• OPEN ACCESS

Safety and Immunogenicity After Primary and Booster Inactivated SARS-Cov-2 Vaccination in Patients with Autoimmune Liver Diseases

  • Zhiwei Chen1,#,
  • Yuting Wang2,#,
  • Taiyu He2,
  • Hu Li1,
  • Ling Ao2,
  • Qingbo Pan2,
  • Yingzhi Zhou2,
  • Qian Zhu2,
  • Dejuan Xiang2,
  • Gaoli Zhang2,
  • Ning Ling1,
  • Min Chen2,
  • Peng Hu1,2,
  • Mingli Peng2,
  • Dachuan Cai1,
  • Dazhi Zhang1,* and
  • Hong Ren1,2,* 
 Author information
Journal of Clinical and Translational Hepatology 2024;12(2):162-171

DOI: 10.14218/JCTH.2023.00049

Abstract

Background and Aims

SARS-CoV-2 vaccines-associated autoimmune liver diseases have been reported in several case reports. However, the safety and immunogenicity after primary and booster inactivated SARS-CoV-2 vaccination in patients with autoimmune liver diseases (AILD) is still unknown.

Methods

Eighty-four patients with AILD were prospectively followed up after the second dose (primary) of inactivated SARS-CoV-2 vaccine. Some of them received the third dose (booster) of inactivated vaccine. Adverse events (AEs), autoimmune activation, and liver inflammation exacerbation after primary and booster vaccination were recorded. Meanwhile, dynamics of antireceptor-binding-domain IgG (anti-RBD-IgG), neutralizing antibodies (NAbs) and RBD-specific B cells responses were evaluated.

Results

The overall AEs in AILD patients after primary and booster vaccination were 26.2% and 13.3%, respectively. The decrease of C3 level and increase of immunoglobulin light chain κ and λ levels were observed in AILD patients after primary vaccination, however, liver inflammation was not exacerbated, even after booster vaccination. Both the seroprevalence and titers of anti-RBD-IgG and NAbs were decreased over time in AILD patients after primary vaccination. Notably, the antibody titers were significantly elevated after booster vaccination (10-fold in anti-RBD-IgG and 7.4-fold in NAbs, respectively), which was as high as in healthy controls. Unfortunately, the inferior antibody response was not enhanced after booster vaccination in patients with immunosuppressants. Changes of atypical memory B cells were inversely related to antibody levels, which indicate that the impaired immune memory was partially restored partly by the booster vaccination.

Conclusions

The well tolerability and enhanced humoral immune response of inactivated vaccine supports an additional booster vaccination in AILD patients without immunosuppressants.

Keywords

SARS-CoV-2, Autoimmune liver disease, Inactivated SARS-CoV-2 vaccine, Safety, Antibody responses, Memory B cells

Introduction

More than 3 years after the first reported case of coronavirus disease 2019 (COVID-19), the pandemic is still overwhelming around the world.1 As of December 1, 2022, COVID-19 has led to over 630 million infections worldwide and claimed over 6.6 million lives (World Health Organization COVID-19 Dashboard). Vaccination is still an effective measure for the prevention of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, severe disease, and mortality.2

Recent case reports found that autoimmune hepatitis (AIH) developing after SARS-CoV-2 vaccine,3–8 and vaccine could also act as a trigger in the disease course of AIH.9 In addition, it has been reported that SARS-CoV-2 vaccination can be associated with liver injury, and some patients showed features of immune-mediated hepatitis,10 which lead to concerns of the safety of vaccines. Autoimmune liver diseases (AILD), as a special type of chronic liver disease, including AIH, primary biliary cholangitis (PBC), and AIH + PBC (overlap syndrome). The liver damage of AILD is mainly caused by immune disorders and can be induced or aggravated by drug exposure, viral infection, and even vaccination.5,9–12 Thus, patients with AILD may be more vulnerable to SARS-CoV-2 vaccines in theory, which has discouraged most AILD patients from accepting vaccination. However, relevant safety data are limited.

Considering that antibody responses decay over time, booster dose programs have been recommended by World Health Organization.13 Our previous study has revealed that the antibody responses were compromised in AILD patients after primary inactivated vaccination.14 In addition, another previous study also showed an inferior antibody response to mRNA or Johnson & Johnson vaccine in patients with AILD and with immunosuppressants.15 However, it is currently not known whether booster dose of inactivated vaccine is likely to improve antibody responses in AILD patients.

In this prospective study, we aimed to investigate the safety, humoral responses to primary and booster inactivated vaccines in patients with AILD. This study therefore had three main objectives. First, to evaluate whether the primary and booster vaccination would activate autoimmune response or aggravate liver inflammatory. Second, to investigate whether the booster vaccination could elevate the antibody responses in AILD patients. Third, to depict the dynamic changes of immune memory responses after primary and booster vaccination.

Methods

Study design and participants

In this prospective observational study, patients with AILD after the second dose (primary) of inactivated vaccine (BBIBP-CorV from Beijing Institute of Biological Products/Corona-Vac from the Chinese company Sinovac Biotech) were consecutively recruited from the Second Affiliated Hospital of Chongqing Medical University since August 1, 2021. And the third dose (booster) of inactivated vaccine was recommended to population at least 6 months interval after primary vaccination in China. Participants were followed up at 1 month (T1), 3 months (T2) and 6 months (T3) after primary vaccination, and some of them completed the booster inactivated vaccines and continued to follow-up at 1 month (T4) after booster vaccination. The inclusion criteria for patients with AILD were: (1) 18 years of age or older; 2) diagnosed with an AILD by relevant guidelines, including clinical manifestation, autoantibodies, liver function or liver biopsy.16–18 The inclusion criteria for healthy controls (HCs) were: (1) 18 years of age or older; (2) without chronic liver disease, hypertension, diabetes, and other basic diseases. The exclusion criteria were: (1) history of SARS-CoV-2 infection; (2) Hepatitis B/C virus or human immunodeficiency virus infection; (3) other major diseases, such as tumors, renal failure; and (4) pregnancy. In addition, we included, as an HC group, 68 healthcare workers at our hospital over 6 months after primary inactivated vaccination, without personal history of COVID-19 or major comorbidities. All the 68 HCs blood sampling before and after booster vaccination.

