1,216
Views
0
CrossRef citations to date
0
Altmetric
Coronavirus

Immunogenicity of COVID-19 vaccines in patients with cirrhosis: A meta-analysis

, &
Article: 2326316 | Received 15 Jan 2024, Accepted 29 Feb 2024, Published online: 11 Mar 2024

ABSTRACT

The immunogenicity of COVID-19 vaccines in patients with liver cirrhosis remains largely unknown. The purpose of this meta-analysis was to investigate the immunogenicity of COVID-19 vaccines in patients with cirrhosis and compare the humoral and cellular immune responses following complete COVID-19 vaccination between cirrhosis patients and healthy controls. A systematic literature search was conducted in PubMed, EMBASE, and Web of Science from 1 January 2020 to 22 August 2023. Sixteen studies with 2127 cirrhosis patients were included. The pooled seroconversion rate in patients with cirrhosis following complete COVID-19 vaccination was 92.4% (95% CI, 86.2%–96%, I2= 90%) with significant between-study heterogeneity. Moreover, COVID-19 vaccination elicited a higher humoral immune response in patients of compensated cirrhosis as compared with decompensated cirrhosis (RR = 1.069, 95% CI, 1.011–1.131, I2= 17%, p = .019). Additionally, 10 studies were included for comparison analysis of seroconversion rate between cirrhosis patients and healthy controls. The results showed that the seroconversion rate in patients with cirrhosis was slightly lower compared with healthy controls (RR = 0.972, 95% CI, 0.955–0.989, I2= 66%, p = .001). Meanwhile, the pooled RR of cellular immune response rate for cirrhosis patients vs. healthy controls was 0.678 (95% CI, 0.563–0.817, I2= 0, p < .0001). Our meta-analysis demonstrated that COVID-19 vaccination elicited diminished humoral and cellular immune responses in patients of cirrhosis. Patients with cirrhosis particularly decompensated cirrhosis who have completed full-doses of COVID-19 vaccination should receive continuous attention and preemptive measures.

View responses to this article:
Immunogenicity of COVID-19 vaccines in patients with cirrhosis: Expected advantage for additional dose of vaccine

Introduction

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the culprit of the coronavirus disease 2019 (COVID-19), has been responsible for almost 770 million cases and more than 6.9 million deaths worldwide by 30th August 2023.Citation1 Patients with comorbidities including chronic liver diseases, particularly cirrhosis, are at a high risk of severe COVID-19 and COVID-19-associated mortality.Citation2–4 Vaccination is the most effective measure to protect the population from severe COVID-19. Therefore, the European Association for the Study of Liver Disease (EASL) and the American Association for the Study of Liver Disease (AASLD) guidelines recommended that patients with liver cirrhosis be prioritized for COVID-19 vaccination.Citation5,Citation6

Cirrhosis is characterized by immune dysregulation with several abnormalities in components of the innate and adaptive immune system, which is associated with poor immune response to vaccines against hepatitis B, influenza, and pneumococcus.Citation7–9 Therefore, it is important to evaluate the immune response against COVID-19 vaccines in patients with liver cirrhosis. Despite the safety and effectiveness of COVID-19 vaccines in phase III, placebo-controlled clinical trials have been reported in the general population,Citation10–12 limited data are available on the immunogenicity of COVID-19 vaccines in patients with cirrhosis. A few observational studies have reported a suboptimal antibody response following COVID-19 vaccination in patients with cirrhosis compared to healthy controls.Citation13–15 However, another study suggested that the seroconversion rate was similar between cirrhotic patients and a reference group of health-care workers.Citation16 In view of the inconsistent results in the above-mentioned studies, the immunogenicity of COVID-19 vaccine in patients with cirrhosis remains controversial. A meta-analysis with large clinical samples is warranted to draw a reliable conclusion.

In this study, we performed a systematic review and quantitative meta-analysis to compare the humoral and cellular immune response following COVID-19 vaccination in patients with cirrhosis and healthy controls.

