Skip to main content

Immune and inflammation features of severe and critical Omicron infected patients during Omicron wave in China

Abstract

Objective

The current study aimed to investigate the baseline immune and inflammatory features and in-hospital outcomes of patients infected with the Omicron variant (PIWO) who presented with different disease severities during the first wave of mass Omicron infections in the Chinese population has occurred.

Method

A cross‐sectional study was conducted on 140 hospitalized PIWO between December 11, 2022, and February 16, 2023. The clinical features, antibodies against SARS-CoV-2, immune cells, and inflammatory cytokines among mildly, severely, and critically ill PIWO at baseline and during follow-up period were compared.

Result

Patients with severe (n = 49) and critical (n = 35) disease were primarily male, needed invasive mechanical ventilation treatment, and exhibited higher mortality than those with mild disease (n = 56). During acute infection, SARS-CoV-2-specific antibody levels fluctuated with disease severity, serum antibodies increased and the incidence of severe cases decreased in critically ill PIWO over time. Antibody titers in severe or critical PIWO with no antibody responses at baseline did not increase significantly over time. Meanwhile, CD4+T cell, CD8+T cell, and natural killer cell counts were negatively correlated with disease severity, whereas interleukin (IL)-6 and IL-10 levels were positively correlated. In addition, combined diabetes, immunosuppressive therapy before infection, serum amyloid A, IL-10 and neutrophil counts were independently associated with severe and critical illness in PIWO. Among the 11 nonsurvivors, 8, 2, 1 died of respiratory failure, sudden cardiac death, and renal failure, respectively. Compared with survivors, nonsurvivors exhibited lower seropositivity of SARS-CoV-2-specific antibody, reduced CD3+T and CD4+T cell counts, and higher IL-2R, IL-6, IL-8, and IL-10 levels. Of note, lactate dehydrogenase was a significant risk factor of death in severe or critically ill PIWO.

Conclusion

This present study assessed the dynamic changes of SARS-CoV-2-specific antibodies, immune cells and inflammatory indexes between severely and critically ill PIWO. Critical and dead PIWO featured compromised humoral immune response and excessive inflammation, which broadened the understanding of the pathophysiology of Omicron infection and provides warning markers for severe disease and poor prognosis.

Highlights 

1. Specific anti-SARS-CoV-2 antibodies showed a trend of increasing first and then decreasing with the progression of disease severity.

2. The severity of infection decreased and the levels of the four antibodies against SARS-CoV-2 increased in critically ill PIWO over time.

3. Negative antibodies in severe/critical PIWO at baseline did not increase significantly with therapeutic intervention.

4. Dead PIWO exhibited disordered features of immunocompromised, hyperinflammatory, abnormal coagulation and myocardial injury.

5. Combined diabetes, immunosuppressive therapy before infection, high levels of SAA, IL-10 and neutrophil counts were the risk factors for severe/critical illness.

6. High LDH was related to death event in severely/critically ill PIWO.

Peer Review reports

Introduction

Due to large number of mutations in the spike origin, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant not only reduced the neutralizing function of antibodies induced by prior infection or vaccination but also enhanced its infectivity, worsening the epidemiological situation worldwide [1]. In addition to immune evasion, mutations in the Omicron sublineage reduced the effectiveness of sotrovimab, the only approved monoclonal antibody against the Omicron variant [2, 3]. Although the clinical efficacy of other available small molecule antivirals (remdesivir, molnupiravir, and nirmatrelvir) had remained effective against Omicron, their efficacy was limited to patients with early and mild disease stages [4].

Compared with the other predecessors of SARS-CoV-2, Omicron showed decreased lung infectivity and became less pathogenic [5]. Of note, patients infected with the Omicron variant (PIWO) exhibited more severe extrapulmonary organ disruptions, including renal and hematological failures, along with more frequent comorbidities [6]. In addition, the Omicron variant has been associated with less severe disease, lower in-hospital mortality, and less frequent need for intensive care than the Delta variant [7,8,9,10], which can be attributed to the evolved immune recognition processes (such as adaptive T cell responses induced by vaccination or infection to cross-recognize the Omicron variant) [11, 12], slow spread of Omicron from the main bronchus to the distal portion of the bronchioles [5], and reduced production of proinflammatory cytokines [13]. However, recent observational and prospective studies have reported that the mortality and invasive mechanical ventilation (IMV) requirement in critically ill PIWO were not significantly different from that in patients infected with the previous variant Delta [6, 14]. During the acute infection period, PIWO with severe disease were characterized by elevated interleukin (IL)-6, C-reactive protein (CRP), ferritin, D-Dimer, and lactate dehydrogenase (LDH), which is also associated with oxygen therapy, ICU admission, disease progression, and deceased[15,16,17,18,19,20,21]. Compared with mild patients, severely ill PIWO exhibited higher levels of white blood cells and neutrophils, lower levels of lymphocytes and its subsets. Lower CD4+T cell counts reflected the longer time of viral shedding [15, 16, 19, 22,23,24]. In addition, neutralizing antibody against Omicron variants were higher in vaccinated PIWO with pneumonia or hypoxemic respiratory failure than in those without [24].

Above studies combined severely and critically ill PIWO in one group to analyze their inflammatory and immunological trajectory, and the number of critical patients was too small to show the differences between severe and critically ill PIWO. Therefore, in the present study, clinical features and outcomes, the expression pattern and kinetics of antibodies against SARS-CoV-2, immune cells, and inflammatory cytokine expression among mildly, severely, and critically ill PIWO were compared; the characteristics of nonsurviving patients were explored; and the potential risk factors associated with coronavirus disease 2019 (COVID-19) severity and mortality were identified among PIWO from the North China area from December, 11, 2022, to February, 16, 2023, during the first Omicron infection wave.

Methods

Patient enrollment and data collection

PIWO were recruited from Baoding First Central Hospital (designated hospital for patients with COVID-19) during the first Omicron pandemic wave in China. Inclusion criteria for participants were as follows: (1) age ≥ 18 years; (2) documented positive result for SARS-CoV-2 using real-time reverse transcriptase-polymerase chain reaction from nasopharyngeal and/or oropharyngeal swabs; (3) no previous infection with other SARS-CoV-2 strains from self-report and medical records; (4) acute illness that necessitated hospitalization for their symptoms; and (5) ability to provide informed consent. Based on the data published by the Chinese Center for Disease Control and Prevention, all the included patients were diagnosed as infected with the Omicron variant (https://www.chinacdc.cn/jkzt/crb/zl/szkb_11803/jszl_13141/202301/t20230125_263519.html). Finally, 140 patients who met the inclusion criteria were enrolled in this study between December, 11, 2022, and February, 16, 2023.

The baseline information of the included PIWO at the time of admission, including age, sex, clinical symptoms and vital signs, SARS-CoV-2 vaccination status, comorbidity, and laboratory parameter tests, was collected. In addition, the data on treatment drugs, disease progression, and outcome after Omicron infection, of the participants were also recorded during their hospitalization. The patients were classified as per latest recommendations on the diagnosis and clinical management of COVID-19 infection (2023) [25] for disease severity. PIWO with mild disease were defined as those with the symptoms of fever, sore throat, and headache, but not of severe or critical illness. Patients who met any of the following were classified as having severe disease: (1) respiratory distress with respiratory rate ≥ 30 times/min; (2) oxygen saturation ≤ 93% at rest; (3) partial pressure of oxygen in arterial blood/fraction of inspiration oxygen ≤ 300 mm Hg; and (4) imaging suggesting the extent of lung involvement was > 50%. PIWO with critical condition were those who developed respiratory failure requiring IMV, or whose blood pressure drop required vasopressor drugs, or who were diagnosed with acute respiratory distress syndrome or septic shock. A total of 140 PIWO were divided into mild (n = 56), severe (n = 49), and critical (n = 35) illness groups.

The data on routine laboratory indices that reflect inflammation, coagulation, myocardial injury, blood oxygen, blood cell parameters, liver and kidney function, and electrolytes were derived from the laboratory information system. The details of laboratory index and the SARS-CoV-2 vaccination program are listed in the Supplementary materials.

For all enrolled PIWO, blood samples were collected at the time of their first examination after hospitalization (baseline). For severe and critically ill PIWO, blood samples were collected at baseline and were followed up once a week during hospitalization for the dynamic monitoring of antibody levels and inflammatory cytokines in PIWO. According to the sampling time after symptom onset, the time points of dynamic monitoring classified as follows: within 10 days (time point 1, T1), 10–17 days (T2), 17–24 days (T3), and > 24 days (T4) after symptom onset. The study design is shown in Fig. 1. Whole peripheral blood was obtained for lymphocyte subset analysis, where the serum was separated for cytokine and antibody assessments. Serum samples were isolated, numbered, and stored at − 80℃ until use.

Fig. 1
figure 1

Study design

The present study was approved by the Medical Ethics Committee of Baoding First Central Hospital ([2022]059), and informed consent was obtained from all enrolled patients.

