98%
18
14.75%
23
19.49%
12
23.08%
37
25%
20
19.05%
4
5.56%
4
10.26%
8
34.78%
 IgA RBD
82
64.57%
86
70.49%
75
63.56%
40
76.92%
114
75.50%
84
80.00%
32
44.44%
14
35.90%
17
73.91%
 IgA S
105
82.68%
91
74.59%
80
67.80%
42
80.77%
115
76.16%
82
78.10%
52
72.22%
28
71.79%
22
95.65%
 IgA S2
97
76.38%
73
59.84%
72
61.02%
40
76.92%
112
74.17%
78
74.29%
40
55.56%
19
48.72%
21
91.30%
IgG seropositivity
118
92.91%
110
90.16%
108
91.53%
47
90.38%
143
94.70%
104
99.05%
70
97.22%
38
97.44%
22
95.65%
 IgG N CT
14
11.02%
100
81.97%
83
70.34%
29
55.77%
97
64.24%
25
23.81%
3
4.17%
2
5.13%
0
0%
 IgG N FL
110
86.61%
98
80.33%
83
70.34%
23
44.23%
76
50.33%
49
46.67%
8
11.11%
5
12.82%
9
39.13%
 IgG RBD
118
92.91%
110
90.16%
108
91.53%
47
90.38%
142
94.04%
104
99.05%
64
88.89%
35
89.74%
22
95.65%
 IgG S
118
92.91%
109
89.34%
106
89.83%
47
90.38%
142
94.04%
102
97.14%
70
97.22%
38
97.44%
22
95.65%
 IgG S2
118
92.91%
108
88.52%
104
88.14%
44
84.62%
141
93.38%
102
97.14%
66
91.67%
38
97.44%
22
95.65%
T, time point; T0, July–August 2020; T1, September 2020; T2, October 2020; T3, November 2020; T4, January–February 2021; T5, March–April 2021; T6, May–June 2021; T7, July 2021; T8, November 2021. Numbers in parentheses refer to samples tested
n, number of individuals who donated samples per time point (first row) and were positive by serology for each antibody/antigen pair (subsequent rows). Those who just received a first vaccine dose in the prior 6 days were included. The total number of pre-exposed individuals in the study cohort who were not vaccinated in 2021 was 161 at T4, 109 at T5, 77 at T6, 44 at T7, and 30 at T8. N nucleocapsid, FL full length, CT C-terminus, spike, RBD receptor-binding domain
We repeated the longitudinal analysis excluding post-reinfection samples from the 8 participants for which we had RT-qPCR diagnosed reinfections and obtained nearly identical results. Some quadratic models gave a positive slope at long times since infection, which could be due to asymptomatic reinfections by Delta and/or Omicron variants, and/or poor goodness of fit for antibody levels owing to the sparsity of data at this interval of time and to the relative simplicity of the model we chose (quadratic) to avoid overfitting.
There was substantially lower binding of circulating antibodies to RBD Beta, followed by Gamma and Delta variants, compared to the wild type, and less difference for Alpha, with an increase in seroprevalence at the later time points (Additional file 1: Table S2). Alpha (B.1.1.7) was first detected in the study area in the summer of 2020 (when B.1.177 was the predominant lineage) [35] and prevailed from February (> 50%) till June 2021 (80–99% cases). Delta (B.1.617.2) was first detected in May 2021, raising to 10% in June, and predominating since July (> 50%) till November 2021 (80–100% cases). Omicron (B.1.529) was first detected early December and was already majoritarian (> 56%) in January 2022. Beta and Gamma frequencies were negligible. Thus, the raise in seropositivity against Delta by T8 could be a mixture of cross-recognition and undetected asymptomatic reinfections at the fifth Spanish pandemic wave (summer–fall 2020).
According to the USA Centers for Disease Control and Prevention [36], reinfection is defined as occurring ≥ 90 days after initial positive testing or ≥ 45 days with background information supporting contact with confirmed cases or the reappearance of COVID-19–like symptoms. In our high-risk population (frontline unvaccinated HCW), there were 8/247 reinfections (incidence of 3.23%), with a mean time between first and second infection of 279 days (range 58–586). In a meta-analysis of 19 studies [37], the incidence of reinfection in recovered COVID-19 patients ranged from 0 to 20%. The pooled reinfection rate was 0.65% (95% CI 0.39–0.98%), with high heterogeneity (I2 = 99%). One of the studies showing a higher incidence of reinfection (15%) was in HCW from a hospital in Barcelona [38]. In our cohort, the mean age of reinfected individuals was 43.9 ± 9.5 years, 7 were female, and 62.5% had a comorbidity. The comorbidities, clinical presentations, dates of infections, and serology are presented in Additional file 1: Table S3. Seven of the reinfections were symptomatic, 85.7% had similar clinical symptoms in both episodes, and 14.3% had a milder form of disease in the second episode. In no case was the second infection more severe than the first, in contrast to another study where 27.8% of reinfected patients had more severe symptoms in the second episode [39].
Before the second positive RT-qPCR diagnosis, five reinfection cases had negative serology, one was undetermined, and two had positive serology. Among the latter, one (asymptomatic) had a weak antibody positive response, and the other (reporting a close positive contact) had a strong serological response (RBD IgG 10 times above the cutoff, S IgG 8 times above the cutoff). In this second case, the reinfection was with Delta. According to the Public Health England report, Delta increased the chances of reinfection by up to 46% compared to Alpha [40]. Overall, serology data suggest that most of the reinfections were due to insufficient natural immunity [36, 41], and the last case was probably due to immune escape, i.e., naturally acquired immunity to the original variant was not effective against Delta [42, 43]. In this subset of individuals with reinfections, there were significant increases in the slope for IgG (RBD, p = 0.027; S, p = 0.008; S2, p = 0.014) and IgA (S, p = 0.023; S2, p = 0.014) levels, all with rho > 0.21.
Two hundred twenty-three patients (90%) had at least one comorbidity. The most frequent was depression/anxiety (19.3%), followed by having had previous allergies (15.7%) and dyslipidemia (14.8%). We assessed baseline factors and comorbidities associated to antibody levels measured in the first sample post-infection available from each participant (from 5 to 9 months post infection) by multivariable stepwise regression models adjusting by time since infection (Table 2). Baseline variables most consistently and significantly associated with higher antibody levels 5–9 months after infection were age, obesity (n = 24), hypertension (n = 18), and variables related to the initial COVID-19 episode: hospitalization (n = 25), fever (n = 163), anosmia and/or hypogeusia (n = 133), chest pain (n = 41), and duration of symptoms (Table 2). Specifically, age was positively associated with anti-N IgA and IgG responses, having 2–2.5% higher levels with each year older. Hypertensive individuals had 57% higher N FL IgA levels, and obesity was associated with 25% lower N FL IgM levels. HCW who had anosmia/hypogeusia or fever had significantly higher IgG levels to all antigens than those without these conditions. Chest pain was associated with 20% higher N CT IgM levels. Higher IgA was positively associated with symptoms duration (median 22 days, IQR 12–34; N FL, rho = 0.116, p = 0.083; RBD, rho = 0.238, p < 0.001; S, rho = 0.244, p < 0.001). Hospitalized patients had 79% times higher RBD IgA levels than those non-hospitalized. Baseline factors associated with lower IgG levels included smoking, with 44% less IgG to N CT, 36% less to N FL and 51% less to RBD than non-smokers (Table 2). Variables significantly associated with antibody levels ~1 year after infection and just before most HCW received the first vaccine dose (T4), are shown in Additional file 1: Table S1. Additional factors significantly associated with lower IgA and IgG levels later on were being physician or nurse compared to other occupations in the primary care health sector and headache symptoms during the initial COVID-19 episode. All other variables, symptoms, or sequelae were either not statistically significantly associated with antibody levels or weakly associated in univariable models. Apart from the reported associations with antibody levels at the time closest to and farthest from infection, we also assessed a potential association of the same variables with differences in the rate of antibody changes as estimated in Fig. 1. The most consistent significant variables were tachycardia and cutaneous symptoms, associated with slower antibody decay, and oxygen supply, with faster antibody decay (Additional file 1: Table S4).
Table 2
Factors affecting Ig levels (log10 median fluorescent intensity) 5–9 months after COVID-19 by multivariable stepwise regression models
  