Data and sample collection

For all participants recruited in this study, adverse events (AEs) within 7 days were recorded by questionnaire.19 All AEs were recorded and graded according to the scale issued by National Medical Products Administration of China (version 2019). AEs related to vaccination were judged by investigators. Demographic characteristics and clinical data (including autoimmune test and liver function) were obtained by questionnaire or electronic medical record. At each visit, serum was used to test the antibody responses and PBMCs was used to examined the B-cell responses.

Assay of liver function and autoimmune indexes

The liver function indices in serum samples were detected by biochemical detection instrument, including albumin (ALB), aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transferase (GGT), total bilirubin (TB), etc. Autoimmune indexes were detected by western blotting, including antinuclear antibody (ANA), antimitochondrial antibody (AMA), antimitochondrial M2 (AMA-M2), IgA, IgG, IgM, complement 3 (C3), C4, Ig light chains kappa (κ) and lambda (λ), and others.

Assay of anti-RBD-IgG and NAbs

The antireceptor-binding-domain IgG (anti-RBD-IgG) and neutralizing antibodies (NAbs) in serum samples were evaluated using capture chemiluminescence immunoassays by MAGLUMI X8 (Snibe, Shenzhen, China), as previously described.20 The sensitivity and specificity of the kits (Snibe) for anti-RBD-IgG are 100% and 99.6%, respectively, and those for NAbs are 100% and 100%, respectively. The cut-off value was 1.00 AU/mL for anti-RBD-IgG and 0.15 µg/mL for NAbs.

Flow cytometry assay of memory B cells

Detection of SARS-CoV-2-specific memory B cells (MBCs) by flow cytometry was as previously described.21 In brief, for SARS-CoV-2 specific MBCs responses, biotinylated SARS-CoV-2 Spike RBD protein (40592-V08H2-B; Sino Biological, Beijing, China) was mixed with Streptavidin BV421 (405225; Biolegend, San Diego, CA, USA) at 4:1 molar ratio for 1 h at 4°C to obtain the antigen probe. According to the manufacturer’s instruction, peripheral blood mononuclear cells (PBMCs) were stained for 30 m at 4°C using antigen probe (1:33.3) and the following conjugated antibodies: antihuman CD3 (1:50) (300430; Biolegend), antihuman CD19 (1:50) (302212; Biolegend), antihuman CD21 (1:50) (354918; Biolegend), antihuman CD27 (1:50) (356406; Biolegend). After staining, cells were rewashed and resuspended in a 200ul FACS buffer. Samples were then evaluated by flow cytometry (CytoFLEX; Beckman Coulter, Brea, CA, USA) and analyzed using FlowJo (version 10.0.7r2; Treestar, Woodburn, OR, USA).

Statistical analysis

For categorical variables, chi-square and Fisher’s exact tests were used determine significance. For continuous variables, Wilcoxon signed-rank tests were used to compare between-group differences, Mann-Whitney U tests were used for unpaired group comparisons, and Kruskal-Wallis H tests were used for multiple-group comparisons. Change of antibody titers and memory B cells with time were described by using geom_smooth [ggplot2 package] of a linear model. A two-sided p-value of <0.05 was considered significant. Data were analyzed with SPSS (version 24.0.0; IBM Corp., Armonk, NY, USA), and visualized with GraphPad Prism (version 9.2.0; GraphPad Software Inc, La Jolla, CA, USA) and R (version 3.5.3).

Results

Participant characteristics

Overall, 84 patients with AILD and 68 HCs were enrolled between August 1, 2021 and May 6, 2022. Some patients were difficult to complete the follow-up, and did not receive the third dose (booster) of inactivated vaccine due to their own concerns and prevention and control policy of COVID-19 in China. Lastly, of the 84 patients, 15 completed booster inactivated vaccination, 31 provided longitudinal blood samples during the observation period (two to four time points; Table 1). The demographic characteristics of patients with AILD are shown in Table 1 and those of the 68 HCs in are shown in Supplementary Table 1. The median age of AILD patients was 54.9 years (IQR 49.3–60.8). Over half the patients were women (84.5%, 71/84). Of the 84 AILD patients, 47 had AIH, 17 patients had PBC, and 20 had AIH + PBC. A total of 34.5% of patients were receiving immunosuppressive therapy, and glucocorticoids were the most used. Liver function indexes were normal or mild elevated in these patients (Supplementary Table 2). Overall, 145 blood samples from the 84 AILD patients were collected, and of them, 31 patients were followed up. The median elapsed time between primary vaccination and blood sampling in AILD patients was 33.0 (IQR 27.0–43.0) days at T1, 92.0 (82.5–98.0) days at T2, 179.5 (170.5–193.8) days at T3, and 35.0 (21.0–40.0) days at T4 between booster vaccination and blood sampling. The control group underwent blood sampling at 246.5 (191.3–274.8) days at T3 after primary vaccination and at 29.0 (28.0–38.8) days at T4 after booster vaccination.