Methods

Search strategy

We searched PubMed, EMBASE, and Web of Science for available studies published in English from 1 January 2020 to 22 August 2023. To identify all the articles regarding the immunogenicity of COVID-19 vaccines in patients with cirrhosis, we used the following keywords alone or in combination for literature search: “SARS-CoV-2,” “COVID-19,” “2019-nCoV,” “severe acute respiratory syndrome coronavirus 2,” “liver disease,” “liver cirrhosis,” “hepatic cirrhosis,” “vaccine,” “vaccination,” and “immunization.” References of the included studies were also manually searched to select additional studies. This systematic review and meta-analysis were conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guideline.Citation17

Inclusion and exclusion criteria

Studies were included if they met the following criteria: (1) Study population: adult patients with liver cirrhosis following full dose of COVID-19 vaccination without prior COVID-19 infection; (2) Outcomes: We calculated the pooled seroconversion rate of anti-SARS-CoV-2 spike antibody or anti-receptor-binding domain (RBD) antibody or neutralizing antibody (Nab) against SARS-CoV-2 in patients of liver cirrhosis following vaccination. The seroconversion rate was reported in the following sequence: anti-spike antibody, anti-RBD antibody, or neutralization antibody. The pooled relative risk (RR) for seroconversion rate in patients with compensated cirrhosis (CC) as compared to patients with decompensated cirrhosis (DC) was estimated. We also compared the seroconversion rate and detectable T-cell immune response rate between patients with liver cirrhosis and healthy controls.

Exclusion criteria included: (1) case reports, reviews, meta-analyses, guidelines, editorials, and comments; (2) reports with less than 20 patients; (3) publications with insufficient data and studies in non-English languages; (4) If participants with and without prior COVID-19 infection were enrolled in a study, subjects with prior COVID-19 infection were excluded. The flow diagram of the study selection has been drafted in accordance with the PRISMA principle.Citation17

Data extraction and quality assessment

Two authors screened independently to determine which studies should be included, and any discrepancy was resolved by a third author. The extracted data included the first author, publication date, country, gender and mean age of participants, type of vaccine, time interval between the second dose of vaccination and antibody testing, type of antibody, method of antibody testing, and outcomes of interest. The data are shown to be median and interquartile range were transformed into mean and standard deviation (SD) according to the formula below (https://smcgrath.shinyapps.io/estmeansd/). The quality of the included observational studies was assessed using the Newcastle-Ottawa scale (NOS) which consists of three categories (quality of the selection, comparability, and the outcome of study). Scores range from 0 to 9, and the risk of bias in the studies was considered as low risk (7–9 points), moderate risk (4–6 points), and high risk (0–3 points), respectively.

Statistical analysis

Statistical analyses were performed using R statistical software version 4.2.2.Citation18 The “metaprop” command in R was performed to estimate the pooled seroconversion rate with 95% confidence interval (CI) in patients, with liver cirrhosis following full-dose of COVID-19 vaccination. The “metabin” function was used to estimate the risk ratio (RR) with 95% confidence interval of humoral and cellular immune response rates between patients and healthy controls as well as between CC and DC. The I2 statistic was used to evaluate the heterogeneity of the pooled estimate. Heterogeneity was considered as low (<25%), moderate (25%–75%) and high (>75%). If there was no evidence of between-studies heterogeneity (I2 ≤ 50%), a fixed-effects model was used to combine the data. Otherwise, a random-effects model was selected. To explore the potential source of heterogeneity, subgroup analysis and meta-regression based on type of vaccine, type of antibody, method of antibody testing, the gender, mean age of participants and time interval between the second dose of vaccination and antibody testing were performed. Sensitivity analysis was performed using a leave-one-out method to confirm the robustness of outcomes. Publication bias was determined by funnel plot and Egger's test if the number of included studies >10. A trim-and-fill method was used to adjust for publication bias. p value less than .05 was regarded as statistically significant.

Results

Study characteristics

A total of 4074 articles were identified according to the search terms. Firstly, duplicated articles (n = 968) were excluded. After reviewing the titles and abstracts, 3007 articles were ruled out and 99 articles underwent full-text screening. Eighty-three articles were excluded due to the following reasons: reviews, editorials, comments, studies with wrong patient population or overlapping cohort or no outcome of interest. Finally, 16 articles with 2127 cirrhosis patients and 843 healthy controls were included in our meta-analysis. shows the full PRISMA flow diagram of study selection. The characteristics of included studies are shown in . Most of the studies were from Europe and Asia, with only one study from Americas. The quality of the studies was evaluated and shown in . Eight studies showed a low risk of bias and other eight studies showed a moderate risk of bias. The time interval between the last dose and antibody testing ranged from 14 to 112 days.