SARS2-CoV-2-specific antibodies

Total antibodies against SARS-CoV-2 (including IgM, IgG, and IgA) targeting the receptor‐binding domain (RBD) region of the S1 subunit (Beijing Wantai Biological Pharmacy Enterprise, Beijing, China), neutralizing antibodies (NAbs) against SARS-CoV-2 wild type (WT) and Omicron BA.4/5 subvariant, which block the interaction between the RBD of the viral spike glycoprotein and human angiotensin-converting enzyme 2 (ACE2) (GenScript cPass™ SARS-CoV-2 Neutralization Antibody Detection Kit, USA), along with IgG antibodies against SARS-CoV-2 spike RBD (Hangzhou Proprium Biotech Co, Ltd., China) were measured as previously described [26]. ELISA plates were scanned at the wavelengths of 450 nm and 620 nm. For interpretation, the OD value of total anti-SARS‐CoV‐2 antibodies ≥ 0.19, the inhibition rate of SARS‐CoV‐2-NAbs ≥ 30%, and the concentration of IgG anti-RBD antibody ≥ 11.6 BAU/mL were considered positive.

Lymphocyte subsets

Lymphocyte subsets were measured on a flow cytometer (BeamCyte1026M) using a fluorescence monoclonal antibody kit (Tianjin QuantoBio Biotechnology Co.Ltd). The antibody clones that were used for flow cytometric analysis were as follows: FITC anti-CD3, PE anti-CD16 + 56, PerCP-Cy5.5 anti-CD45, PC7 anti-CD4, APC anti-CD19, and APC-Cy7 anti-CD8. First, a total of 20 µL antibody reagent and 50 µL whole blood were added to a tube, and incubated at room temperature in the dark for 20 min. Then 450 µL hemolysin was added to the tube, and after incubation at room temperature in the dark for another 15 min, the percentages of lymphocyte subsets were detected on the flow cytometer. The cell populations were as follows: B lymphocytes (CD45+CD19+), CD3+T lymphocytes (CD45+CD3+), CD4+T lymphocytes (CD45+CD3+CD4+), CD8+T lymphocytes (CD45+CD3+CD8+), and natural killer (NK) cells (CD3CD16+CD56+). The absolute values of total B cells, NK cells, T cells, and T cell subsets were calculated from the absolute values of lymphocytes enumerated using a hematology analyzer.

Cytokines

Six inflammatory cytokines including IL-1β, IL-2R, IL-6, IL-8, IL-10, and tumor necrosis factor (TNF)-α were detected using immunofluorescence luminescence (Tianjin QuantoBio Biotechnology Co.Ltd). First, 20 µL of capture microspheres coated with different specific antibodies against cytokines was mixed with 20 µL of serum and 20 µL of biotin-labeled detection antibodies. Then incubate the mixture at room temperature for 2 h in the dark with shaking (the shaking frequency was approximately 500 rpm) to form a complex of antibody capture microspheres-cytokine detection antibodies. Then, 20 μL of PE-labeled streptavidin was added to each tube, incubated at room temperature for 30 min with shaking in the dark, and analyzed on the flow cytometer. The flow cytometry data were processed using the FCAP software for obtaining cytokine concentration.

Statistical analysis

The primary outcome of the current study was severe/critical illness in PIWO and death in severe/critical PIWO. Ordinal logistic regression analyses were performed to investigate independent factors that influence COVID-19 severity in PIWO (from mild illness to severe to critical). Four characteristics of PIWO, including clinical parameters (time from symptom onset to hospitalization, sex, comorbidities, vaccination status, and previous immunosuppressive therapy), titers of the abovementioned four SARS-CoV-2-specific antibodies, count of immune subsets, and level of inflammatory factors (IL-1β, IL-2R, IL-6, IL-8, IL-10, TNF-α, CRP, serum amyloid A (SAA), and procalcitonin (PCT)), were included in the logistic regression models and analyzed using the step by step option. The models included the adjusted Model 1 (intaking clinical parameters, antibodies, or immune cells or inflammatory factors) and adjusted Model 2 (intaking all clinical parameters, antibodies, immune cells and inflammatory factors). Binary logistic regression was initially performed to define the clinical and laboratory risk factors associated with the mortality of PIWO. The statistically significant indicators identified between the survivors and nonsurvivors were included in univariate regression analyses. Next, statistically significant variables (p < 0.05) associated with deceased PIWO in the univariable analyses were analyzed using multivariable logistic regression analyses. Meanwhile, the variance inflation factor was < 3 and tolerance was > 0.3, indicating no multicollinearity.

The Shapiro–Wilk test was performed to assess the normality of distribution of the values. Independent sample t-test or one-way analysis of variance, respectively, was used to compare two groups or multiple groups for values with normal distribution. By contrast, the Wilcoxon rank-sum test or Kruskal–Wallis test was used for non-normally distributed data. The χ2 test was used to compare differences between categorical variables. Mean (standard deviation, SD) and median (interquartile range, IQR) are used to denote quantitative data that followed normal and non-normal distribution, respectively, whereas categorical variables are presented as numbers and percentages. The correlation analysis of the non-normally distributed data was performed using Spearman’s correlation coefficient. All statistical analyses were performed using R software (version 4.1.3), IBM SPSS Statistics (version 26.0), and or GraphPad Prism (version 8.0). P < 0.05 indicated statistical significance.

Results

Demographics and clinical characteristics of PIWO

The clinical and laboratory characteristics at baseline, along with the treatment and outcomes of PIWO during hospitalization are shown in Table 12 and Supplementary Table 1. Of all PIWO, 86% (120/140) had at least one underlying disease, with the most common comorbidities being hypertension (58%), heart disease (35%), diabetes (32%), and brain stroke (23%). No statistical difference was observed in the comorbidities, time from symptom onset to hospitalization, vaccination status, and second infection among the mildly, severely, and critically ill patients. A higher proportion of males, IMV treatment, and poor clinical outcome was observed in PIWO with severe and critical illness. The immunosuppressd medications before infection administered to severely ill (16%, 8/49) and critically ill patients (11%, 4/35) were similar (P = 0.753). In addition, the level of indicators that reflected inflammation, abnormal coagulation, and myocardial injury increased with disease severity.

Table 1 The baseline demographic and clinical characteristics of PIWO
Table 2 The treatment and prognosis of PIWO during hospitalization

Antibody signature of severely or critically ill PIWO

The level of total anti-SARS-CoV-2 antibodies, IgG anti-RBD antibodies, and NAbs against SARS-CoV-2 WT and Omicron BA.4/5 variant in PIWO at baseline showed a trend of an initial increase and subsequent decrease with disease severity (Fig. 2A-D). Severe patients had the highest total anti-SARS-CoV-2 antibodies titers, IgG anti-SARS-CoV-2 spike RBD antibody concentrations, the inhibition rates of NAb-WT and -BA.4/5 than mild or critical individuals. However, there was no significant difference in the titers of four specific antibodies against SARS-CoV-2 between the mild and critical patient group (Supplementary Table 1).

Fig. 2
figure 2

Comparison of SARS-CoV-2-specific antibody titers and seropositivity at hospitalization among PIWO with mild, severe, and critical disease in PIWO. A-D. Box plots show the level of total SARS-CoV-2-specific antibodies, anti-RBD IgG, and neutralizing antibodies against SARS-CoV-2 wild type and BA.4/5 in the mild, severe, and critical groups. EH. Seropositivity of SARS-CoV-2-specific antibodies among different COVID-19 disease severity groups. The numbers on the orange columns represent the number of individuals with positive SARS-CoV-2-specific antibodies in different groups

The seropositivity of the total antibodies against SARS‐CoV‐2 and anti-RBD IgG after the Omicron infection was comparable among the mild, severe and critical patient groups (Fig. 2E, F). While the positivity of NAb against WT and BA.4/5 were higher among severely ill patients than that among mildly or critically ill patients (Fig. 2G, H), exhibiting similar results to the inhibition rate.

The neutralizing effect of NAb-WT and NAb-BA.4/5 in blocking infection against SARS‐CoV‐2 WT and Omicron were similar in PIWO (Supplementary Fig. 1), indicating that the Omicron variant might harbor the same conserved sites as the WT strain, facilitating the development of therapeutically NAbs.

Additionally, we followed-up 12 critically ill PIWO and found that the incidence of severe cases decreased and serum levels of the four specific antibodies against SARS-CoV-2 increased over time (Fig. 3A-H. Supplementary Tables 2, 3). The enhancement of antibody titers and inhibition rate was more obvious in critical PIWO who had the good prognosis at the time of discharge (Fig. 3I-L). The kinetics of antibodies of the 13 severely ill PIWO were not consistent with those of critically ill. The inhibition rate of NAb-WT and -BA.4/5 remained high, while the anti-RBD IgG and total anti-SARS-CoV-2 antibodies fluctuated (Supplementary Fig. 2).