N CT
N FL
RBD
S
S2
Beta (%) 95% CI
Beta (%) 95% CI
Beta (%) 95% CI
Beta (%) 95% CI
Beta (%) 95% CI
IgA
Age
0.9 (0.15; 1.66)
1.9 (0.77; 3.04)
0.48 (− 0.46; 1.43)
0.98 (− 0.23; 2.22)
1.02 (− 0.54; 2.61)
Shivers
7.38 (− 8.68; 26.27)
17.13 (− 7.94; 49.02)
18 (− 3.79; 44.74)
18.04 (− 9.27; 53.59)
9.08 (− 22.22; 52.98)
Symptoms duration
0.21 (− 0.09; 0.52)
0.46 (0.01; 0.92)
0.48 (0.1; 0.87)
0.57 (0.07; 1.06)
0.35 (− 0.29; 0.99)
Sputum
− 0.53 (− 27.1; 35.73)
21.77 (− 23.28; 93.26)
26.52 (− 14.49; 87.19)
4.24 (− 37.08; 72.7)
− 12.9 (− 54.47; 66.64)
Fever
− 5.07 (− 21; 14.07)
9.16 (− 16.92; 43.43)
13.39 (− 10.05; 42.93)
28 (− 5.02; 72.5)
26.53 (− 13.77; 85.65)
Anosmia/hypogeusia
7.96 (− 7.85; 26.48)
− 1.43 (− 22.1; 24.72)
3.22 (− 15.45; 26.03)
24.88 (− 3.44; 61.51)
49.2 (7.2; 107.64)
Hospitalization
14.35 (− 23.18; 70.2)
− 8.98 (− 49.6; 64.39)
79.25 (8.58; 195.91)
47.25 (− 22.82; 180.94)
85.13 (− 19.29; 324.64)
Hypertension
18.89 (− 11.13; 59.07)
56.55 (1.57; 141.29)
8.63 (− 24.73; 56.77)
− 12.94 (− 45.74; 39.69)
12.92 (− 38.5; 107.32)
Dizziness
− 4.2 (− 21.91; 17.53)
− 1.46 (− 27.28; 33.52)
18.82 (− 8.17; 53.73)
20.71 (− 13.39; 68.25)
35.75 (− 11.4; 107.98)
Oxygen
17.09 (− 27.3; 88.57)
35.15 (− 33.44; 174.42)
− 18.38 (− 55.23; 48.81)
− 13.7 (− 60.2; 87.14)
− 34.56 (− 75.8; 76.93)
Cough
9.41 (− 7.11; 28.87)
4.24 (− 18.27; 32.95)
6.83 (− 13.09; 31.31)
14.26 (− 12.42; 49.07)
3.92 (− 26.15; 46.25)
IgG
Age
2.71 (1.37; 4.08)
2.23 (1.2; 3.27)
2.3 (0.66; 3.97)
1.31 (− 0.2; 2.84)
0.69 (− 0.4; 1.79)
Shivers
27.7 (− 5.25; 72.12)
12.11 (− 10.82; 40.94)
27.37 (− 11.63; 83.59)
17.65 (− 16.21; 65.21)
6.24 (− 16.88; 35.79)
Dyspnea
8.58 (− 20.4; 48.09)
1.35 (− 20.11; 28.57)
− 0.57 (− 32.02; 45.42)
3.96 (− 26.96; 47.97)
− 4.24 (− 25.81; 23.59)
Fever
107.38 (49.19; 188.25)
89.46 (47.19; 143.88)
192.5 (95.41; 337.82)
152.84 (73.85; 267.7)
82.01 (38.84; 138.59)
Anosmia/hypogeusia
50.43 (13.84; 98.8)
58.13 (27.7; 95.8)
90.52 (35.41; 168.06)
104.14 (48.67; 180.3)
73.92 (38.3; 118.71)
Hospitalization
36.15 (− 31.97; 172.47)
7.73 (− 36.71; 83.38)
52.13 (− 34.96; 255.88)
38.26 (− 37.19; 204.38)
29.59 (− 26.74; 129.24)
Dizziness
4.2 (− 27.37; 49.48)
12.64 (− 14.58; 48.54)
17.46 (− 24.5; 82.75)
6.62 (− 29.27; 60.73)
3.68 (− 22.94; 39.49)
Myalgia/arthralgia
− 0.88 (− 26.47; 33.62)
− 5.9 (− 25.16; 18.31)
− 16.5 (− 42.08; 20.39)
− 9.84 (− 35.81; 26.64)
− 8.22 (− 28.2; 17.32)
Oxygen
13.09 (− 51.02; 161.11)
14.62 (− 39.65; 117.71)
38.62 (− 50.26; 286.33)
27.63 (− 50.73; 230.58)
6.18 (− 46.63; 111.25)
Cough
23.2 (− 7.49; 64.07)
21 (− 2.86; 50.73)
29.93 (− 8.52; 84.56)
23.34 (− 10.96; 70.86)
21.7 (− 3.84; 54.03)
Ex-smoker
0.69 (− 27.97; 40.75)
− 4.64 (− 26.23; 23.28)
4.65 (− 30.57; 57.72)
5.92 (− 27.63; 55.04)
2.3 (− 22.32; 34.72)
Smoking
− 43.78*(− 66.34; − 6.09)
− 36.12 (− 56.9; − 5.34)
− 51.11 (− 73.92; − 8.34)
− 44.12 (− 68.82; 0.16)
− 32.05 (− 55.43; 3.61)
IgM
Shivers
− 6.18 (− 18.05; 7.4)
0.41 (− 15.79; 19.72)
14.29 (− 9.05; 43.62)
15.08 (− 5.92; 40.77)
7.81 (− 12.62; 33.01)
Pain chest
19.63 (0.22; 42.79)
6.45 (− 15.45; 34.01)
2.84 (− 23.74; 38.68)
8.62 (− 16.56; 41.39)
20.01 (− 8.85; 57.99)
Sputum
3.67 (− 20.83; 35.77)
7.94 (− 24.01; 53.31)
45.59 (− 7.68; 129.61)
24.46 (− 16.72; 86.02)
0.26 (− 34.07; 52.45)
Anosmia/hypogeusia
− 1.56 (− 14.2; 12.94)
− 2.61 (− 18.55; 16.45)
3.32 (− 18.08; 30.31)
4.06 (− 15.21; 27.7)
22.74 (− 0.86; 51.96)
Hospitalization
4.63 (− 25.82; 47.59)
8.15 (− 30.87; 69.19)
38.26 (− 22.67; 147.19)
31.32 (− 21.34; 119.23)
33.55 (− 21.74; 127.92)
Oxygen
− 6.06 (− 37.76; 41.79)
24.27 (− 27.26; 112.31)
40.07 (− 30.12; 180.75)
22.2 (− 33.82; 125.64)
− 19.77 (− 57.68; 52.09)
Cough
7.06 (− 7.33; 23.46)
3.06 (− 13.72; 25.31)
16.5 (− 8.05; 49.27)
14.89 (− 6.65; 43.13)
20.07 (− 3.52; 50.67)
Obesity
− 15.41 (− 31.54; 4.53)
− 25.48 (− 43.42; − 1.86)
− 19.05 (− 43.38; 15.73)
− 19.68 (− 41.4; 10.09)
− 13.48 (− 37.72; 20.21)
Statistically significant variables indicated in bold font. SARS-CoV-2 N nucleocapsid, FL full length, CT C-terminus, S spike, RBD receptor-binding domain
Previous acute phase studies have shown that COVID-19 severity is associated with higher antibody responses. Here, hospitalization was associated with higher Ig levels many months after convalescence, suggesting that severity does not affect the stability of memory B cells and antibody-producing plasma cells [4447]. Common symptoms such as fever and very specific symptoms such as altered smell and taste were also associated with higher antibody levels. Interestingly, hypertension was also positively associated with higher antibodies levels, consistent with some studies [29, 48] but contrary to others [49, 50]. We found that obesity was negatively related to IgM levels, similarly to post-vaccination studies in Italian HCW [50]. Smoking has been previously associated with lower antibody responses [28, 5052], and we showed that this effect persists after several months, mainly affecting IgG. Finally, lower antibody levels in physicians and nurses in later time points could be due to work-related stress or burn out, which might affect immune memory fitness [5355].
Limitations of this study include the lack of cellular or neutralizing antibody data, the specific focus on symptomatic HCW, and the limited sample size at later visits due to high vaccination coverage. Because of the screening of only those HCW with symptoms or contact with infected cases, we may have missed several reinfections. Another limitation is that we did not sequence the virus genome of the first infection and only some of the second infections; therefore, we cannot confirm reinfection with another SARS-CoV-2 variant. However, reinfections described occurred > 45 days after the first infection, and all of them had a negative RT-qPCR after the first infection and an increase in antibody levels after the 2nd infection. Future investigations should elucidate what threshold of antibodies correlate with protection against infection and disease, the determinants of antibody longevity, and what features of naturally-acquired antibody kinetics may predict that of vaccine-elicited responses.
In conclusion, our study demonstrates a robust persistence of SARS-CoV-2 antibodies after ~1.7 years, with seropositivity greater than 90% in unvaccinated individuals up to 20.5 months after COVID-19 symptoms onset. The maintenance of anti-S IgG, whose levels highly correlate with neutralizing antibodies [31], appears to be clinically relevant in protecting individuals particularly against the wild type and Alpha variants, despite lack of vaccination, consistent with having symptomatic reinfections in low responders and those reinfected with the more transmissible Delta variant. Antibody kinetics after natural infection appear to be stably sustained, more so than after vaccination, which has led to the implementation of booster immunizations, particularly in face of more contagious VoCs like Omicron. However, previously infected individuals also benefit from vaccination, as hybrid immunity seems to confer the greatest protection against SARS-CoV-2 infections and their symptoms [56].