Table 1

Demographic characteristics of AILD patients

VariableAll patients, n=84AIH, n=47PBC, n=17AIH + PBC, n=20
Age in years54.9 (49.3–60.8)54.5 (48.0–60.0)54.5 (46.0–63.0)56.0 (52.0–58.8)
Sex
  Male13 (15.5)7 (14.9)4 (23.5)2 (10.0)
  Female71 (84.5)40 (85.1)13 (76.5)18 (90.0)
BMI in kg/m222.4 (21.0–23.8)22.7 (21.2–24.1)22.0 (21.0–23.0)21.9 (21.0–23.4)
Therapy
  Immunosuppressants29 (34.5)16 (34.0)013 (65.0)
  Prednisolone/Prednisone27 (32.1)15 (31.9)012 (60.0)
    Azathioprine16 (19.0)9 (19.1)07 (35.0)
    Mycophenolate mofetil2 (2.4)1 (2.1)01 (5.0)
  Other therapiesa52 (61.9)28 (59.6)17 (100.0)7 (35.0)
  Treatment naïve3 (3.6)3 (6.4)00
Liver cirrhosis
  Yes19 (22.6)10 (21.3)6 (35.3)3 (15.0)
  No5 (77.4)37 (78.7)11 (64.7)17 (85.0)
Vaccine
  BBIBP-CorV23 (27.4)14 (29.8)3 (17.6)6 (30.0)
  Corona-Vac54 (64.3)27 (57.4)13 (76.5)14 (70.0)
  mixed7 (8.3)6 (12.8)1 (5.9)0
Days after primary vaccination81.1 (35.8–99.0)81.3 (34.5–98.5)78.0 (30.0–97.0)83.6 (41.5–105.0)
Days after booster vaccination32.3 (21.0–40.0)32.6 (23.0–41.0)31.0 (21.0–35.0)31.7 (12.0–21.0)
Blood collection frequency
  Multiple time points31 (36.9)16 (34.0)9 (52.9)6 (30.0)
  Single-time point53 (63.1)31 (66.0)8 (47.1)14 (70.0)

Safety of inactivated SARS-CoV-2 vaccine in patients with AILD

Firstly, we evaluated the general AEs within 7 days after primary and booster vaccination. As shown in Table 2, the overall incidence of AEs was 26.2% (22/84) after primary vaccination, and the most common local and systemic AEs were pain at injection site (7.1%), fatigue (6.0%), and headache (6.0%). After booster vaccination, only two patients reported AEs (13.3%, 2/15). All AEs were mild or moderate (grade 1 or 2). No severe AEs (grade 3 or 4), such as severe thromboembolism and myocarditis were observed in patients with AILD.

Table 2

Adverse events after primary and booster inactivated vaccination in AILD patients

Adverse eventsPrimary vaccination, n=84Booster vaccination, n=15
Overall, within 7 days22 (26.2%)2 (13.3%)
Local
  Pain6 (7.1%)/
  Swelling2 (2.4%)/
  Redness//
  Itch//
  Induration//
Systemic
  Muscle pain//
  Pruritus//
  Rash//
  Fatigue5 (6.0%)/
  Drowsiness//
  Dizziness/1 (6.7%)
  Headache5 (6.0%)/
  Rhinorrhea//
  Laryngeal pain//
  Fever//
  Chill//
  Cough//
  Inappetence//
  Abdominal pain//
  Abdominal distension//
  Diarrhea//
  Hepatalgia//
  Nausea1 (1.2%)1 (6.7%)
  Chest distress//
  Constipation//
  Numbness of limb2 (2.4%)/
  Lower extremity edema1 (1.2%)/
Grade 3 and 4//

Further, we focused to whether inactivated vaccines led to autoimmune response activation or liver inflammatory aggravation. As shown in Figure 1, autoimmune indexes such as ANA, AMA, C4, IgG and IgM were unchanged before and after primary vaccination (Fig. 1A). However, the C3 level was decreased after primary vaccination (1.1 g/L (IQR 1.0–1.2) vs. 1.0 g/L (0.9–1.1), p<0.01). Meanwhile, Ig light-chain kappa (κ) and lambda (λ) levels were increased (4.3 (3.4–10.1) g/L vs. 10.5 (8.9–12.7) g/L; 2.6 (2.2–5.5) g/L vs. 6.6 (5.1–7.9) g/L, both p<0.001). The results indicate that the autoimmune response was partially activated in AILD patients after primary vaccination. Fortunately, liver function indices such as ALB, ALT, AST, GGT, TB, were unchanged after primary vaccination (Fig. 1B) and after booster vaccination (Fig. 1C). Meanwhile, there were no changes in routine blood indicators after vaccination (Supplementary Fig. 1). In detail, seven patients had mild/moderate increases of liver enzymes (ALT, AST, GGT, etc.) after primary vaccination (Supplementary Table 3). But none of them was judged to be related to vaccine by the investigator after reviewed their past medical and treatment history, due to the aminotransferases increase was related to disease activity, and interruption, or self-modification of treatment regimen. In addition, no one was hospitalized due to this reason. Moreover, we observed four cases of AIH with no AILD before inactivated vaccination by the questionnaire and clinical lab examination. Diagnosis and treatment information are shown in Supplementary Table 4. In brief, no aggravated liver inflammation was seen after primary and booster vaccination in patients with AILD, and one had recurrent abnormal liver function. Taken together, the inactivated vaccines were well tolerated in AILD patients.

Autoimmune responses and liver function changes after primary and booster inactivated vaccination in AILD patients.
Fig. 1  Autoimmune responses and liver function changes after primary and booster inactivated vaccination in AILD patients.

(A) Changes of autoantibodies and immunoglobulins levels before and after primary inactivated vaccination. (B and C) Changes of liver function tests indexes before and after the primary (B) and booster (C) inactivated vaccination in AILD patients. *p<0.05; **p<0.01; ***p<0.001; ns, not significant. ALB, albumin; ALP, alkaline phosphatase; ALT, alanine aminotransferase; AMA, antimitochondrial antibody; AMA-M2, antimitochondrial antibody-M2; ANA, antinuclear antibody; AST, aspartate aminotransferase; C3, complement 3; C4, complement 4; DB, direct bilirubin; GGT, gamma-glutamyl transferase; HB, hemoglobin; Ig, immunoglobulin; PLT, platelet; RBC, red blood cell; TB, total bilirubin; WBC, white blood cell; κ, kappa; λ, lambda.