Fig. 3
figure 3

Dynamics of changing antibody levels in critically ill PIWO during hospitalization. A-D. The line chart shows the trend of the NAb-BA.4/5, NAb-WT, anti-RBD IgG, total anti-SARS-CoV-2 antibodies and the proportion of severe disease in critically ill patients over time. Red line represents the levels of anti-SARS-CoV-2 antibody, and blue line represents the percentage of severe cases. EH. Kinetics of four SARS-CoV-2-specific antibodies levels of PIWO with critical disease. I-L. The dynamic changes of SARS-CoV-2-specific antibodies in each critically ill patient. Red lines represent critically ill PIWO become asymptomatic or mild during follow-up. MN. Kinetics of SARS-CoV-2-specific antibodies levels in PIWO with negative antibody responses at admission in severe and critical group. Note: A-P: The x-axis represents sampling time after symptom, T1: within 10 days, T2: 10–17 days, T3: 10–24 days, T4: > 24 days. A-D: The right y-axis displays the antibody level, and the left y-axis shows the percentage of severe/critical cases. The dotted line represents the positive threshold. A-L: T1 (n = 11), T2 (n = 10), T3 (n = 12), T4: (n = 6)

Nine of the 25 PIWO followed-up had negative antibody responses at baseline. Among these patients, the proportions of males and critical cases were 67% (6/9) and 67% (6/9), respectively. Four patients had not received any COVID-19 vaccine, four underwent IMV therapy, three experienced secondary infections, and two died (Supplementary Table 4). Except for the inhibition rate of NAb-WT, the levels of the other three antibodies did not increase with therapeutic intervention during hospitalization. Of note, the inhibition rate of NAb-WT remained below the positive threshold (30%) in the later stages of hospitalization (Fig. 3M-P, Supplementary Table 5). The levels of the four antibodies in PIWO with severe or critical disease did not enhance significantly over time, which may be related to the subsequent poor prognosis.

In the current study, 74% (104/140) PIWO received SARS-CoV-2 vaccination and the median sampling time after the last vaccine was 376 days (Table 1). The doses of SARS-CoV-2 vaccine were positively correlated to the levels of four antibodies (Supplementary Fig. 3). Given that vaccination rates and sampling times after the last vaccine dose were comparable across PIWO in the mild, severe and critical groups (Table 1, Supplementary Fig. 4), we think that the difference in SARS-CoV-2-specific antibody levels among the three groups may be due to the severity of the Omicron infection in this situation.

Immune cell profiling of severely and critically ill PIWO

PIWO in the severe and critical groups were characterized by high counts of neutrophil and monocyte counts (Fig. 4A, B) and low level of lymphocyte counts (Fig. 4C) compared with those in the mild group, indicating obvious active inflammatory response. However, CD3+T, CD4+T, and CD8+T lymphocytes that participate in cellular immunity (Fig. 4D-F), B lymphocytes that impart humoral immunity (Fig. 4G), and NK cells that kill targeted cells (Fig. 4H) were reduced to varying degrees in severe, particularly critical, PIWO.

Fig. 4
figure 4

Immune cell differences among PIWO with different disease severities at the first sampling. A-C. Violin plots show the comparison of the counts (left) and proportions (right) of neutrophils (A), monocytes (B), and lymphocytes (C). D-H. Count (left) and percentage (right) differences in CD3+T cells (D), CD4+T cells (E), CD8+T cells (F), B cells (G), and NK cells (H) among different disease severity groups. Note: n = 56, 47, and 33 for PIWO with mild, severe, and critical condition, respectively. Four patients with severe or critical disease who had hematological malignancies were excluded

The Qin’s study observed the irreversible reduction of NK and CD8+T cells with over-expressed activation marker (CD38) and proliferation marker (Ki-67) in severe and critical PIWO. And overactivated NK cells and/or cytotoxic T-lymphocytes induced by the persistence of SARS-CoV-2 in patients with severe or critical illness could infiltrate lung tissue, leading to a worse clinical outcome [27]. In present study, CD4+T cells showed obviously increase in both critically and severely ill PIWO. PIWO with critical disease featured slight elevated CD8+T cells, significantly reduced B cells, and unchanged NK cell, which was inconsistent with the trend of cell changes in severe patients (Supplementary Figs. 5, 6).

Besides, preinfection immunosuppressive therapy including glucocorticoids and immunosuppressants such as prednisone, hydroxychloroquine sulfate had no effect on the count and percentage of neutrophils, monocytes, lymphocytes and lymphocyte subsets at baseline (Supplementary Fig. 7).

Inflammatory factors in PIWO of different severities

According to the heatmap of cytokines, critical PIWO presented with the characteristics of increased inflammation (Fig. 5A). Multivariable comparisons showed that IL-6, IL-10, SAA, and PCT levels were significantly different among the different groups (Fig. 5B-G and Supplementary Table 1). Besides, IL-1β, IL-6, SAA, PCT, and CRP were positively associated with disease severity (Fig. 5H, I), supplementary Fig. 8).

Fig. 5
figure 5

Cytokine characteristics of severely and critically ill PIWO at baseline A. Heatmap showing six inflammation-related cytokines in the mild (n = 56), severe (n = 49), and critical (n = 35) groups. Log10 (cytokine expression) was calculated for each row (each cytokine) and used for graphical visualization. B-G. Serum concentration of cytokines with different disease severities at baseline. H-I. Spearman rank correlation analysis was performed to evaluate the correlation of serum IL-6 (H) and IL-10 (I) with COVID-19 severity in PIWO

The longitudinal changes of different inflammation markers in 13 severe and 12 critical PIWO showed that above six cytokines waned with the extension of hospital stay among the majority of patients in severe group, while critically ill PIWO demonstrated the stable and high levels of inflammatory cytokines, so is CRP and PCT (Supplementary Figs. 9, 10).

Spearman’s correlation analysis demonstrated that the inhibition rate of NAb against WT and BA.4/5, the concentrations of IgG anti-RBD antibodies, and the levels of total anti-SARS-CoV-2 antibodies were all negatively correlated with the expression of IL-1β, IL-10 and TNF-α, while the level of total anti-SARS-CoV-2 antibodies was positively correlated with the counts of B lymphocytes. In addition, IL-2R and IL-10 were also inversely associated with the level of T lymphocyte subsets such as CD3+T, CD4+T, CD8+T cells. Furthermore, the severity of infection had positively correlation with male gender, and previous immunosuppressive therapy, and the levels of IL-6 and IL-10. Poor prognosis at the time of discharge is associated with high inflammatory cytokines, low B lymphocyte and T-cell subsets (Supplementary Fig. 11).

Risk factors associated with the COVID-19 severity and PIWO death

According to the adjusted Model 1, we identified total anti-SARS-CoV-2 antibodies and CD4+T cell counts were associated with mild condition, whereas male, concomitant diabetes, neutrophil counts and SAA were significantly risk factors linked with severity. When all variables were included into the adjusted Model 2, immunosuppressive therapy before infection and IL-10 had also been proved as the independent risk factors (Table 3).

Table 3 Risk factors for severe and critical illness in PIWO

Because no patient with mild disease were died of the disease progression, the characteristics of surviving and nonsurviving PIWO in severe and critical groups were compare. Compared with the survivors, nonsurvivors who were mainly in the critical condition (10/11, 90.91%) and had received IMV (8/22, 72.72%), exhibited immunosuppression, which manifested as the lower positive rates of total anti-SARS-CoV-2 antibodies, anti-RBD IgG, and NAbs against WT and BA.4/5 along with lower counts of CD3+T cells and CD4+T cells. In addition, the level of IL-6, IL-8, and IL-10 in nonsurviving PIWO were significantly higher than that in surviving PIWO. Furthermore, baseline D-Dimer, creatine kinase, myoglobin, LDH, and creatinine levels in PIWO who died were higher than those who survived, indicating abnormal coagulation, myocardial injury, or kidney damage. Among the 11 nonsurvivors, 8 died of respiratory failure, 2 of sudden cardiac death, and 1 of renal failure (Table 3 and Supplementary Fig. 12).

The statistically significant indicators in Table 4 were included in univariate regression analyses. Myoglobin, LDH, CD4+T cells, negative total anti-SARS-CoV-2 antibody, negative IgG anti-RBD antibody, negative NAb-BA.4/5, and negative NAb-WT were the independent factors associated with the death outcome in PIWO with severe or critical condition. Multivariate logistic regression analyses with the above risk and protective factors showed that LDH was a significant risk factors contributing to the death of PIWO (Table 5).