Acknowledgements

We thank the HCW volunteers for their participation, the clinical staff for recruitment and sample and data collection, the microbiology labs for virology data, and the biobanks for baseline (T0) sample handling. We are grateful to Jordi Vila and Tomàs Pumarola from the VisCat study for T0 samples. We thank Chenjerai Jairoce, Robert A. Mitchell, and Inocencia Cuamba for assistance with sample processing, and Emma Cascant for logistics coordination.

Declarations

The study protocols were approved by the IRB Comitè Ètic d’Investigació Clínica IDIAP Jordi Gol (codes 20/186-PCV, 20/094-PCV and 20/162-PCV), and written informed consent was obtained from participants.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Dobaño C, Ramírez-Morros A, Alonso S, Vidal-Alaball J, Ruiz-Olalla G, Vidal M, et al. Persistence and baseline determinants of seropositivity and reinfection rates in health care workers up to 12.5 months after COVID-19. Res Sq. 2021;19:155. Dobaño C, Ramírez-Morros A, Alonso S, Vidal-Alaball J, Ruiz-Olalla G, Vidal M, et al. Persistence and baseline determinants of seropositivity and reinfection rates in health care workers up to 12.5 months after COVID-19. Res Sq. 2021;19:155.
2.
Zurück zum Zitat Moncunill G, Aguilar R, Ribes M, Ortega N, Rubio R, Salmerón G, et al. Determinants of early antibody responses to COVID-19 mRNA vaccines in a cohort of exposed and naïve healthcare workers. EBioMedicine. 2022;75:103805.CrossRef Moncunill G, Aguilar R, Ribes M, Ortega N, Rubio R, Salmerón G, et al. Determinants of early antibody responses to COVID-19 mRNA vaccines in a cohort of exposed and naïve healthcare workers. EBioMedicine. 2022;75:103805.CrossRef
3.
Zurück zum Zitat Shi D, Weng T, Wu J, Dai C, Luo R, Chen K, et al. Dynamic characteristic analysis of antibodies in patients with COVID-19: a 13-month study. Front Immunol. 2021;12:708184. Shi D, Weng T, Wu J, Dai C, Luo R, Chen K, et al. Dynamic characteristic analysis of antibodies in patients with COVID-19: a 13-month study. Front Immunol. 2021;12:708184.
4.
Zurück zum Zitat Gallais F, Gantner P, Bruel T, Velay A, Planas D, Wendling MJ, et al. Evolution of antibody responses up to 13 months after SARS-CoV-2 infection and risk of reinfection. EBioMedicine. 2021;71:103561. Gallais F, Gantner P, Bruel T, Velay A, Planas D, Wendling MJ, et al. Evolution of antibody responses up to 13 months after SARS-CoV-2 infection and risk of reinfection. EBioMedicine. 2021;71:103561.
5.
Zurück zum Zitat Pradenas E, Trinité B, Urrea V, Marfil S, Tarrés-Freixas F, Ortiz R, Rovirosa C, Rodon J, Vergara-Alert J, Segalés J, Guallar V, Valencia A, Izquierdo-Useros N, Noguera-Julian M, Carrillo J, Paredes R, Mateu L, Chamorro A, Toledo R, Massanella M, Clotet B, Blanco J. Clinical course impacts early kinetics,magnitude, and amplitude of SARS-CoV-2 neutralizing antibodies beyond 1 year after infection. J Cell Rep Med. 2022;3(2):100523. https://doi.org/10.1016/j.xcrm.2022.100523. Pradenas E, Trinité B, Urrea V, Marfil S, Tarrés-Freixas F, Ortiz R, Rovirosa C, Rodon J, Vergara-Alert J, Segalés J, Guallar V, Valencia A, Izquierdo-Useros N, Noguera-Julian M, Carrillo J, Paredes R, Mateu L, Chamorro A, Toledo R, Massanella M, Clotet B, Blanco J. Clinical course impacts early kinetics,magnitude, and amplitude of SARS-CoV-2 neutralizing antibodies beyond 1 year after infection. J Cell Rep Med. 2022;3(2):100523. https://​doi.​org/​10.​1016/​j.​xcrm.​2022.​100523.
6.
Zurück zum Zitat Eyran T, Vaisman-Mentesh A, Taussig D, Dror Y, Aizik L, Kigel A, Rosenstein S, Bahar Y, Ini D, Tur-Kaspa R, Kournos T, Marcoviciu D, Dicker D, Wine Y. Longitudinal kinetics of RBD+ antibodies in COVID-19 recovered patients over 14 months. PLoS Pathog. 2022;18(6):e1010569. https://doi.org/10.1371/journal.ppat.1010569. Eyran T, Vaisman-Mentesh A, Taussig D, Dror Y, Aizik L, Kigel A, Rosenstein S, Bahar Y, Ini D, Tur-Kaspa R, Kournos T, Marcoviciu D, Dicker D, Wine Y. Longitudinal kinetics of RBD+ antibodies in COVID-19 recovered patients over 14 months. PLoS Pathog. 2022;18(6):e1010569. https://​doi.​org/​10.​1371/​journal.​ppat.​1010569.
7.
Zurück zum Zitat Violán C, Torán P, Quirant B, Lamonja-Vicente N, Carrasco-Ribelles LA, Chacón C, et al. Antibody kinetics to SARS-CoV-2 at 13.5 months, by disease severity. medRxiv. 2021;13:2021.09.10.21262527. Violán C, Torán P, Quirant B, Lamonja-Vicente N, Carrasco-Ribelles LA, Chacón C, et al. Antibody kinetics to SARS-CoV-2 at 13.5 months, by disease severity. medRxiv. 2021;13:2021.09.10.21262527.
8.