Figure 1. PRISMA flowchart of included and excluded studies.

Figure 1. PRISMA flowchart of included and excluded studies.

Table 1. Characteristics of the included studies.

Table 2. Quality assessment of the included studies by Newcastle-Ottawa scale (NOS).

Humoral immune response after full dose of COVID-19 vaccination in cirrhosis patients

Sixteen eligible studies reported humoral immune response in 2127 cirrhosis patients following a full dose of COVID-19 vaccination. Among the 16 studies, 4 studiesCitation19–22 reported both Nab and anti-spike antibody, 2 studies only reported NabCitation15,Citation23 and 10 studies only reported anti-spike or anti-RBD antibody.Citation13,Citation14,Citation16,Citation24–30 The pooled seroconversion rate (14 studies of anti-spike and/or anti-RBD and 2 studies of Nab) was 92.4% (95% CI, 86.2%–96%) () with considerable heterogeneity (I2= 90%, p < .01). Sensitivity analysis showed that the pooled estimate was robust (Supplemental Figure S1). The funnel plot (Supplemental Figure S2) and the result of Egger's test (p < .0001) indicated the existence of publication bias. Trim-and-fill method was used to adjust for publication bias, and the pooled seroconversion rate was 80.1% (95% CI, 60.5%–91.3%) after adjusting.

Figure 2. Forest plot of the seroconversion rate following complete COVID-19 vaccination in patients of cirrhosis. CI, confidence interval.

Figure 2. Forest plot of the seroconversion rate following complete COVID-19 vaccination in patients of cirrhosis. CI, confidence interval.

Subgroup analysis and meta-regression

Subgroup analysis was performed to identify potential sources of heterogeneity based on type of antibody, type of vaccine and method of antibody testing. The pooled seroconversion rate of anti-spike antibody in cirrhosis patients was significantly higher than that of neutralizing antibody (94% (95% CI, 88.4%–97%) vs 74.8% (95% CI, 64.3%–83%), p < .01) (Supplemental Figure S3). The pooled seroconversion rate after complete vaccination with mRNA vaccine, adeno-associated virus (AAV), and inactivated vaccine was 94.7% (95% CI, 88.7%–97.6%), 93.7% (95% CI, 70.2%–98.9%) and 81.9% (95% CI, 62%–92.6%), respectively (Supplemental Figure S4). The pooled seroconversion rate detected with ECLIA, CLIA, and ELISA was 95.1% (95% CI, 71.9%–99.3%), 89.1% (95% CI, 78.8%–94.8%), and 96.5% (95% CI, 91.2%–98.6%), respectively (Supplemental Figure S5).

Meta-regression shows a trend toward significance between seroconversion rate and antibody type (p = .06). However, no association of pooled seroconversion rate with type of vaccine (p = .11), method of antibody testing (p = .3), time interval between the last dose and antibody testing (p = .76), mean age (p = .62), and male ratio (p = .77) were found.

Comparison of humoral immune response between compensated cirrhosis and decompensated cirrhosis

Nine studies compared the humoral immune response between compensated cirrhosis (n = 955) and decompensated cirrhosis (n = 486) after complete vaccination. The seroconversion rate in patients with compensated cirrhosis was higher as compared to patients with decompensated cirrhosis (RR = 1.069, 95% CI = 1.011–1.131, I2= 17%, p = .019) (). The sensitivity analysis confirmed the robustness of the result (Supplemental Figure S6).

Figure 3. Forest plot of the comparison of seroconversion rate between compensated cirrhosis and decompensated cirrhosis.

Figure 3. Forest plot of the comparison of seroconversion rate between compensated cirrhosis and decompensated cirrhosis.

Comparison of humoral immune response between cirrhosis patients and healthy controls

There were 10 studies (1077 patients and 843 controls) that compared seroconversion rate following complete COVID-19 vaccination between cirrhosis patients and healthy controls. Compared with healthy controls, the cirrhosis patients exhibited a slightly lower seroconversion rate with moderate heterogeneity (RR = 0.972, 95% CI = 0.955–0.989, I2 = 66%, p = .001) (). The RR value did not change markedly by sensitivity analysis, confirming the stability of the result (Supplemental Figure S7). Subgroup by type of vaccine demonstrated significant between-group differences with a higher rate of heterogeneity in the group of inactivated vaccines (Supplemental Figure S8).