Table 4 The comparison between dead and surviving patients in severe and critical group
Table 5 Identified risk factors for death in severe/critical PIWO

Discussion

With the high incidence of Omicron infections but lower disease severity, Chinese authorities altered the epidemic prevention and control strategy, and changed the “dynamic zeroing policy” policy to the “reopening in an orderly and effective manner” policy on December, 7, 2022. However, owing to the high infectivity of Omicron, the strategy led to a huge health burden on the public, with the number of PIWO surging and reaching a peak of 6.94 million [28]. Moreover, severely ill and nonsurviving patients in the hospital reached a peak of 128,000 and daily peak of 4,273, respectively [28]. Therefore, the early identification and dynamic monitoring of the immune responses and inflammation characteristics of severe and critical PIWO can help to explore the potential pathological mechanism of disease progression. This present study accurately and extensively analyzes the anti-SARS-CoV-2 specific antibodies and lymphocyte subset alterations in addition to the expression pattern of inflammation factors among severely and critically ill PIWO as well as nonsurvivors during acute infection.

This retrospective study included 56 mild, 49 severe, and 35 critical PIWO during the loosen policy of China's epidemic control. Compared with previous studies (Supplementary Table 6), our research further demonstrates the levels of CD4+T cell, CD8+T cell, B cell, and NK cell were negatively correlated with COVID-19 severity, while pro-inflammation cytokines IL-6 and IL-10 positively. In addition, we found that (1) During acute infection, we showed an incremental trend of specific anti-SARS-CoV-2 antibodies first, which then decreased with disease severity; (2) The incidence of severe cases and the levels of the four specific antibodies against SARS-CoV-2 displayed the opposite trend in PIWO with critical illness over time; (3) Negative antibody responses in severe/critical PIWO at baseline did not increase with therapeutic intervention; (4) Nonsurvivors exhibited disordered features of immunocompromised, hyperinflammatory, abnormal coagulation and myocardial injury; (5) Combined diabetes, receiving immunosuppressive drugs, SAA, IL-10 and neutrophil counts were independently associated with severe and critical illness in PIWO, and high LDH was the significant risk factor for death in PIWO with severe or critical condition.

Several studies have reported that SARS-CoV-2-specific antibody titers were higher in patients with moderate/severe illness than in patients with asymptomatic/mild illness over the course of SARS-CoV-2 infection [29,30,31]. Moreover, males produced stronger SARS-CoV-2 -specific antibody responses than females [32] because of higher levels of ACE2 expression in men than in women [33]. Likewise, in the present study, anti-RBD IgG and NAbs against WT and BA.4/5 in PIWO with severe disease were higher than those with mild disease. The body's immune system generates immunoglobulin M (IgM) within 3–5 days of infection and produces IgG antibodies within 7 days to response the SARS-CoV-2 and exert protective functions[34]. NAbs prevent SARS-CoV-2 from binding to host cells, or prevent the conformational changes required to mediate fusion with host cell membranes, non-NAbs can activate immune effector cells through the interaction between Fc region and Fc receptor to clear viruses or infected cells [35]. However, antibody-dependent phagocytosis and antibody-dependent cellular cytotoxicity [36, 37], aberrant glycosylation patterns were observed in the anti-SARS-CoV-2 IgG antibodies among patients with severe disease but not in the those with mild illness, which induced high cytokine production, immune cell infiltration into the lungs, or platelet-mediated thrombosis, aggravating pathological damage in severe patients [38]. Thus, additional studies are required to interpret this dual role of antibodies in COVID-19, particular with regard to their pathological functions.

PIWO with non-mild (severe plus critical) disease had higher inhibition rates of NAb against WT and BA.4/5 and concentrations of anti-RBD IgG compared to those with mild disease (Supplementary Fig. 13), which is similar to patients infected with other SARS-CoV-2 strains [39, 40]. Of note, critically ill PIWO had the lower titers of total antibodies against SARS-CoV-2, IgG anti-RBD, and NAb against WT inhibition rates than severely. This characteristic was also observed in previous studies. Due to the decreased B cells and impaired immune response, patients admitted to ICU had lower anti-SARS-CoV-2 IgG than those not admitted to ICU [41], as did critically ill patients compared to severely ill patients [42]. Non-survivors also exhibited the deficient anti-RBD or/and anti-S IgG expression [43, 44]. The reasons for this impaired humoral immunity signature are as follows: (1) Patients with fatal outcomes showed the slower NAbs dynamics, even though they reached higher levels later in the disease trajectory, which is consistent with our current research. The delayed and incomplete antibody responses prolonged viral shedding. Elicited NAb responses within 14 day of symptom onset contributed to good recovery, whereas those induced at later time points lose protective effect [44] Anna et.al detected the anti-RBD IgG titers 34 days after onset of symptoms and found critical patients had higher antibodies, but the time of antibody production and the quality of antibodies are not enough to effectively control viral load [45]. (2) Critically ill COVID-19 patients exhibited functionally compromised antiviral T-cell response, whose SARS-CoV-2-specific CD4+ T cell inhibited IFN-γ, IL4, and IL-21 production [45]. IFN-γ could recruit other immune cells and dampens viral replication [46]; IL-4 induced suppressive functions of regulatory T cells through antagonizing Th1 and Th17 responses [47]; and IFN-γ and IL-4 inhibited the expression of ACE2 on cell surface and hinder SARS-CoV replication [48]. Furthermore, IL-21 can act directly on B cells and regulate the germinal center response [49]. Meanwhile, PIWO receiving immunosuppressive drug before infection was the risk factors for disease severity, suggesting the impaired B- and T-cell responses cannot withhold SARS-CoV-2 infection. (3) Moreover, in critically ill patients, vascular leakage enhanced circulating antigen-inexperienced naïve T cells infiltration to lungs and subsequent pulmonary edema. In view of the defects in germinal center induction (such as inefficient generation of T follicular helper cell (Tfh)) in the pathological tissues of deceased patients [50] and circulating Tfh dysfunction in critically ill patients [45]. Future studies should focus on the exact mechanism of late antibody response failure and the role of Tfh in COVID-19 immunity.

The expression of IL-6 and IL-10 gradually increases with disease severity in PIWO, this phenomenon was also seen in patients infected with other SARS-CoV-2 strains [51,52,53,54]. Previous studies have shown that IL-6 was the key molecule related to cytokine storm [55], and that it is extremely elevated in acute SARS-CoV-2 infection [56, 57]. Elevated serum IL-6 levels causing immune defects such as lymphopenia, impaired lymphocyte cytotoxicity, and endothelial activation could be partially restored by IL-6 blockade treatment with tocilizumab [55]. Additionally, IL-10 had dual role of anti-inflammatory and immune stimulation during the progression of COVID-19. It could inhibit Th1 cell activity and enhance ACE2 expression against increased pro-inflammatory mediators after infection [58,59,60]. However, elevated IL-10 leading to overstimulation of CD8+ T cells (potentially pro-inflammatory function) and IL-10 “resistance” (impaired anti-inflammatory function) amplify systemic inflammation [61]. Although IL-6 and IL-10 showed a consistent rise in levels independently of the genetic variant of the virus (ancestral Wuhan strain, Alpha, Delta and Omicron) compared with healthy donors, the level of these cytokines reduced with virus mutation [62, 63]. The current study yielded similar results. Severely ill, critically ill, and nonsurviving PIWO with higher IL-1β, IL-2R, IL-6, IL-8, or IL-10 levels did not develop cytokine storm syndrome. Upon infection, endothelial cells, epithelial cells, neutrophils, and monocytes cells release inflammatory mediators that activate immune cells, which collectively trigger a release of multiple proinflammatory cytokines and chemokines to eliminate SARS-CoV-2 virions [64]. However, a hyperactive cytokine response could increase the frequency of CD3+CD4+CD28CD57+ T cells with the expression of programmed death 1, thereby contributing to T cell exhaustion or senescence and subsequent acute immunodeficiency [65]. This suggests that the unregulated activation of the immune system can easily cause severe and critical illness, leading to poor outcomes. In addition the levels of the aforementioned cytokines vary depending on the variant of the virus, indicating that mutations in viral proteins may be associated with the pathogenesis of the infection and mechanisms of immune responses. Debmalya et al. have found that mutations in structural S and N proteins and the pathogenicity-associated accessory protein ORF3a in the Omicron variant produced less pro-inflammatory epitopes and more anti-inflammatory epitopes compared with the Delta and other variants, which hampered proinflammatory IL-6 stimulation and upgraded IFN-γ and IL-4 induction efficacy [66].

As a proinflammation cytokine, IL-2R is a risk factor of severe disease and death prognosis [67, 68]. The present study revealed that IL-2R level increased with disease severity and was higher in nonsurvivors than in survivors. In addition, IL-2R was associated with decreased T lymphocyte subset counts [69, 70]. IL-2 is essential for the proliferation, differentiation, and function of Tregs, CD4, and CD8 cells [71, 72]. Thus, the increase in circulating IL-2R level reflects a decrease in cell response to IL-2 [73], indicating that IL-2R may act as a T cell negative regulatory factor contributing to lymphopenia. On the one hand, high CD25 (IL-2R ɑ chain) level inhibited Ki67 expression among CD3/CD4/CD8 T cells through IL-2 signaling inhibition [74], and on the other hand, overactivated inflammatory response promoted T cell apoptosis [75]. However, the situation of patients with COVID-19 is more complicated, and future research should focus on whether IL-2R is a decisive factor for lymphopenia in COVID-19.