Antibody response to inactivated SARS-CoV-2 vaccine

Next, we wanted to observe the dynamic changes of antibody response after primary and booster vaccination in this study. As expected, both the seroprevalence and titers of anti-RBD-IgG declined over time in patients with AILD after primary vaccination (87% vs. 45% positive and 7.45 AU/mL (95% confidence interval (CI): 5.0–9.9) vs. 1.7 AU/mL 95% CI: (0.4–3.0) at T1 and T3, respectively, p<0.01) (Fig. 2A). After booster vaccination, almost all the patients had detectable anti-RBD-IgG, and the titer was significantly elevated compared with before booster vaccination (10-fold: 1.7 AU/mL 95% CI: (0.4–3.0) vs. 17.0 AU/mL 95% CI: (8.4–25.7), p<0.001). Similar results were observed for NAbs responses (7.4-fold) (Fig. 2B). The two antibodies were highly correlated (Supplementary Fig. 2). Further longitudinal study analysis showed similar but more obvious trends (Fig. 2C, D). Strikingly, both the titers of anti-RBD-IgG and NAbs in AILD patients were increased as high as in the HCs after booster inactivated vaccination (anti-RBD-IgG: 21.1 AU/mL 95% CI: (8.9–33.3) vs. 26.7 AU/mL 95% CI: (16.9–36.5); NAbs: 1.8 AU/mL 95% CI: (0.6–3.0) vs. 1.7 AU/mL 95% CI: (1.1–2.3), both p>0.05) (Fig. 2E, F).

Antibody responses after primary and booster inactivated vaccinations in AILD patients.
Fig. 2  Antibody responses after primary and booster inactivated vaccinations in AILD patients.

(A and B) Cross-sectional changes of Anti-RBD-IgG (A) and NAbs titers (B) over time after primary and booster vaccination in AILD patients. (C and D) Longitudinal changes of Anti-RBD-IgG (C) and NAbs (D) titers in patients over time after primary and booster vaccination in AILD patients. (E and F) Longitudinal changes of Anti-RBD-IgG (E) and NAbs (F) titers in AILD patients and HCs at T3 and T4 point after vaccination. Dotted lines indicate the detection limit for anti-RBD-IgG and NAbs. The trendlines were produced using a linear model fit, and the shaded area showed the 95% CI for each fit. *p<0.05; **p<0.01; ***p<0.001; ns, not significant. AILD, autoimmune liver disease; anti-RBD-IgG, antireceptor-binding-domain IgG; BV, booster vaccination; HCs, healthy controls; NAbs, neutralizing antibodies; PV, primary vaccination.

As expected, subgroup analysis showed that the anti-RBD-IgG titer after primary vaccination was lower in patients with immunosuppressants than in patients without immunosuppressants (3.9 AU/mL (95% CI: 1.7–6.2) vs. 5.9 AU/mL (95% CI: 4.1–7.8), p<0.05). However, the antibody response was not enhanced significantly in patients with immunosuppressants after booster vaccination, and this discrepancy of anti-RBD-IgG titer between the two subgroups was persisted (5.9 AU/mL (95% CI: 2.3–14.0) vs. 22.6 AU/mL (95% CI: 11.1–34.1), p<0.05) (Fig. 3A). A similar trend was observed in NAbs responses (Fig. 3B). Further, patients with PBC, and AIH without immunosuppressants subgroups showed a significantly enhanced antibody responses after booster vaccination (both p<0.01). However, the difference was not observed in AIH with immunosuppressants and AIH + PBC subgroups (Fig. 3B). Of note, two patients (one AIH with immunosuppressants and one AIH + PBC with immunosuppressants) did not mount an antibody response after the primary vaccination. After booster vaccination, the former was induced a de novo response, but the latter was nonresponse (Fig. 2C and Fig. 3B). Patients with cirrhosis showed a lower trend of antibody responses than patients without cirrhosis after primary and booster vaccination, but the difference was not significant (Fig. 3C). Subgroup analysis in different vaccine types, similar trend was observed between BBIBP-CorV, Corona-Vac and mixed subgroups (Fig. 3D). In brief, booster inactivated vaccines significantly enhanced antibody responses in AILD patients, but not in patients with immunosuppressive therapy.

Subgroup analysis of antibody responses in AILD patients after booster vaccination.
Fig. 3  Subgroup analysis of antibody responses in AILD patients after booster vaccination.

(A) The anti-RBD-IgG (left panel) and NAbs (right panel) titers after booster vaccination in patients with and without immunosuppressants. (B) The anti-RBD-IgG (left panel) and NAbs (right panel) titers after booster vaccination in patients with and without cirrhosis. (C) The anti-RBD-IgG (left panel) and NAbs (right panel) titers after booster vaccination in patients with AIH and PBC/PSC. (D) The anti-RBD-IgG (left panel) and NAbs (right panel) titers in AILD patients after different inactivated vaccines. Dotted lines indicate the detection limit for anti-RBD-IgG and NAbs. *p<0.05; **p<0.01; ***p<0.001; ns, not significant. AIH, autoimmune hepatitis; AILD, autoimmune liver disease; anti-RBD-IgG, antireceptor-binding-domain IgG; BV, booster vaccination; NAbs, neutralizing antibodies; PBC, primary biliary cholangitis; PSC, primary sclerosing cholangitis.