Zurück zum Zitat Marcotte H, Piralla A, Zuo F, Du L, Cassaniti I, Wan H, et al. Immunity to SARS-CoV-2 up to 15 months after infection. iScience. 2022. PMID: 35018336. Marcotte H, Piralla A, Zuo F, Du L, Cassaniti I, Wan H, et al. Immunity to SARS-CoV-2 up to 15 months after infection. iScience. 2022. PMID: 35018336.
9.
Zurück zum Zitat Alejo JL, Mitchell J, Chang A, Chiang TPY, Massie AB, Segev DL, et al. Prevalence and durability of SARS-CoV-2 antibodies among unvaccinated US adults by history of COVID-19. JAMA. 2022;327:1085-7. Alejo JL, Mitchell J, Chang A, Chiang TPY, Massie AB, Segev DL, et al. Prevalence and durability of SARS-CoV-2 antibodies among unvaccinated US adults by history of COVID-19. JAMA. 2022;327:1085-7.
10.
Zurück zum Zitat Yang Y, Yang M, Peng Y, et al. Longitudinal analysis of antibody dynamics in COVID-19 convalescents reveals neutralizing responses up to 16 months after infection. Nat Microbiol. 2022;7:423-33. Yang Y, Yang M, Peng Y, et al. Longitudinal analysis of antibody dynamics in COVID-19 convalescents reveals neutralizing responses up to 16 months after infection. Nat Microbiol. 2022;7:423-33.
11.
Zurück zum Zitat Sterlin D, Mathian A, Miyara M, Mohr A, Anna F, Claër L, et al. IgA dominates the early neutralizing antibody response to SARS-CoV-2. Sci Transl Med. 2021;13:eabd2223.CrossRef Sterlin D, Mathian A, Miyara M, Mohr A, Anna F, Claër L, et al. IgA dominates the early neutralizing antibody response to SARS-CoV-2. Sci Transl Med. 2021;13:eabd2223.CrossRef
12.
Zurück zum Zitat Zhang J, Zhang H, Sun L. Therapeutic antibodies for COVID-19: is a new age of IgM, IgA and bispecific antibodies coming? MAbs. 2022;14:2031483.CrossRef Zhang J, Zhang H, Sun L. Therapeutic antibodies for COVID-19: is a new age of IgM, IgA and bispecific antibodies coming? MAbs. 2022;14:2031483.CrossRef
13.
Zurück zum Zitat Klingler J, Weiss S, Itri V, Liu X, Oguntuyo KY, Stevens C, et al. Role of immunoglobulin M and A antibodies in the neutralization of severe acute respiratory syndrome coronavirus 2. J Infect Dis. 2021;223:957–70.CrossRef Klingler J, Weiss S, Itri V, Liu X, Oguntuyo KY, Stevens C, et al. Role of immunoglobulin M and A antibodies in the neutralization of severe acute respiratory syndrome coronavirus 2. J Infect Dis. 2021;223:957–70.CrossRef
14.
Zurück zum Zitat Andreano E, Rappuoli R. SARS-CoV-2 escaped natural immunity, raising questions about vaccines and therapies. Nat Med. 2021;27:759–61.CrossRef Andreano E, Rappuoli R. SARS-CoV-2 escaped natural immunity, raising questions about vaccines and therapies. Nat Med. 2021;27:759–61.CrossRef
15.
Zurück zum Zitat Davies NG, Abbott S, Barnard RC, Jarvis CI, Kucharski AJ, Munday JD, et al. Estimated transmissibility and impact of SARS-CoV-2 lineage B.1.1.7 in England. Science. 2021;372:eabg3055. Davies NG, Abbott S, Barnard RC, Jarvis CI, Kucharski AJ, Munday JD, et al. Estimated transmissibility and impact of SARS-CoV-2 lineage B.1.1.7 in England. Science. 2021;372:eabg3055.
16.
Zurück zum Zitat Hu J, Peng P, Wang K, Fang L, Yang LF, Shun JA, et al. Emerging SARS-CoV-2 variants reduce neutralization sensitivity to convalescent sera and monoclonal antibodies. Cell Mol Immunol. 2021;18:1061–3.CrossRef Hu J, Peng P, Wang K, Fang L, Yang LF, Shun JA, et al. Emerging SARS-CoV-2 variants reduce neutralization sensitivity to convalescent sera and monoclonal antibodies. Cell Mol Immunol. 2021;18:1061–3.CrossRef
17.
Zurück zum Zitat Sabino EC, Buss LF, Carvalho MPS, Prete CA, Crispim MAE, Fraiji NA, et al. Resurgence of COVID-19 in Manaus, Brazil, despite high seroprevalence. Lancet (London, England). 2021;397:452–5.CrossRef Sabino EC, Buss LF, Carvalho MPS, Prete CA, Crispim MAE, Fraiji NA, et al. Resurgence of COVID-19 in Manaus, Brazil, despite high seroprevalence. Lancet (London, England). 2021;397:452–5.CrossRef
18.
Zurück zum Zitat Tegally H, Wilkinson E, Giovanetti M, Iranzadeh A, Fonseca V, Giandhari J, et al. Detection of a SARS-CoV-2 variant of concern in South Africa. Nat. 2021;592:438–43.CrossRef Tegally H, Wilkinson E, Giovanetti M, Iranzadeh A, Fonseca V, Giandhari J, et al. Detection of a SARS-CoV-2 variant of concern in South Africa. Nat. 2021;592:438–43.CrossRef
19.
Zurück zum Zitat Zheng Z, Peng F, Xu B, Zhao J, Liu H, Peng J, et al. Risk factors of critical & mortal COVID-19 cases: a systematic literature review and meta-analysis. J Inf Secur. 2020;81:e16–25. Zheng Z, Peng F, Xu B, Zhao J, Liu H, Peng J, et al. Risk factors of critical & mortal COVID-19 cases: a systematic literature review and meta-analysis. J Inf Secur. 2020;81:e16–25.
20.