In the current study, the total mortality rate of PIWO was 8% (11/140). Of note, 0%, 2%, and 29% deaths were observed in mild, severe, and critical groups, respectively, which was lower than the 35% or 37.8% mortality rate observed in critically ill French or American PIWO [6, 76].The deceased PIWO exhibited immunosuppressed features on hospitalization, which was demonstrated by low SARS-CoV-2 specific antibody seroconversion rates, lymphopenia, active proinflammatory cytokine production, and heart and kidney damage, which is similar to the dead patients infected with other strains who were characterized by lymphopenia, high inflammation status (like neutrophils to lymphocytes ratio, platelet to lymphocytes ratio, and systematic immune-inflammation index), and increased serum BUN and creatinine at admission [77]. Meanwhile, certain risk factors in PIWO with severe and critical illness also influenced the clinical prognosis. Indeed, myoglobin and LDH, which reflect myocardial injury, were independently risk factors associated with PIWO death. It has been shown that SARS-CoV-2 infection causes severe cardiovascular complications such as acute myocardial infarction and heart failure [78]. Cardiac myocytes specifically express ACE2 to which the SARS-CoV-2 spike protein RBD binds with high affinity, which explains cardiac involvement in COVID-19 [79]. Two patients in this study died of cardiac involvement, both had a history of coronary atherosclerotic heart disease. This is consistent with the findings of Loba et al. [80]. Meanwhile, compared with surviving PIWO, the nonsurvivor group had a far lower positive rate of the four SARS-CoV-2 antibodies, negative specific antibodies against SARS-CoV-2 increased the risk of death by five to nine times. Several studies have reported that B-cell deficient patients had prolonged in-hospital stay and higher mortality [81,82,83,84], and that convalescent plasma therapy relieved their clinical symptoms [85]. Because the included PIWO had no previous infection with other variants, vaccination might have produced a neutralizing effect to SARS-CoV-2 WT. Indeed, SARS-CoV-2-specific T cell responses with cross-reactivity can recognize multiple epitopes across the SARS-CoV-2 proteome, thereby providing protection against other variants [12, 86]. In addition, different vaccine platforms exhibited > 80% preservation of memory T cell responses, and CD4 and CD8 cell responses after vaccination were shown to be active against other variants (including Omicron) [87]. Therefore, it is necessary to vaccinate different target groups to resolve the gap in immunity and reduce the risk of severe illness and death.

This study has some limitations. As the monitoring of viral variants was based on data from the Chinese Center for Disease Control and Prevention, we did not perform for all included patients with COVID-19. The relatively small number of longitudinally followed-up hospitalized patients have yielded statistically insignificant results. Third, due to the complexity of treatment protocols and clinical complications such as secondary infections, we were unable to quantify their effects on immune and inflammatory parameters. Finally, because of sample restrictions, differences in SARS‐CoV‐2-specific CD4 cells, and activated/functional T cells among PIWO groups with different severity could not be analyzed.

Conclusion

Taking together, the trajectory of the peripheral antibodies, immune cells and inflammatory features were different between severely and critically ill PIWO. Impaired SARS-CoV-2-specific antibody responses were observed in PIWO with critical illness and adverse outcomes. Combined diabetes, immunosuppressive therapy before infection, SAA, IL-10 and neutrophil counts were the risk factors of severe/critical illness, and high LDH was correlated with death in PIWO. The current research and results are valuable to save lives, especially PIWO with elderly age, comorbidities and inadequate antibody responses.

Availability of data and materials

The data will be available upon requested. Professor Yongzhe Li (yongzhelipumch@126.com) and Shuqin Guo (baoding8888@126.com) are responsible for data

provision.

Abbreviations

ACE2:

Angiotensin-converting enzyme 2

COVID-19:

Coronavirus disease 2019

CRP:

C-reactive protein

IL:

Interleukin

IQR:

Interquartile range

IMV:

Invasive mechanical ventilation

LDH:

Lactate dehydrogenase

NAb:

Neutralizing antibodies

NK cells:

Natural killer cells

PIWO:

Patients infected with Omicron

PCT:

Procalcitonin

RBD:

Receptor‐binding domain

SARS-CoV-2:

Severe acute respiratory syndrome coronavirus 2

SAA:

Serum amyloid A

SD:

Standard deviation

TNF-α:

Tumor necrosis factor α

WT:

Wild type

References

  1. Chen J, Wang R, Gilby NB, Wei GW. Omicron Variant (B.1.1.529): Infectivity, Vaccine Breakthrough, and Antibody Resistance. J Chem Inf Model [Journal Article; Research Support, NIH, Extramural; Research Support, US Gov’t, Non-PHS]. 2022;62(2):412–22.

    CAS  PubMed  Google Scholar 

  2. Takashita E, Kinoshita N, Yamayoshi S, Sakai-Tagawa Y, Fujisaki S, Ito M, et al. Efficacy of antibodies and antiviral drugs against Covid-19 Omicron Variant. N Engl J Med [Letter; Research Support, Non-US Gov’t]. 2022;386(10):995–8.

    PubMed  Google Scholar 

  3. Rockett R, Basile K, Maddocks S, Fong W, Agius JE, Johnson-Mackinnon J, et al. Resistance Mutations in SARS-CoV-2 Delta Variant after Sotrovimab Use. N Engl J Med [Letter]. 2022;386(15):1477–9.

    Article  PubMed  Google Scholar 

  4. Focosi D, McConnell S, Casadevall A, Cappello E, Valdiserra G, Tuccori M. Monoclonal antibody therapies against SARS-CoV-2. LANCET INFECT DIS [Journal Article; Review]. 2022;22(11):e311–26.

    Article  CAS  PubMed  Google Scholar 

  5. Suzuki R, Yamasoba D, Kimura I, Wang L, Kishimoto M, Ito J, et al. Attenuated fusogenicity and pathogenicity of SARS-CoV-2 Omicron variant. Nature. [Journal Article; Research Support, Non-US Gov’t]. 2022;603(7902):700–5.

    CAS  PubMed  Google Scholar 

  6. de Prost N, Audureau E, Heming N, Gault E, Pham T, Chaghouri A, et al. Clinical phenotypes and outcomes associated with SARS-CoV-2 variant Omicron in critically ill French patients with COVID-19. Nat Commun [Journal Article; Multicenter Study; Research Support, Non-US Gov’t]. 2022;13(1):6025.

    PubMed  Google Scholar 

  7. Bouzid D, Visseaux B, Kassasseya C, Daoud A, Femy F, Hermand C, et al. Comparison of patients infected with delta versus Omicron COVID-19 variants presenting to paris emergency departments : a retrospective cohort study. Ann Intern Med. 2022;175(6):831–7.

    Article  PubMed  Google Scholar 

  8. Wolter N, Jassat W, Walaza S, Welch R, Moultrie H, Groome M, et al. Early assessment of the clinical severity of the SARS-CoV-2 omicron variant in South Africa: a data linkage study. Lancet. [Journal Article; Research Support, Non-US Gov’t; Research Support, US Gov’t, PHS]. 2022;399(10323):437–46.

    CAS  PubMed  Google Scholar 

  9. Ledford H. How severe are Omicron infections? Nature [News]. 2021;600(7890):577–8.

    Article  CAS  PubMed  Google Scholar 

  10. Menni C, Valdes AM, Polidori L, Antonelli M, Penamakuri S, Nogal A, et al. Symptom prevalence, duration, and risk of hospital admission in individuals infected with SARS-CoV-2 during periods of omicron and delta variant dominance: a prospective observational study from the ZOE COVID Study. Lancet. [Journal Article; Observational Study; Research Support, Non-US Gov’t]. 2022;399(10335):1618–24.

    CAS  PubMed  Google Scholar 

  11. Keeton R, Tincho MB, Ngomti A, Baguma R, Benede N, Suzuki A, et al. T cell responses to SARS-CoV-2 spike cross-recognize Omicron. Nature. [Journal Article; Research Support, NIH, Extramural; Research Support, Non-US Gov’t]. 2022;603(7901):488–92.

    CAS  PubMed  Google Scholar 

  12. GeurtsvanKessel CH, Geers D, Schmitz KS, Mykytyn AZ, Lamers MM, Bogers S, et al. Divergent SARS-CoV-2 Omicron-reactive T and B cell responses in COVID-19 vaccine recipients. Sci Immunol [Journal Article; Research Support, NIH, Extramural; Research Support, Non-US Gov’t]. 2022;7(69):o2202.

    Google Scholar 

  13. Barh D, Tiwari S, Rodrigues GL, Ramalho PC, Andrade BS, Ahmad S, et al. SARS-CoV-2 Variants Show a Gradual Declining Pathogenicity and Pro-Inflammatory Cytokine Stimulation, an Increasing Antigenic and Anti-Inflammatory Cytokine Induction, and Rising Structural Protein Instability: A Minimal Number Genome-Based Approach. Inflamation [Journal Article]. 2023;46(1):297–312.