B-cell response to inactivated SARS-CoV-2 vaccine

Lastly, we evaluated the changes of immune memory function in AILD patients after primary and booster vaccination. Overall, the frequency and percentage of total B cells of RBD+ MBCs decreased over time after primary vaccination (6.7 (IQR 5.1–10.1) % at T1 vs. 3.9 (2.8–5.6)% at T3, p<0.05), but had an trend for increase after booster vaccination (3.9 (2.8–5.8)% vs. 5.8 (3.2–8.7) %, p>0.05) (Fig. 4A). Further longitudinal study analysis showed a similar but more obvious trend (p<0.05) (Fig. 4B). To better understand the functional phenotypes of the RBD+ MBCs, we further classed the RBD+ MBCs into four subsets:22 resting MBCs (rMBCs), activated MBCs (actMBCs), atypical MBCs (atyMBCs), and intermediate MBCs (intMBCs). The gating strategy and representative results are shown in Supplementary Figure 3. After primary vaccination, the frequencies of rMBCs and intMBCs decreased over time. On the contrary, the frequency of atyMBCs increased (20.3 (13.9–25.7)% vs. 29.5 (21.2–35.2)%, p<0.05). Interestingly, the frequency of atyMBCs had a decreasing trend (29.5 (21.2–35.2)% vs. 23.3 (18.8–31.0)%, p>0.05) (Fig. 4C) and the frequency of rMBCs and intMBCs increased after booster vaccination (Supplementary Fig. 4A–C). Further longitudinal study analysis showed more obvious trend in atyMBCs (p<0.05) (Fig. 4D) and other subsets (Supplementary Fig. 4D, F). Considering the function of atyMBCs,23 our results indicated that booster inactivated vaccination might restore partly the damaged immune memory function in AILD patients.

RBD-specific memory B cells responses after primary and booster vaccination in patients with AILD.
Fig. 4  RBD-specific memory B cells responses after primary and booster vaccination in patients with AILD.

(A and B) Cross-sectional analysis (A) and longitudinal analysis (B) of frequency of RBD+ MBCs over time after primary and booster vaccination in AILD patients. (C and D) Cross-sectional analysis (C) and longitudinal analysis (D) of frequency of atyMBCs over time after primary and booster vaccination in AILD patients. The trendlines were produced using a linear model fit, and the shaded area showed the 95% CI for each fit. *p<0.05; **p<0.01. AILD, autoimmune liver disease; atyMBCs, atypical MBCs; BV, booster vaccination; MBCs, memory B cells; RBD, receptor binding domain; PV, primary vaccination.

Discussion

In this prospective observational study, we focused on the safety and humoral responses in AILD patients after primary and booster inactivated vaccination. The main findings of this study are: (1) inactivated vaccines were safe for AILD patients; (2) booster dose of inactivated vaccine significantly enhanced the antibody responses in AILD patients without immunosuppressants; (3) booster vaccination partially repaired the impaired immune memory function in AILD patients. Therefore, booster dose of inactivated vaccine is recommended for patients with AILD.

The SARS-CoV-2 vaccine might induce autoimmune responses or aggravate autoimmune diseases.24–26 Several instances of people who developed AIH after mRNA or adenovirus vector vaccines have been reported.3–8 Hence, the safety profile in AILD patients after primary and booster inactivated vaccination was evaluated in this study. The overall occurrence of AEs in AILD patients after primary vaccination was 26.2%, which was lower than that reported in previous studies of patients with severe liver disease (33.3%),27 but was higher than that in chronic hepatitis B patients (14.1%).21 Interestingly, the total incidence of AEs after booster vaccination was significantly decreased (13.3%). A similar phenomenon was reported in a previous study.28 Moreover, although IgG is the main indicator of AIH immune activity, C3 is also used to evaluate immunoinflammatory activity in some autoimmune diseases (such as primary glomerulonephritis, and systemic lupus erythematosus nephritis). When liver function is impaired, complement synthesis is affected, and C3 can also decrease. Our results showed the level of C3 was decreased after primary vaccination, which indicated that the autoimmune response might be partially activated in AILD patients. However, significant vaccine-related elevation of liver enzymes levels was not observed. The reason may be that different type of vaccines were used and most patients are receiving treatment in this study. Altogether, both the primary and booster inactivated vaccines were safe in patients with AILD.

Next, we evaluated dynamic changes of antibody responses after primary and booster vaccination in AILD patients. After primary vaccination, the titers of anti-RBD-IgG and NAbs declined over time, which was consistent with a previous study of BNT162b2 Vaccine in HCs.29 After booster vaccination, both antibody titers significantly increased, and were higher than those 1 month after primary vaccination, which was similar to previous results in healthy individuals.30–32 This indicated the immune memory was stimulated after the booster dose of inactivated vaccine in AILD patients.33 Notably, both the anti-RBD-IgG and NAbs titers in AILD patients were elevated as high as a control group of health care workers after booster vaccination. A recent study15 and our previous data14 have shown the inadequate immune responses at early stage after primary vaccination in AILD patients. Therefore, this result indicated that booster vaccination reversed the poor antibody responses of patients with AILD. Unfortunately, booster vaccination did not significantly enhance the antibody responses in patients receiving immunosuppressants, which differed from the results in immunocompromised patients given an mRNA booster dose.34 The vaccine type may contribute to this discrepancy. Similarly, the lower antibody titers in patients with immunosuppressants than in patients without immunosuppressants were observed after the booster vaccination.

Although several previous studies have shown weakened antibody responses to COVID-19 vaccines in patients with cirrhosis,35–38 the finding is still controversial39 Because some studies found no differences in the humoral responses of cirrhotic and noncirrhotic patients.40,41 In this study, the antibody responses in the two groups were similar (data not shown), which may be explained by the small sample size or different vaccine types.

Lastly, we found that the frequency of aytMBCs increased significantly over time after primary vaccination in AILD patients. AtyMBCs are short-lived activated cells with low binding to the spike protein of SARS-CoV-223 and was also increased in patients with common variable immunodeficiency,42 solid tumors,43 and severe liver diseases.27 This indicated that the immune memory function was damaged over time in patients with AILD. Interestingly, the percentage of atyMBCs was decreased after booster vaccination, which suggests that booster vaccination may have partially restored impaired immune memory.