Zurück zum Zitat Fang X, Li S, Yu H, Wang P, Zhang Y, Chen Z, et al. Epidemiological, comorbidity factors with severity and prognosis of COVID-19: a systematic review and meta-analysis. Aging (Albany NY). 2020;12:12493.CrossRef Fang X, Li S, Yu H, Wang P, Zhang Y, Chen Z, et al. Epidemiological, comorbidity factors with severity and prognosis of COVID-19: a systematic review and meta-analysis. Aging (Albany NY). 2020;12:12493.CrossRef
21.
Zurück zum Zitat Yang J, Zheng Y, Gou X, Pu K, Chen Z, Guo Q, et al. Prevalence of comorbidities and its effects in patients infected with SARS-CoV-2: a systematic review and meta-analysis. Int J Infect Dis. 2020;94:91–5.CrossRef Yang J, Zheng Y, Gou X, Pu K, Chen Z, Guo Q, et al. Prevalence of comorbidities and its effects in patients infected with SARS-CoV-2: a systematic review and meta-analysis. Int J Infect Dis. 2020;94:91–5.CrossRef
22.
Zurück zum Zitat Ssentongo P, Ssentongo AE, Heilbrunn ES, Ba DM, Chinchilli VM. Association of cardiovascular disease and 10 other pre-existing comorbidities with COVID-19 mortality: A systematic review and meta-analysis. PLoS One. 2020;15:e0238215.CrossRef Ssentongo P, Ssentongo AE, Heilbrunn ES, Ba DM, Chinchilli VM. Association of cardiovascular disease and 10 other pre-existing comorbidities with COVID-19 mortality: A systematic review and meta-analysis. PLoS One. 2020;15:e0238215.CrossRef
23.
Zurück zum Zitat Földi M, Farkas N, Kiss S, Zádori N, Váncsa S, Szakó L, et al. Obesity is a risk factor for developing critical condition in COVID-19 patients: A systematic review and meta-analysis. Obes Rev. 2020;21:e13095. Földi M, Farkas N, Kiss S, Zádori N, Váncsa S, Szakó L, et al. Obesity is a risk factor for developing critical condition in COVID-19 patients: A systematic review and meta-analysis. Obes Rev. 2020;21:e13095.
24.
25.
Zurück zum Zitat Hussain A, Mahawar K, Xia Z, Yang W, EL-Hasani S. Obesity and mortality of COVID-19. Meta-analysis. Obes Res Clin Pract. 2020;14:295–300.CrossRef Hussain A, Mahawar K, Xia Z, Yang W, EL-Hasani S. Obesity and mortality of COVID-19. Meta-analysis. Obes Res Clin Pract. 2020;14:295–300.CrossRef
26.
Zurück zum Zitat Aghili SMM, Ebrahimpur M, Arjmand B, Shadman Z, Pejman Sani M, Qorbani M, et al. Obesity in COVID-19 era, implications for mechanisms, comorbidities, and prognosis: a review and meta-analysis. Int J Obes. 2021;45:998–1016.CrossRef Aghili SMM, Ebrahimpur M, Arjmand B, Shadman Z, Pejman Sani M, Qorbani M, et al. Obesity in COVID-19 era, implications for mechanisms, comorbidities, and prognosis: a review and meta-analysis. Int J Obes. 2021;45:998–1016.CrossRef
27.
Zurück zum Zitat Biswas M, Rahaman S, Biswas TK, Haque Z, Ibrahim B. Association of sex, age, and comorbidities with mortality in COVID-19 patients: a systematic review and meta-analysis. Intervirology. 2021;64:36–47.CrossRef Biswas M, Rahaman S, Biswas TK, Haque Z, Ibrahim B. Association of sex, age, and comorbidities with mortality in COVID-19 patients: a systematic review and meta-analysis. Intervirology. 2021;64:36–47.CrossRef
28.
Zurück zum Zitat Gerhards C, Thiaucourt M, Kittel M, Becker C, Ast V, Hetjens M, et al. Longitudinal assessment of anti-SARS-CoV-2 antibody dynamics and clinical features following convalescence from a COVID-19 infection. Int J Infect Dis. 2021;107:221.CrossRef Gerhards C, Thiaucourt M, Kittel M, Becker C, Ast V, Hetjens M, et al. Longitudinal assessment of anti-SARS-CoV-2 antibody dynamics and clinical features following convalescence from a COVID-19 infection. Int J Infect Dis. 2021;107:221.CrossRef
29.
Zurück zum Zitat Horton DB, Barrett ES, Roy J, Gennaro ML, Andrews T, Greenberg P, et al. Determinants and dynamics of SARS-CoV-2 infection in a diverse population: 6-month evaluation of a prospective cohort study. J Infect Dis. 2021;224:1345.CrossRef Horton DB, Barrett ES, Roy J, Gennaro ML, Andrews T, Greenberg P, et al. Determinants and dynamics of SARS-CoV-2 infection in a diverse population: 6-month evaluation of a prospective cohort study. J Infect Dis. 2021;224:1345.CrossRef
31.
Zurück zum Zitat Ortega N, Ribes M, Vidal M, Rubio R, Aguilar R, Williams S, et al. Seven-month kinetics of SARS-CoV-2 antibodies and role of pre-existing antibodies to human coronaviruses. Nat Commun. 2021;12:4740.CrossRef Ortega N, Ribes M, Vidal M, Rubio R, Aguilar R, Williams S, et al. Seven-month kinetics of SARS-CoV-2 antibodies and role of pre-existing antibodies to human coronaviruses. Nat Commun. 2021;12:4740.CrossRef
33.
Zurück zum Zitat Naaber P, Tserel L, Kangro K, Sepp E, Jürjenson V, Adamson A, et al. Dynamics of antibody response to BNT162b2 vaccine after six months: a longitudinal prospective study. Lancet Reg Heal Eur. 2021;10:100208.CrossRef Naaber P, Tserel L, Kangro K, Sepp E, Jürjenson V, Adamson A, et al. Dynamics of antibody response to BNT162b2 vaccine after six months: a longitudinal prospective study. Lancet Reg Heal Eur. 2021;10:100208.CrossRef