    CAS  Google Scholar 

  14. Vieillard-Baron A, Flicoteaux R, Salmona M, Chariot A, De Maupeou DB, Darmon M, et al. Omicron Variant in the Critical Care Units of the Paris Metropolitan Area: The Reality Research Group. Am J Respir Crit Care Med [Letter; Research Support, Non-US Gov’t]. 2022;206(3):349–63.

    Article  CAS  PubMed  Google Scholar 

  15. Muntean M, Briciu V, Lupse M, Colcear D, Macicasan RV, Csiszer A, et al. Effects of COVID-19 on the Liver and Mortality in Patients with SARS-CoV-2 Pneumonia Caused by Delta and Non-Delta Variants: An Analysis in a Single Centre. Pharmaceuticals (Basel). 2023;17(1):3. https://doi.org/10.3390/ph17010003.

  16. Chu J, Hua L, Liu X, Xiong H, Jiang F, Zhou W, et al. Superoxide dismutase alterations in COVID-19: implications for disease severity and mortality prediction in the context of omicron variant infection. Front Immunol [Journal Article; Research Support, Non-US Gov’t]. 2024;15:1362102.

    CAS  PubMed  Google Scholar 

  17. Peng H, Xiang T, Xu F, Jiang Y, Zhong L, Peng Y, et al. Redistribution and Activation of CD16brightCD56dim NK Cell Subset to Fight against Omicron Subvariant BA.2 after COVID-19 Vaccination. Microorganisms. 2023;11(4):940. https://doi.org/10.3390/microorganisms11040940.

  18. Zhou X, Huang X, Sun T, Jin X, Tian Z, Xue M, et al. Chronological changes of viral shedding in adult inpatients with Omicron infection in Shanghai. China Front Immunol. [Journal Article; Research Support, Non-US Gov’t]. 2023;14:1090498.

    CAS  PubMed  Google Scholar 

  19. Wang J, Choy KW, Lim HY, Ho P. Laboratory markers of severity across three COVID-19 outbreaks in Australia: has Omicron and vaccinations changed disease presentation? Intern Emerg Med. [Journal Article]. 2023;18(1):43–52.

    Article  PubMed  Google Scholar 

  20. Yadav R, Yadav V, Pokhriyal S, Zahid U, Gandhi A. Retrospective Observational Study of Complete Blood Count (CBC) Parameters and ICU Mortality of COVID-19 Disease in Delta Variant and Omicron Variant in a Community-Based Hospital in New York City. Cureus [Journal Article]. 2023;15(2):e34894.

    PubMed  Google Scholar 

  21. Hamar A, Mohammed D, Varadi A, Herczeg R, Balazsfalvi N, Fulesdi B, et al. COVID-19 mortality prediction in Hungarian ICU settings implementing random forest algorithm. Sci Rep [Journal Article]. 2024;14(1):11941.

    Article  CAS  PubMed  Google Scholar 

  22. Xia W, Jiang T, Tan Y, Li C, Wu S, Mei B. Characteristics of hematological parameters on admission in COVID-19 Omicron variant infected in Chinese population: a large-scale retrospective study. Bmc Infect Dis. [Journal Article]. 2023;23(1):835.

    Article  CAS  PubMed  Google Scholar 

  23. Wang J, Dong H, Zhao J, Li T, Wang M, Zhou C, et al. Effects of vaccines on clinical characteristics of convalescent adult patients infected with SARS-CoV-2 Omicron variant: A retrospective study. Front Microbiol. [Journal Article]. 2023;14:1096022.

    Article  PubMed  Google Scholar 

  24. Kawasuji H, Morinaga Y, Nagaoka K, Tani H, Yoshida Y, Yamada H, et al. High interleukin-6 levels induced by COVID-19 pneumonia correlate with increased circulating follicular helper T cell frequency and strong neutralization antibody response in the acute phase of Omicron breakthrough infection. Front Immunol [Journal Article; Observational Study; Research Support, Non-US Gov’t]. 2024;15:1377014.

    CAS  PubMed  Google Scholar 

  25. 北京协和医院新型冠状病毒感染诊疗多学科专家组. 北京协和医院成人新型冠状病毒感染实用诊疗建议(2023). 协和医学杂志. 2023 2023–01–11;14(01):50–9.

  26. Zhan H, Gao H, Liu Y, Zhang X, Li H, Li X, et al. Booster shot of inactivated SARS-CoV-2 vaccine induces potent immune responses in people living with HIV. J Med Virol [Journal Article; Research Support, Non-US Gov’t]. 2023;95(1):e28428.

    CAS  PubMed  Google Scholar 

  27. Qin L, Duan X, Dong JZ, Chang Y, Han Y, Li Y, et al. The unreversible reduced but persistent activated NK and CD8(+) T cells in severe/critical COVID-19 during omicron pandemic in China. Emerg Microbes Infect [Journal Article]. 2023;12(1):2208679.

    Article  PubMed  Google Scholar 

  28. https://www.chinacdc.cn/jkzt/crb/zl/szkb_11803/jszl_13141/202301/t20230125_263519.html.

  29. Legros V, Denolly S, Vogrig M, Boson B, Siret E, Rigaill J, et al. A longitudinal study of SARS-CoV-2-infected patients reveals a high correlation between neutralizing antibodies and COVID-19 severity. Cell Mol Immunol [Journal Article; Research Support, Non-US Gov’t]. 2021;18(2):318–27.

    CAS  PubMed  Google Scholar 

  30. Havervall S, Jernbom FA, Klingstrom J, Ng H, Greilert-Norin N, Gabrielsson L, et al. SARS-CoV-2 induces a durable and antigen specific humoral immunity after asymptomatic to mild COVID-19 infection. PLOS ONE. [Journal Article; Research Support, Non-US Gov’t]. 2022;17(1):e262169.

    Google Scholar 

  31. Ye X, Angelo LS, Nicholson EG, Iwuchukwu OP, Cabral DRW, Rajan A, et al. Serum IgG anti-SARS-CoV-2 Binding Antibody Level Is Strongly Associated With IgA and Functional Antibody Levels in Adults Infected With SARS-CoV-2. Front Immunol [Journal Article]. 2021;12:693462.

    Article  CAS  PubMed  Google Scholar 

  32. Shrock E, Fujimura E, Kula T, Timms RT, Lee IH, Leng Y, et al. Viral epitope profiling of COVID-19 patients reveals cross-reactivity and correlates of severity. Science. 2020;370(6520):eabd4250. https://doi.org/10.1126/science.abd4250.

  33. Patel SK, Velkoska E, Burrell LM. Emerging markers in cardiovascular disease: where does angiotensin-converting enzyme 2 fit in? Clin Exp Pharmacol Physiol. [Journal Article; Research Support, Non-US Gov’t; Review]. 2013;40(8):551–9.

    CAS  PubMed  Google Scholar 

  34. Guo YR, Cao QD, Hong ZS, Tan YY, Chen SD, Jin HJ, et al. The origin, transmission and clinical therapies on coronavirus disease 2019 (COVID-19) outbreak - an update on the status. Mil Med Res. [Journal Article; Research Support, Non-US Gov’t; Review]. 2020;7(1):11.

    CAS  PubMed  Google Scholar 

  35. Kim W, Zhou JQ, Horvath SC, Schmitz AJ, Sturtz AJ, Lei T, et al. Germinal centre-driven maturation of B cell response to mRNA vaccination. NATURE. [Journal Article]. 2022;604(7904):141–5.

    CAS  PubMed  Google Scholar 

  36. Tortorici MA, Beltramello M, Lempp FA, Pinto D, Dang HV, Rosen LE, et al. Ultrapotent human antibodies protect against SARS-CoV-2 challenge via multiple mechanisms. Sci. [Journal Article; Research Support, NIH, Extramural; Research Support, Non-US Gov’t]. 2020;370(6519):950–7.

    CAS  Google Scholar 

  37. Pinto D, Park YJ, Beltramello M, Walls AC, Tortorici MA, Bianchi S, et al. Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody. Nature. [Journal Article; Research Support, NIH, Extramural; Research Support, Non-US Gov’t; Research Support, US Gov’t, Non-PHS]. 2020;583(7815):290–5.

    CAS  PubMed  Google Scholar 

  38. Yaugel-Novoa M, Bourlet T, Paul S. Role of the humoral immune response during COVID-19: guilty or not guilty? Mucosal Immunol. [Journal Article; Research Support, Non-US Gov’t; Review]. 2022;15(6):1170–80.

    CAS  PubMed  Google Scholar 

  39. Peghin M, De Martino M, Palese A, Chiappinotto S, Fonda F, Gerussi V, et al. Antibody response and risk of reinfection over 2 years among the patients with first wave of COVID-19. Clin Microbiol Infect. [Journal Article]. 2024;30(4):522–30.