Our study had some limitations. First, the sample size was relatively small. Because COVID-19 patients were unwilling or unable to come to the hospital to take part in the trial, enrollment and follow-up were difficult. Second, the observation period after booster vaccination was short, which hindered extended analysis of changes of the immune response longer than 6 months after booster vaccination. Third, T-cell responses after primary and booster vaccination in AILD patients were not analyzed. A larger and well-designed study is needed in future. Despite some of these limitations, we believe our findings are still important and meaningful to clinicians. In conclusion, our results indicate that the inactivated SARS-CoV-2 vaccine was well tolerated in patients with AILD, regardless of primary or booster vaccination. Booster vaccination significantly enhanced the antibody responses of AILD patients without immunosuppressants and may partially recover immune memory function.

Supporting information

Supplementary Table 1

Characteristics of healthy controls.

(DOCX)

Supplementary Table 2

Clinical characteristics of AILD patients.

(DOCX)

Supplementary Table 3

Characteristics of patients with liver enzymes elevated after inactivated vaccination.

(DOCX)

Supplementary Table 4

Characteristics of four patients who developed AIH after inactivated vaccination.

(DOCX)

Supplementary Fig. 1

Blood routine examination after primary inactivated vaccination in AILD patients.

The RBC levels (A), Hb levels (B), WBC levels (C), Lymphocyte levels (D) and PLT levels (E) before and after primary inactivated vaccination in patients with AILD. AILD, autoimmune liver disease; RBC, red blood cell; Hb, hemoglobin; WBC, white blood cell; PLT, platelet; ns, not significant.

(TIF)

Supplementary Fig. 2

Correlation between Anti-RBD-IgG and NAbs titers after vaccination in AILD patients.

Anti-RBD-IgG, antireceptor-binding-domain IgG; NAb, neutralizing antibodies; AILD, autoimmune liver disease.

(TIF)

Supplementary Fig. 3

Gating strategies of SARS-CoV-2-specific B cells. RBD, receptor binding domain.

(TIF)

Supplementary Fig. 4

MBC changes over time after primary and booster inactivated vaccination in AILD patients.

(A–C) The overall frequencies of rMBCs (A), intMBCs (B) and actMBCs (C) after primary and booster vaccination. (D–F) Longitudinal dynamics of rMBCs (D), intMBCs (E) and actMBCs (F) after primary and booster vaccination in patients with AILD. *p<0.05; **p<0.01. MBCs, memory B cells; AILD, autoimmune liver diseases; rMBCs, resting MBCs; intMBCs, intermediate MBCs; actMBCs, active MBCs.

(TIF)

Abbreviations

AEs: 

adverse events

AIH: 

autoimmune hepatitis

AILD: 

autoimmune liver disease

ALB: 

albumin

ALP: 

alkaline phosphatase

ALT: 

alanine aminotransferase

AMA: 

antimitochondrial antibody

AMA-M2: 

antimitochondrial antibody-M2

ANA: 

antinuclear antibody

AST: 

aspartate aminotransferase

BMI: 

body mass index

CI: 

confidential interval

C3: 

complement 3

C4: 

complement 4

DB: 

direct bilirubin

GGT: 

gamma-glutamyl transferase

HB: 

hemoglobin

HCs: 

health care workers

Ig: 

immunoglobulin

MBCs: 

memory B cells

NAb: 

neutralizing antibody

PBC: 

primary biliary cirrhosis

PBMC: 

peripheral blood mononuclear cell

PLT: 

platelet

RBC: 

red blood cell

RBD: 

receptor binding domain

SAE: 

serious adverse event

SARS-CoV-2: 

severe acute respiratory syndrome coronavirus 2

TB: 

total bilirubin

WBC: 

white blood cell

Declarations

Acknowledgement

We thank the Health Care Center and Department of Clinical Laboratory of the Second Affiliated Hospital, Chongqing Medical University for their support.

Ethical statement

This study was approved by the Ethics Committee of the Second Affiliated Hospital of Chongqing Medical University and in accordance with the ethical guidelines of the Declaration of Helsinki (Ratification No. 94/2021). Written informed consent was obtained from all participants. This study has been registered at ClinicalTrials.gov (NCT05007665).

Data sharing statement

The datasets used and analyzed during the current study are available from the corresponding author upon reasonable request.

Funding

This work was supported by the National Science and Technology Major Project of China (Nos 2017ZX10202203-007, 2017ZX10202203-008, 2018ZX10302206-003) and a pilot project of clinical cooperation between traditional Chinese and western medicine for significant and complicated diseases of National Administration of Traditional Chinese Medicine: hepatic fibrosis. We also acknowledge the support of the National Natural Science Foundation of China (No 81772198), Natural Science Foundation of Chongqing, China (No. cstc2020jcyj-msxmX0389).

Conflict of interest

HR has been an editor-in-chief of Journal of Clinical and Translational Hepatology since 2013, PH and DZ have been associate editors of Journal of Clinical and Translational Hepatology since 2013. The other authors have no conflict of interests related to this publication.

Authors’ contributions

Participated in the conception and design of this study (DC, DZ, HR), project manager and coordinated patient recruitment (DC), coordinated the serological analysis (MC, MP), performed patient recruitment (YW, TH, GZ, LA, QP, YZ, QZ, NL, DZ, DC, PH), performed antibody testing (DX, GZ), acquisition, analysis, or interpretation of data (ZC, YW, HL), drafting of the manuscript (ZC, YW), obtained funding for the study (PH, MP, MC HR). All the authors contributed to the critical review and final approval of the manuscript. All authors were responsible for the decision to submit the manuscript.