Figure 4. (a) Forest plot of the comparison of humoral immune response between cirrhosis patients and healthy controls; (b) Forest plot of the comparison of cellular immune response between cirrhosis patients and healthy controls.

Figure 4. (a) Forest plot of the comparison of humoral immune response between cirrhosis patients and healthy controls; (b) Forest plot of the comparison of cellular immune response between cirrhosis patients and healthy controls.

Comparison of cellular immune response between cirrhosis patients and healthy controls

Only two studies reported detectable T-cell immune response rate in 68 patients and 50 controls. The T-cell immune response rate following complete COVID-19 vaccination in cirrhosis patients was significantly lower as compared with healthy controls (RR = 0.678, 95% CI = 0.563–0.817, I2= 0, p < .0001) (). We compare the method used to analyze cellular immunity and type of vaccine employed between the two studies. Both two studies quantified interferon-gama (IFN-γ) production which was released by T cells upon in vitro stimulation with peptides derived from the SARS-COV-2 spike antigen to analyze cellular immunity. Although both studies used ELISA as the detection method, the ELISA test kit used in the two studies was from different companies, resulting in difference in cutoff values which were regarded as positive cellular immunity. In addition, patients and healthy controls in both two studies mainly received full doses of mRNA vaccination. Although the method applied and type of vaccine employed in the two studies were similar, the results of the cellular immunity should be interpreted with caution considering only two studies were included in the meta-analysis.

Discussion

To our knowledge, this is the first meta-analysis to report immunogenicity of COVID-19 vaccines in patients with liver cirrhosis. Our results show that the pooled seroconversion rate following complete COVID-19 vaccination was 92.4% in patients with cirrhosis with considerable heterogeneity. The humoral immune response in patients with decompensated cirrhosis was significantly lower as compared with compensated cirrhosis. In addition, both the humoral and cellular immune response in patients with cirrhosis were impaired when compared to healthy controls.

Up to now, only two meta-analysesCitation31,Citation32 have reported the seroconversion rate after COVID-19 vaccination in patients with chronic liver disease, in which subgroup analysis based on cirrhosis status has been performed. The pooled rate of humoral immune response in the cirrhosis patients was found 94% (95% CI, 82%–100%) in one study (328 patients)Citation32 and 85% (95% CI, 75%–91%) in another study (406 patients).Citation31 Our study observed that the pooled seroconversion rate was 92.4% (95% CI, 86.2%–96%) in 2127 patients of cirrhosis with considerable heterogeneity. This inconsistency may be explained by different sample sizes and types of detected antibodies. As all of the included articles in our meta-analysis, except for two reported seroconversion rates of anti-spike or anti-RBD antibody, we calculated the pooled seroconversion rate mainly based on data of anti-spike or anti-RBD antibody with only two data of Nab. Neutralizing antibody has been considered as proxy indicator of protection from symptomatic COVID-19 infection.Citation33 Studies have demonstrated that measurement of anti-spike or anti-RBD IgG results in a slightly higher seroconversion rate than that of Nab.Citation19–22 Consistent with these studies, subgroup analysis in our study confirmed that the pooled seroconversion rate of anti-spike or anti-RBD antibody in cirrhosis patients was 94% (95% CI, 88.4%–97%), which was significantly higher than that of neutralizing antibody (74.8%; 95% CI, 64.3%–83%).

Patients with cirrhosis display cirrhosis-associated immune dysfunction (CAID) that predisposes patients to bacterial and viral infections as well as poor response to various vaccinations.Citation34 Several studies have showed that patients with cirrhosis, particularly decompensated cirrhosis, have diminished antibody levels after COVID-19 vaccination compared with healthy controls.Citation13–15,Citation27 Recently, Simão et al. have reported that cirrhosis is associated with a lower serological response to COVID-19 vaccines in patients with chronic liver disease.Citation35 In line with these studies, our results suggested that the cirrhosis patients exhibited a slightly lower seroconversion rate after COVID-19 vaccination compared to healthy controls. Additionally, patients with decompensated cirrhosis manifested inferior vaccine response compared to patients with compensated cirrhosis.