    Article  CAS  PubMed  Google Scholar 

  40. Kaygusuz S, Korukluoglu G, Cosgun Y, Sahin O, Arslan F. Investigation and long-term monitoring of the presence of neutralizing antibody in patients with COVID-19 disease of different clinical severity. J Med Virol. [Journal Article; Research Support, Non-US Gov’t]. 2022;94(8):3596–604.

    CAS  PubMed  Google Scholar 

  41. Dobi A, Sandenon SA, Lalarizo RM, Lebeau G, Vagner D, Frumence E, et al. Serological Surveillance of COVID-19 Hospitalized Patients in Reunion Island (France) Revealed that Specific Immunoglobulin G Are Rapidly Vanishing in Severe Cases. J Clin Med. 2020;9(12):3847. https://doi.org/10.3390/jcm9123847.

  42. Hou H, Wang T, Zhang B, Luo Y, Mao L, Wang F, et al. Detection of IgM and IgG antibodies in patients with coronavirus disease 2019. Clin Transl Immunology. [Journal Article]. 2020;9(5):e1136.

    Article  CAS  PubMed  Google Scholar 

  43. Ozonoff A, Schaenman J, Jayavelu ND, Milliren CE, Calfee CS, Cairns CB, et al. Phenotypes of disease severity in a cohort of hospitalized COVID-19 patients: Results from the IMPACC study. Ebiomedicine [Journal Article; Observational Study]. 2022;83:104208.

    CAS  PubMed  Google Scholar 

  44. Lucas C, Klein J, Sundaram ME, Liu F, Wong P, Silva J, et al. Delayed production of neutralizing antibodies correlates with fatal COVID-19. Nat Med. 2021;27(7):1178–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Oja AE, Saris A, Ghandour CA, Kragten N, Hogema BM, Nossent EJ, et al. Divergent SARS-CoV-2-specific T- and B-cell responses in severe but not mild COVID-19 patients. EUR J IMMUNOL. [Clinical Trial; Journal Article]. 2020;50(12):1998–2012.

    Article  CAS  PubMed  Google Scholar 

  46. Schroder K, Hertzog PJ, Ravasi T, Hume DA. Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol [Journal Article; Review]. 2004;75(2):163–89.

    Article  CAS  PubMed  Google Scholar 

  47. Guenova E, Skabytska Y, Hoetzenecker W, Weindl G, Sauer K, Tham M, et al. IL-4 abrogates T(H)17 cell-mediated inflammation by selective silencing of IL-23 in antigen-presenting cells. Proc Natl Acad Sci USA [Journal Article; Research Support, Non-US Gov’t]. 2015;112(7):2163–8.

    CAS  PubMed  Google Scholar 

  48. de Lang A, Osterhaus AD, Haagmans BL. Interferon-gamma and interleukin-4 downregulate expression of the SARS coronavirus receptor ACE2 in Vero E6 cells. Virology. [Journal Article; Research Support, Non-US Gov’t]. 2006;353(2):474–81.

    PubMed  Google Scholar 

  49. Linterman MA, Beaton L, Yu D, Ramiscal RR, Srivastava M, Hogan JJ, et al. IL-21 acts directly on B cells to regulate Bcl-6 expression and germinal center responses. J Exp Med. [Journal Article; Research Support, Non-US Gov’t]. 2010;207(2):353–63.

    CAS  PubMed  Google Scholar 

  50. Kaneko N, Kuo HH, Boucau J, Farmer JR, Allard-Chamard H, Mahajan VS, et al. Loss of Bcl-6-expressing T follicular helper cells and germinal centers in COVID-19. Cell. [Journal Article; Research Support, NIH, Extramural; Research Support, Non-US Gov’t]. 2020;183(1):143–57.

    CAS  PubMed  Google Scholar 

  51. Liu XQ, Xue S, Xu JB, Ge H, Mao Q, Xu XH, et al. Clinical characteristics and related risk factors of disease severity in 101 COVID-19 patients hospitalized in Wuhan China. Acta Phamarcol Sin. [Journal Article]. 2022;43(1):64–75.

    CAS  Google Scholar 

  52. Islam H, Chamberlain TC, Mui AL, Little JP. Elevated Interleukin-10 Levels in COVID-19: Potentiation of Pro-Inflammatory Responses or Impaired Anti-Inflammatory Action? Front Immunol. 2021;12:677008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Henry BM, de Oliveira M, Benoit S, Plebani M, Lippi G. Hematologic, biochemical and immune biomarker abnormalities associated with severe illness and mortality in coronavirus disease 2019 (COVID-19): a meta-analysis. Clin Chem Lab Med. [Journal Article; Meta-Analysis; Review]. 2020;58(7):1021–8.

    CAS  PubMed  Google Scholar 

  54. Albini A, Calabrone L, Carlini V, Benedetto N, Lombardo M, Bruno A, et al. Preliminary Evidence for IL-10-Induced ACE2 mRNA Expression in Lung-Derived and Endothelial Cells: Implications for SARS-Cov-2 ARDS Pathogenesis. Front Immunol [Journal Article; Research Support, Non-US Gov’t]. 2021;12:718136.

    CAS  PubMed  Google Scholar 

  55. Kim JS, Lee JY, Yang JW, Lee KH, Effenberger M, Szpirt W, et al. Immunopathogenesis and treatment of cytokine storm in COVID-19. Theranostics. [Journal Article; Review]. 2021;11(1):316–29.

    Article  CAS  PubMed  Google Scholar 

  56. Hoiland RL, Stukas S, Cooper J, Thiara S, Chen L, Biggs CM, et al. Amelioration of COVID-19-related cytokine storm syndrome: parallels to chimeric antigen receptor-T cell cytokine release syndrome. Br J Haematol. 2020;190(3):e150–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wang C, Kang K, Gao Y, Ye M, Lan X, Li X, et al. Cytokine Levels in the Body Fluids of a Patient With COVID-19 and Acute Respiratory Distress Syndrome: A Case Report. Ann Intern Med [Case Reports; Letter]. 2020;173(6):499–501.

    Article  PubMed  Google Scholar 

  58. Zhao W, Li Y, Xie R, Dong Y, Wei Y, Cheng C, et al. Real-World Evidence for COVID-19 Delta Variant’s Effects on the Digestive System and Protection of Inactivated Vaccines from a Medical Center in Yangzhou, China: A Retrospective Observational Study. Int J Clin Pract [Journal Article; Observational Study]. 2022;2022:7405448.

    PubMed  Google Scholar 

  59. Islam F, Habib S, Badruddza K, Rahman M, Islam MR, Sultana S, et al. The Association of Cytokines IL-2, IL-6, TNF-alpha, IFN-gamma, and IL-10 With the Disease Severity of COVID-19: A Study From Bangladesh. Cureus [Journal Article]. 2024;16(4):e57610.

    PubMed  Google Scholar 

  60. Carlini V, Noonan DM, Abdalalem E, Goletti D, Sansone C, Calabrone L, et al. The multifaceted nature of IL-10: regulation, role in immunological homeostasis and its relevance to cancer, COVID-19 and post-COVID conditions. Front Immunol. [Journal Article; Research Support, Non-US Gov’t; Review]. 2023;14:1161067.

    CAS  PubMed  Google Scholar 

  61. Qudus MS, Tian M, Sirajuddin S, Liu S, Afaq U, Wali M, et al. The roles of critical pro-inflammatory cytokines in the drive of cytokine storm during SARS-CoV-2 infection. J Med Virol. [Journal Article; Research Support, Non-US Gov’t; Review]. 2023;95(4):e28751.

    CAS  PubMed  Google Scholar 

  62. Korobova ZR, Arsentieva NA, Liubimova NE, Batsunov OK, Dedkov VG, Gladkikh AS, et al. Cytokine Profiling in Different SARS-CoV-2 Genetic Variants. Int J Mol Sci. 2022;23(22):14146. https://doi.org/10.3390/ijms232214146.

  63. Korobova ZR, Arsentieva NA, Liubimova NE, Dedkov VG, Gladkikh AS, Sharova AA, et al. A Comparative Study of the Plasma Chemokine Profile in COVID-19 Patients Infected with Different SARS-CoV-2 Variants. Int J Mol Sci. 2022;23(16):9058. https://doi.org/10.3390/ijms23169058.

  64. Merad M, Martin JC. Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages. Nat Rev Immunol [Journal Article; Review]. 2020;20(6):355–62.

    Article  CAS  PubMed  Google Scholar 

  65. Arcanjo A, Guimaraes PK, Logullo J, Leite P, Menezes C, Freire-de-Lima L, et al. Critically Ill coronavirus disease 2019 patients exhibit hyperactive cytokine responses associated with effector exhausted senescent T cells in acute infection. J Infect Dis [Journal Article; Research Support, Non-US Gov’t]. 2021;224(10):1672–83.