References

  1. Hu B, Guo H, Zhou P, Shi ZL. Characteristics of SARS-CoV-2 and COVID-19. Nat Rev Microbiol 2021;19:141-154 View Article PubMed/NCBI
  2. Dagan N, Barda N, Kepten E, Miron O, Perchik S, Katz MA, et al. BNT162b2 mRNA Covid-19 Vaccine in a Nationwide Mass Vaccination Setting. N Engl J Med 2021;384:1412-1423 View Article PubMed/NCBI
  3. Palla P, Vergadis C, Sakellariou S, Androutsakos T. Letter to the editor: Autoimmune hepatitis after COVID-19 vaccination: A rare adverse effect?. Hepatology 2022;75:489-490 View Article PubMed/NCBI
  4. Suzuki Y, Kakisaka K, Takikawa Y. Letter to the editor: Autoimmune hepatitis after COVID-19 vaccination: Need for population-based epidemiological study. Hepatology 2022;75:759-760 View Article PubMed/NCBI
  5. Garrido I, Lopes S, Simoes MS, Liberal R, Lopes J, Carneiro F, et al. Autoimmune hepatitis after COVID-19 vaccine - more than a coincidence. J Autoimmun 2021;125:102741 View Article PubMed/NCBI
  6. Avci E, Abasiyanik F. Autoimmune hepatitis after SARS-CoV-2 vaccine: New-onset or flare-up?. J Autoimmun 2021;125:102745 View Article PubMed/NCBI
  7. Zin Tun GS, Gleeson D, Al-Joudeh A, Dube A. Immune-mediated hepatitis with the Moderna vaccine, no longer a coincidence but confirmed. J Hepatol 2022;76:747-749 View Article PubMed/NCBI
  8. Efe C, Harputluoglu M, Soylu NK, Yilmaz S. Letter to the editor: Liver transplantation following severe acute respiratory syndrome-coronavirus-2 vaccination-induced liver failure. Hepatology 2022;75:1669-1671 View Article PubMed/NCBI
  9. Cao Z, Gui H, Sheng Z, Xin H, Xie Q. Letter to the editor: Exacerbation of autoimmune hepatitis after COVID-19 vaccination. Hepatology 2022;75:757-759 View Article PubMed/NCBI
  10. Efe C, Kulkarni AV, Terziroli Beretta-Piccoli B, Magro B, Stättermayer A, Cengiz M, et al. Liver injury after SARS-CoV-2 vaccination: Features of immune-mediated hepatitis, role of corticosteroid therapy and outcome. Hepatology 2022;76(6):1576-1586 View Article PubMed/NCBI
  11. Czaja AJ. Examining pathogenic concepts of autoimmune hepatitis for cues to future investigations and interventions. World J Gastroenterol 2019;25:6579-6606 View Article PubMed/NCBI
  12. Premkumar M, Kedarisetty CK. Cytokine Storm of COVID-19 and Its Impact on Patients with and without Chronic Liver Disease. J Clin Transl Hepatol 2021;9:256-264 View Article PubMed/NCBI
  13. The World Health Organization. Interim statement on booster doses for COVID-19 vaccination. Available from: https://www.who.int/news/item/04-10-2021-interim-statement-on-booster-doses-for-covid-19-vaccination View Article PubMed/NCBI
  14. Li H, Wang Y, Ao L, Ke M, Chen ZW, Chen M, et al. Association between immunosuppressants and poor antibody responses to inactivated SARS-CoV-2 vaccines in patients with autoimmune liver diseases. J Hepatol 2022;77:S240 View Article PubMed/NCBI
  15. Thuluvath PJ, Robarts P, Chauhan M. Analysis of antibody responses after COVID-19 vaccination in liver transplant recipients and those with chronic liver diseases. J Hepatol 2021;75:1434-1439 View Article PubMed/NCBI
  16. Chinese Society of Hepatology, Chinese Medical Association. Guidelines on the diagnosis and management of autoimmune hepatitis (2021). Zhonghua Nei Ke Za Zhi 2021;60:1038-1049 View Article PubMed/NCBI
  17. Zhang FC, Wang L, Shuai ZW, Wu ZB, Zhang W, Zhang ZL, et al. Recommendations for diagnosis and treatment of primary biliary cholangitis in China (2021). Zhonghua Nei Ke Za Zhi 2021;60:709-715 View Article PubMed/NCBI
  18. Chinese Society of Hepatology, Chinese Medical Association. Guidelines on the diagnosis and management of primary sclerosing cholangitis (2021). Zhonghua Gan Zang Bing Za Zhi 2022;30:169-189 View Article PubMed/NCBI
  19. Kremsner PG, Ahuad Guerrero RA, Arana-Arri E, Aroca Martinez GJ, Bonten M, Chandler R, et al. Efficacy and safety of the CVnCoV SARS-CoV-2 mRNA vaccine candidate in ten countries in Europe and Latin America (HERALD): a randomised, observer-blinded, placebo-controlled, phase 2b/3 trial. Lancet Infect Dis 2022;22(3):329-340 View Article PubMed/NCBI
  20. Chen Z, Zhu P, Liu Z, Zhu B, Yin G, Ming J, et al. Weakened humoral immune responses of inactivated SARS-CoV-2 vaccines in patients with solid tumors. Cancer Commun (Lond) 2023;43(2):280-284 View Article PubMed/NCBI
  21. He T, Zhou Y, Xu P, Ling N, Chen M, Huang T, et al. Safety and antibody response to inactivated COVID-19 vaccine in patients with chronic hepatitis B virus infection. Liver Int 2022;00:1-10 View Article PubMed/NCBI
  22. Ogega CO, Skinner NE, Blair PW, Park HS, Littlefield K, Ganesan A, et al. Durable SARS-CoV-2 B cell immunity after mild or severe disease. J Clin Invest 2021;131:e145516 View Article PubMed/NCBI
  23. Sosa-Hernández VA, Torres-Ruíz J, Cervantes-Díaz R, Romero-Ramírez S, Páez-Franco JC, Meza-Sánchez DE, et al. B Cell Subsets as Severity-Associated Signatures in COVID-19 Patients. Front Immunol 2020;11:611004 View Article PubMed/NCBI