34.
Zurück zum Zitat Pegu A, O’Connell SE, Schmidt SD, O’Dell S, Talana CA, Lai L, et al. Durability of mRNA-1273 vaccine-induced antibodies against SARS-CoV-2 variants. Science. 2021;373:1372–7.CrossRef Pegu A, O’Connell SE, Schmidt SD, O’Dell S, Talana CA, Lai L, et al. Durability of mRNA-1273 vaccine-induced antibodies against SARS-CoV-2 variants. Science. 2021;373:1372–7.CrossRef
36.
Zurück zum Zitat Investigative Criteria for Suspected Cases of SARS-CoV-2 Reinfection (ICR) | CDC. Centers for Disease Control and Prevention. 2020. Investigative Criteria for Suspected Cases of SARS-CoV-2 Reinfection (ICR) | CDC. Centers for Disease Control and Prevention. 2020.
37.
Zurück zum Zitat Mao Y-J, Wang W-W, Ma J, Wu S-S, Sun F. Reinfection rates among patients previously infected by SARS-CoV-2. Chin Med J. 2021;Publish Ah:1–8. Mao Y-J, Wang W-W, Ma J, Wu S-S, Sun F. Reinfection rates among patients previously infected by SARS-CoV-2. Chin Med J. 2021;Publish Ah:1–8.
38.
Zurück zum Zitat Sánchez-Montalvá A, Fernández-Naval C, Antón A, Durà X, Vimes A, Silgado A, et al. Risk of sars-cov-2 infection in previously infected and non-infected cohorts of health workers at high risk of exposure. J Clin Med. 2021;10. Sánchez-Montalvá A, Fernández-Naval C, Antón A, Durà X, Vimes A, Silgado A, et al. Risk of sars-cov-2 infection in previously infected and non-infected cohorts of health workers at high risk of exposure. J Clin Med. 2021;10.
39.
Zurück zum Zitat Brouqui P, Colson P, Melenotte C, Houhamdi L, Bedotto M, Devaux C, et al. COVID-19 re-infection. Eur J Clin Investig. 2021;51:e13537. Brouqui P, Colson P, Melenotte C, Houhamdi L, Bedotto M, Devaux C, et al. COVID-19 re-infection. Eur J Clin Investig. 2021;51:e13537.
40.
Zurück zum Zitat Health England P. SARS-CoV-2 variants of concern and variants under investigation; Techincal briefing 19; 2021. Health England P. SARS-CoV-2 variants of concern and variants under investigation; Techincal briefing 19; 2021.
41.
Zurück zum Zitat Wajnberg A, Amanat F, Firpo A, Altman DR, Bailey MJ, Mansour M, et al. Robust neutralizing antibodies to SARS-CoV-2 infection persist for months. Science. 2020;370:1227–30.CrossRef Wajnberg A, Amanat F, Firpo A, Altman DR, Bailey MJ, Mansour M, et al. Robust neutralizing antibodies to SARS-CoV-2 infection persist for months. Science. 2020;370:1227–30.CrossRef
42.
Zurück zum Zitat Kerlavage AR, Clayton RA, Sutton GG, Fleischmann RD, Ketchum KA, Klenk HP, et al. COVID-19 re-infection by a phylogenetically distinct SARS-coronavirus-2 strain confirmed by whole genome sequencing. Nature. 2018;388:539–47. Kerlavage AR, Clayton RA, Sutton GG, Fleischmann RD, Ketchum KA, Klenk HP, et al. COVID-19 re-infection by a phylogenetically distinct SARS-coronavirus-2 strain confirmed by whole genome sequencing. Nature. 2018;388:539–47.
43.
Zurück zum Zitat Van Elslande J, Vermeersch P, Vandervoort K, Wawina-Bokalanga T, Vanmechelen B, Wollants E, et al. Symptomatic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) reinfection by a phylogenetically distinct strain. Clin Infect Dis. 2021;73:354–6.CrossRef Van Elslande J, Vermeersch P, Vandervoort K, Wawina-Bokalanga T, Vanmechelen B, Wollants E, et al. Symptomatic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) reinfection by a phylogenetically distinct strain. Clin Infect Dis. 2021;73:354–6.CrossRef
44.
Zurück zum Zitat Abayasingam A, Balachandran H, Agapiou D, Hammoud M, Rodrigo C, Keoshkerian E, et al. Long-term persistence of RBD+ memory B cells encoding neutralizing antibodies in SARS-CoV-2 infection. Cell Reports Med. 2021;2:100228. Abayasingam A, Balachandran H, Agapiou D, Hammoud M, Rodrigo C, Keoshkerian E, et al. Long-term persistence of RBD+ memory B cells encoding neutralizing antibodies in SARS-CoV-2 infection. Cell Reports Med. 2021;2:100228.
45.
Zurück zum Zitat Sokal A, Chappert P, Barba-Spaeth G, Roeser A, Fourati S, Azzaoui I, et al. Maturation and persistence of the anti-SARS-CoV-2 memory B cell response. Cell. 2021;184:1201.CrossRef Sokal A, Chappert P, Barba-Spaeth G, Roeser A, Fourati S, Azzaoui I, et al. Maturation and persistence of the anti-SARS-CoV-2 memory B cell response. Cell. 2021;184:1201.CrossRef
46.
Zurück zum Zitat Sherina N, Piralla A, Du L, Wan H, Kumagai-Braesch M, Andréll J, et al. Persistence of SARS-CoV-2-specific B and T cell responses in convalescent COVID-19 patients 6–8 months after the infection. Med (New York, Ny). 2021;2:281. Sherina N, Piralla A, Du L, Wan H, Kumagai-Braesch M, Andréll J, et al. Persistence of SARS-CoV-2-specific B and T cell responses in convalescent COVID-19 patients 6–8 months after the infection. Med (New York, Ny). 2021;2:281.