    CAS  PubMed  Google Scholar 

  66. Barh D, Tiwari S, Rodrigues GL, Ramalho PC, Andrade BS, Ahmad S, et al. SARS-CoV-2 Variants Show a Gradual Declining Pathogenicity and Pro-Inflammatory Cytokine Stimulation, an Increasing Antigenic and Anti-Inflammatory Cytokine Induction, and Rising Structural Protein Instability: A Minimal Number Genome-Based Approach. Inflammation [Journal Article]. 2023;46(1):297–312.

    Article  CAS  PubMed  Google Scholar 

  67. Qin R, He L, Yang Z, Jia N, Chen R, Xie J, et al. Identification of Parameters Representative of Immune Dysfunction in Patients with Severe and Fatal COVID-19 Infection: a Systematic Review and Meta-analysis. Clin Rev Allergy Immunol. [Journal Article; Meta-Analysis; Review; Systematic Review]. 2023;64(1):33–65.

    CAS  PubMed  Google Scholar 

  68. Li Y, Li X, Geng X, Zhao H. The IL-2A receptor pathway and its role in lymphocyte differentiation and function. Cytokine Growth Factor Rev. [Journal Article; Research Support, Non-US Gov’t; Review]. 2022;67:66–79.

    CAS  PubMed  Google Scholar 

  69. Hou H, Zhang B, Huang H, Luo Y, Wu S, Tang G, et al. Using IL-2R/lymphocytes for predicting the clinical progression of patients with COVID-19. Clin Exp Immunol. [Journal Article; Research Support, Non-US Gov’t]. 2020;201(1):76–84.

    CAS  PubMed  Google Scholar 

  70. Quartuccio L, Fabris M, Sonaglia A, Peghin M, Domenis R, Cifu A, et al. Interleukin 6, soluble interleukin 2 receptor alpha (CD25), monocyte colony-stimulating factor, and hepatocyte growth factor linked with systemic hyperinflammation, innate immunity hyperactivation, and organ damage in COVID-19 pneumonia. Cytokine. [Journal Article]. 2021;140:155438.

    Article  CAS  PubMed  Google Scholar 

  71. Li Y, Li X, Geng X, Zhao H. The IL-2A receptor pathway and its role in lymphocyte differentiation and function. Cytokine Growth Factor Rev. [Journal Article; Research Support, Non-US Gov’t; Review]. 2022;67:66–79.

    CAS  PubMed  Google Scholar 

  72. Zhang Y, Wang X, Li X, Xi D, Mao R, Wu X, et al. Potential contribution of increased soluble IL-2R to lymphopenia in COVID-19 patients. Cell Mol Immunol. [Journal Article]. 2020;17(8):878–80.

    Article  CAS  PubMed  Google Scholar 

  73. Gooding R, Riches P, Dadian G, Moore J, Gore M. Increased soluble interleukin-2 receptor concentration in plasma predicts a decreased cellular response to IL-2. Br J Cancer. [Comparative Study; Journal Article]. 1995;72(2):452–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Zhang Y, Wang X, Li X, Xi D, Mao R, Wu X, et al. Potential contribution of increased soluble IL-2R to lymphopenia in COVID-19 patients. Cell Mol Immunol. [Journal Article]. 2020;17(8):878–80.

    Article  CAS  PubMed  Google Scholar 

  75. Hou H, Zhang B, Huang H, Luo Y, Wu S, Tang G, et al. Using IL-2R/lymphocytes for predicting the clinical progression of patients with COVID-19. Clin Exp Immunol. [Journal Article; Research Support, Non-US Gov’t]. 2020;201(1):76–84.

    CAS  PubMed  Google Scholar 

  76. Tabatabai M, Juarez PD, Matthews-Juarez P, Wilus DM, Ramesh A, Alcendor DJ, et al. An Analysis of COVID-19 Mortality During the Dominancy of Alpha, Delta, and Omicron in the USA. J Prim Care Community Health. [Journal Article]. 2023;14:17176972.

    Google Scholar 

  77. Rokni M, Ahmadikia K, Asghari S, Mashaei S, Hassanali F. Comparison of clinical, para-clinical and laboratory findings in survived and deceased patients with COVID-19: diagnostic role of inflammatory indications in determining the severity of illness. BMC Infect Dis. [Comparative Study; Journal Article]. 2020;20(1):869.

    Article  CAS  PubMed  Google Scholar 

  78. Long B, Brady WJ, Koyfman A, Gottlieb M. Cardiovascular complications in COVID-19. AM J Emerg Med. [Journal Article; Review]. 2020;38(7):1504–7.

    Article  PubMed  Google Scholar 

  79. Chung MK, Zidar DA, Bristow MR, Cameron SJ, Chan T, Harding CR, et al. COVID-19 and Cardiovascular Disease: From Bench to Bedside. Circ Res. [Journal Article; Research Support, NIH, Extramural; Research Support, Non-US Gov’t; Research Support, US Gov’t, Non-PHS; Review]. 2021;128(8):1214–36.

    CAS  PubMed  Google Scholar 

  80. Alam L, Omar A, Talebi S, Narula J, Argulian E. Echocardiographic findings in patients with COVID-19 with myocardial injury during the omicron variant surge. AM J CARDIOL [Letter]. 2022;172:168–9.

    Article  CAS  PubMed  Google Scholar 

  81. Dulery R, Lamure S, Delord M, Di Blasi R, Chauchet A, Hueso T, et al. Prolonged in-hospital stay and higher mortality after Covid-19 among patients with non-Hodgkin lymphoma treated with B-cell depleting immunotherapy. AM J Hematol [Journal Article; Multicenter Study]. 2021;96(8):934–44.

    CAS  PubMed  Google Scholar 

  82. Vergidis P, Levy ER, Ristagno EH, Iyer VN, O’Horo JC, Joshi AY. COVID-19 in patients with B cell immune deficiency. J Immunol Methods [Journal Article]. 2022;510:113351.

    Article  CAS  PubMed  Google Scholar 

  83. Boekel L, Wolbink GJ. Rituximab during the COVID-19 pandemic: time to discuss treatment options with patients. Lancet Rheumatol [Journal Article]. 2022;4(3):e154–5.

    Article  CAS  PubMed  Google Scholar 

  84. Arellano-Llamas AA, Vela-Ojeda J, Hernandez-Caballero A. Chronic Lymphocytic Leukemia in the SARS-CoV-2 Pandemic. Curr Oncol Rep. [Journal Article; Review]. 2022;24(2):209–13.

    Article  CAS  PubMed  Google Scholar 

  85. Hueso T, Pouderoux C, Pere H, Beaumont AL, Raillon LA, Ader F, et al. Convalescent plasma therapy for B-cell-depleted patients with protracted COVID-19. Blood. [Journal Article; Multicenter Study; Observational Study; Research Support, Non-US Gov’t]. 2020;136(20):2290–5.

    CAS  PubMed  Google Scholar 

  86. McLean G, Kamil J, Lee B, Moore P, Schulz TF, Muik A, et al. The Impact of Evolving SARS-CoV-2 Mutations and Variants on COVID-19 Vaccines. Mbio. [Journal Article; Research Support, NIH, Extramural; Research Support, Non-US Gov’t; Review]. 2022;13(2):e297921.

    Google Scholar 

  87. Tarke A, Coelho CH, Zhang Z, Dan JM, Yu ED, Methot N, et al. SARS-CoV-2 vaccination induces immunological T cell memory able to cross-recognize variants from Alpha to Omicron. Cell. [Journal Article]. 2022;185(5):847–59.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not application.

Funding

This work was supported by the Beijing Municipal Natural Science Foundation (M23008, 7234383), National Key Research and Development Program of China (2018YFE0207300), National High Level Hospital Clinical Research Funding (2022-PUMCH-B-124), China Postdoctoral Science Foundation (2023T160060), and the Baoding Key Laboratory of research on the pathogenicity and predictive value of autoantibodies (2163P045).

Author information

Authors and Affiliations

Authors

Contributions

YML and HTZ designed the research, analyzed the data, drafted the first manuscript, and constructed the figures. YPG was responsible for managing the operation of experiments. XL was responsible for the selection of participants, the collection of patient data, and the quality control of experimental operations. JJC undertook the work of reviewing medical records. SF and XYL performed experiment operations. XML, HLL, and LLC organized data and searched literature. YZL, YPG and LLC were responsible for the support of experimental funds. YML, HTZ, YZL, and SYG provided direction and guidance throughout the preparation of this manuscript. YZL and SYG designed the research, supervised the work, and reviewed the manuscript. All authors contributed to the article and approved the submitted version.

Corresponding authors

Correspondence to Shuqin Guo or Yongzhe Li.

Ethics declarations

Ethics approval and consent to participate

This study was approved by Medical Ethics Committee of Baoding First Central Hospital ([2022]059). Informed consents were obtained from all enrolled patients.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, Y., Guo, Y., Zhan, H. et al. Immune and inflammation features of severe and critical Omicron infected patients during Omicron wave in China. BMC Infect Dis 24, 809 (2024). https://doi.org/10.1186/s12879-024-09652-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12879-024-09652-y

Keywords