  24. Chen Y, Xu Z, Wang P, Li XM, Shuai ZW, Ye DQ, et al. New-onset autoimmune phenomena post-COVID-19 vaccination. Immunology 2022;165:386-401 View Article PubMed/NCBI
  25. Klok FA, Pai M, Huisman MV, Makris M. Vaccine-induced immune thrombotic thrombocytopenia. Lancet Haematol 2022;9:e73-e80 View Article PubMed/NCBI
  26. Patrizio A, Ferrari SM, Antonelli A, Fallahi P. Worsening of Graves’ ophthalmopathy after SARS-CoV-2 mRNA vaccination. Autoimmun Rev 2022;21:103096 View Article PubMed/NCBI
  27. Chen Z, Zhang Y, Song R, Wang L, Hu X, Li H, et al. Waning humoral immune responses to inactivated SARS-CoV-2 vaccines in patients with severe liver disease. Signal Transduct Target Ther 2022;7:174 View Article PubMed/NCBI
  28. Cao C, Qiu F, Lou C, Fang L, Liu F, Zhong J, et al. Safety of inactivated SARS-CoV-2 vaccines in patients with allergic diseases. Respir Res 2022;23(1):133 View Article PubMed/NCBI
  29. Levin EG, Lustig Y, Cohen C, Fluss R, Indenbaum V, Amit S, et al. Waning Immune Humoral Response to BNT162b2 Covid-19 Vaccine over 6 Months. N Engl J Med 2021;385:e84 View Article PubMed/NCBI
  30. Cheng ZJ, Huang H, Zheng P, Xue M, Ma J, Zhan Z, et al. Humoral immune response of BBIBP COVID-19 vaccination before and after the booster immunization. Allergy 2022;00:1-11 View Article PubMed/NCBI
  31. Hartl J, Rüther DF, Duengelhoef PM, Brehm TT, Steinmann S, Weltzsch JP, et al. Analysis of the humoral and cellular response after the third COVID-19 vaccination in patients with autoimmune hepatitis. Liver Int 2023;43(2):393-400 View Article PubMed/NCBI
  32. Xia S, Duan K, Zhang Y, Zeng X, Zhao D, Zhang H, et al. Safety and Immunogenicity of an Inactivated COVID-19 Vaccine, WIBP-CorV, in Healthy Children: Interim Analysis of a Randomized, Double-Blind, Controlled, Phase 1/2 Trial. Front Immunol 2022;13:898151 View Article PubMed/NCBI
  33. Sette A, Crotty S. Immunological memory to SARS-CoV-2 infection and COVID-19 vaccines. Immunol Rev 2022;310(1):27-46 View Article PubMed/NCBI
  34. Wagner A, Garner-Spitzer E, Schötta AM, Orola M, Wessely A, Zwazl I, et al. SARS-CoV-2-mRNA Booster Vaccination Reverses Non-Responsiveness and Early Antibody Waning in Immunocompromised Patients - A Phase Four Study Comparing Immune Responses in Patients With Solid Cancers, Multiple Myeloma and Inflammatory Bowel Disease. Front Immunol 2022;13:889138 View Article PubMed/NCBI
  35. Thuluvath PJ, Robarts P, Chauhan M. Analysis of antibody responses after COVID-19 vaccination in liver transplant recipients and those with chronic liver diseases. J Hepatol 2021;75(6):1434-1439 View Article PubMed/NCBI
  36. Simão AL, Palma CS, Izquierdo-Sanchez L, Putignano A, Carvalho-Gomes A, Posch A, et al. Cirrhosis is associated with lower serological responses to COVID-19 vaccines in patients with chronic liver disease. JHEP Rep 2023;5(5):100697 View Article PubMed/NCBI
  37. Giambra V, Piazzolla AV, Cocomazzi G, Squillante MM, De Santis E, Totti B, et al. Effectiveness of Booster Dose of Anti SARS-CoV-2 BNT162b2 in Cirrhosis: Longitudinal Evaluation of Humoral and Cellular Response. Vaccines (Basel) 2022;10(8):1281 View Article PubMed/NCBI
  38. Cao H, Huang Y, Zhong C, Liao X, Tan W, Zhao S, et al. Antibody response and safety of inactivated SARS-CoV-2 vaccines in chronic hepatitis B patients with and without cirrhosis. Front Immunol 2023;14:1167533 View Article PubMed/NCBI
  39. Toutoudaki K, Dimakakou M, Androutsakos T. Efficacy, Safety and Immunogenicity of Anti-SARS-CoV-2 Vaccines in Patients with Cirrhosis: A Narrative Review. Vaccines (Basel) 2023;11(2):452 View Article PubMed/NCBI
  40. Singh A, De A, Singh MP, Rathi S, Verma N, Premkumar M, et al. Antibody Response and Safety of ChAdOx1-nCOV (Covishield) in Patients with Cirrhosis: A Cross-Sectional, Observational Study. Dig Dis Sci 2023;68(2):676-684 View Article PubMed/NCBI
  41. Bakasis AD, Bitzogli K, Mouziouras D, Pouliakis A, Roumpoutsou M, Goules AV, et al. Antibody Responses after SARS-CoV-2 Vaccination in Patients with Liver Diseases. Viruses 2022;14(2):207 View Article PubMed/NCBI
  42. Quinti I, Locatelli F, Carsetti R. The Immune Response to SARS-CoV-2 Vaccination: Insights Learned From Adult Patients With Common Variable Immune Deficiency. Front Immunol 2021;12:815404 View Article PubMed/NCBI
  43. Li T, Song R, Wang J, Zhang J, Cai H, He H, et al. Safety and immunogenicity of inactivated SARS-CoV-2 vaccines in people with gastrointestinal cancer. Int J Infect Dis 2022;122:874-884 View Article PubMed/NCBI
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
  • Copyright © 2022 JCTH. All Rights Reserved.
  • Published by Xia & He Publishing Inc.
  • Address: 14090 Southwest Freeway, Suite 300, Sugar Land, Texas 77478, USA
  • Email: service@xiahepublishing.com