48.
Zurück zum Zitat Ebinger JE, Botwin GJ, Albert CM, Alotaibi M, Arditi M, Berg AH, et al. Seroprevalence of antibodies to SARS-CoV-2 in healthcare workers: a cross-sectional study. BMJ Open. 2021;11:e043584.CrossRef Ebinger JE, Botwin GJ, Albert CM, Alotaibi M, Arditi M, Berg AH, et al. Seroprevalence of antibodies to SARS-CoV-2 in healthcare workers: a cross-sectional study. BMJ Open. 2021;11:e043584.CrossRef
49.
Zurück zum Zitat Karuna S, Li SS, Grant S, Walsh SR, Frank I, Casapia M, et al. Neutralizing antibody responses over time in demographically and clinically diverse individuals recovered from SARS-CoV-2 infection in the United States and Peru: a cohort study. PLoS Med. 2021;18:e1003868.CrossRef Karuna S, Li SS, Grant S, Walsh SR, Frank I, Casapia M, et al. Neutralizing antibody responses over time in demographically and clinically diverse individuals recovered from SARS-CoV-2 infection in the United States and Peru: a cohort study. PLoS Med. 2021;18:e1003868.CrossRef
50.
Zurück zum Zitat Watanabe M, Balena A, Tuccinardi D, Tozzi R, Risi R, Masi D, et al. Central obesity, smoking habit, and hypertension are associated with lower antibody titres in response to COVID-19 mRNA vaccine. Diabetes Metab Res Rev. 2022;38:e3465. Watanabe M, Balena A, Tuccinardi D, Tozzi R, Risi R, Masi D, et al. Central obesity, smoking habit, and hypertension are associated with lower antibody titres in response to COVID-19 mRNA vaccine. Diabetes Metab Res Rev. 2022;38:e3465.
51.
Zurück zum Zitat Schaffner A, Risch L, Aeschbacher S, Risch C, Weber MC, Thiel SL, et al. Characterization of a pan-immunoglobulin assay quantifying antibodies directed against the receptor binding domain of the SARS-CoV-2 S1-subunit of the spike protein: a population-based study. J Clin Med. 2020;9:3989. Schaffner A, Risch L, Aeschbacher S, Risch C, Weber MC, Thiel SL, et al. Characterization of a pan-immunoglobulin assay quantifying antibodies directed against the receptor binding domain of the SARS-CoV-2 S1-subunit of the spike protein: a population-based study. J Clin Med. 2020;9:3989.
52.
Zurück zum Zitat Jonsdottir HR, Bielecki M, Siegrist D, Buehrer TW, Züst R, Deuel JW. Titers of neutralizing antibodies against SARS-CoV-2 are independent of symptoms of non-severe COVID-19 in young adults. Viruses. 2021;13:284. Jonsdottir HR, Bielecki M, Siegrist D, Buehrer TW, Züst R, Deuel JW. Titers of neutralizing antibodies against SARS-CoV-2 are independent of symptoms of non-severe COVID-19 in young adults. Viruses. 2021;13:284.
53.
Zurück zum Zitat Glaser R, Kiecolt-Glaser JK, Malarkey WB, Sheridan JF. The influence of psychological stress on the immune response to vaccines. Ann N Y Acad Sci. 1998;840:649–55.CrossRef Glaser R, Kiecolt-Glaser JK, Malarkey WB, Sheridan JF. The influence of psychological stress on the immune response to vaccines. Ann N Y Acad Sci. 1998;840:649–55.CrossRef
54.
Zurück zum Zitat Kiecolt-Glaser JK, McGuire L, Robles TF, Glaser R. Psychoneuroimmunology and psychosomatic medicine: back to the future. Psychosom Med. 2002;64:15–28.CrossRef Kiecolt-Glaser JK, McGuire L, Robles TF, Glaser R. Psychoneuroimmunology and psychosomatic medicine: back to the future. Psychosom Med. 2002;64:15–28.CrossRef
55.
Zurück zum Zitat Jonsdottir IH, Sjörs DA. Mechanisms in endocrinology: Endocrine and immunological aspects of burnout: a narrative review. Eur J Endocrinol. 2019;180:R147–58.CrossRef Jonsdottir IH, Sjörs DA. Mechanisms in endocrinology: Endocrine and immunological aspects of burnout: a narrative review. Eur J Endocrinol. 2019;180:R147–58.CrossRef
56.
Zurück zum Zitat Pilz S, Theiler-Schwetz V, Trummer C, Krause R, Ioannidis JPA. SARS-CoV-2 reinfections: Overview of efficacy and duration of natural and hybrid immunity. Environ Res. 2022;209:112911.CrossRef Pilz S, Theiler-Schwetz V, Trummer C, Krause R, Ioannidis JPA. SARS-CoV-2 reinfections: Overview of efficacy and duration of natural and hybrid immunity. Environ Res. 2022;209:112911.CrossRef
Metadaten
Titel
Sustained seropositivity up to 20.5 months after COVID-19
verfasst von
Carlota Dobaño
Anna Ramírez-Morros
Selena Alonso
Rocío Rubio
Gemma Ruiz-Olalla
Josep Vidal-Alaball
Dídac Macià
Queralt Miró Catalina
Marta Vidal
Aina Fuster Casanovas
Esther Prados de la Torre
Diana Barrios
Alfons Jiménez
Jasmina Zanoncello
Natalia Rodrigo Melero
Carlo Carolis
Luis Izquierdo
Ruth Aguilar
Gemma Moncunill
Anna Ruiz-Comellas
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Schlagwort
COVID-19
Erschienen in
BMC Medicine / Ausgabe 1/2022
Elektronische ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-022-02570-3

Weitere Artikel der Ausgabe 1/2022

BMC Medicine 1/2022 Zur Ausgabe

Leitlinien kompakt für die Allgemeinmedizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Facharzt-Training Allgemeinmedizin

Die ideale Vorbereitung zur anstehenden Prüfung mit den ersten 24 von 100 klinischen Fallbeispielen verschiedener Themenfelder

Mehr erfahren

Isotretinoin: Risiko für schwere Laboranomalien „marginal erhöht“

08.05.2024 Akne Nachrichten

Die Aknetherapie mit Isotretinoin kann einen Anstieg von Leberenzymen und Blutfetten verursachen. Das Risiko für schwere Störungen ist laut einer Forschungsgruppe der Universität Lübeck aber nur marginal erhöht und auf einen engen Zeitraum konzentriert.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Klinik aktuell Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.

GLP-1-Rezeptoragonisten und SGLT-2-Hemmer: zusammen besser

06.05.2024 Typ-2-Diabetes Nachrichten

Immer häufiger wird ein Typ-2-Diabetes sowohl mit einem GLP-1-Rezeptor-Agonisten als auch mit einem SGLT-2-Inhibitor behandelt. Wie sich das verglichen mit den Einzeltherapien auf kardiovaskuläre und renale Komplikationen auswirkt, wurde anhand von Praxisdaten aus Großbritannien untersucht.

Update Allgemeinmedizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.