T-cell responses have been associated with protection from severe COVID-19 infection despite a lack of seroconversion.Citation36,Citation37 Expectedly, similar to the result found in humoral immune response, we observed that T-cell immune response was significantly impaired compared to the healthy controls. However, only two studies were combined, although no heterogeneity was found and the result should be interpreted with caution. More studies regarding cellular immune response in cirrhosis patients are needed to provide a reliable conclusion.

Given that cirrhosis patients exhibit a diminished humoral and cellular immune response following COVID-19 vaccination compared to healthy controls, measures aim to enhance the immunization rate, e.g. further booster doses or heterologous vaccination which has been observed in other conditions characterized by impaired immunity should be considered in patients with cirrhosis.Citation38,Citation39 Currently, there were limited studies reporting the immune responses to the additional booster doses in patients with cirrhosis. A recent study demonstrated that the booster dose of COVID-19 vaccination significantly increased the SARS-COV-2 specific wild-type antibody levels.Citation40 Another study observed higher immunogenicity of both humoral and cellular responses to the heterologous COVID-19 vaccine regimen compared to homogeneous regimens in patients with cirrhosis.Citation41 Therefore, we speculate that further booster doses of COVID-19 vaccination with heterologous regimens could possibly enhance the immune response of patients with cirrhosis.

Even though humoral and cellular immune responses to COVID-19 vaccination seem to be inferior in patients with cirrhosis, evidence provided by real-world study has demonstrated the effectiveness of COVID-19 vaccines in patients with cirrhosis as shown in reduced rates of SARS-COV-2 infection, severe COVID-19 and COVID-19 related mortalities.Citation42,Citation43 Administration of a third dose of COVID-19 vaccine is associated with a reduced risk of breakthrough infection, severe/critical COVID-19, and COVID-19-related death compared to two doses of vaccine.Citation44,Citation45 With the continuing emergence of novel viral variants, the effectiveness of COVID-19 vaccination could be possibly faded, as evidenced by the finding that serological titers to Omicron subvariants and neutralization activity against Omicron subvariants in patients with cirrhosis following 2 or 3 doses of COVID-19 vaccination were significantly lower compared to wild-type SARS-COV-2.Citation40,Citation46 In addition, another study demonstrated that COVID-19 vaccination was less effective against symptomatic COVID-19 during the Omicron period compared to the Alpha and Delta periods.Citation47 However, they also found that vaccine effectiveness against severe/critical COVID-19 was highest during Omicron and lowest during the Delta period.Citation47 The above evidences provide reassurance to patients with cirrhosis and clinicians treating them.

Inevitably, the present study has several limitations. First, a significant magnitude of heterogeneity exists in our study. To explore the potential origin, we performed subgroup analysis and meta-regression based on antibody type, type of vaccine, method of antibody testing, time interval between the last dose and antibody testing, mean age, and male ratio. However, no significant association of pooled seroconversion rate with these above factors except antibody type was found. Studies have suggested that comorbidities (diabetes, obesity, chronic kidney disease, etc.)Citation48–50 and the etiology of cirrhosis (particularly autoimmune hepatitis taking suppression drugs)Citation51 are associated with the inferior humoral immune response of COVID-19 vaccine. However, the individual study included in this present meta-analysis comprised a heterogeneous population of various comorbidities and disease etiology. Subgroup analysis and meta-regression regarding specific comorbidity and disease etiology could not be performed due to lack of data. Second, all of the included studies are observational which could result in potential bias and cannot be adjusted by meta-analysis. Third, the test kits used to detect antibody and the cutoff value to define a positive seroconversion vary among different studies. Finally, only 2 studies are available to retrieve data regarding the rate of cellular immune response following COVID-19 vaccination, which might not be sufficient.

In conclusion, our study demonstrated that COVID-19 vaccination elicited a diminished humoral and cellular immune response in patients with cirrhosis. Booster vaccination may be considered in patients with cirrhosis who have completed full doses of COVID-19 vaccination. As the continuous emergence of novel SARS-COV-2 variants, self-protective behaviors should be followed by cirrhosis patients even if they have received COVID-19 vaccination. An updated meta-analysis, which includes more well-designed clinical trials with large sample size, is needed in the future.

Author contributions

Lichen Ouyang designed the search strategy, screened studies for eligibility, conducted the data analysis, and wrote the first draft of the manuscript. Yeli Gong conceived the study, screened studies for eligibility, and critically revised the manuscript. Gang Lei addressed discrepancies in screening and data extraction and critically revised the manuscript. All coauthors were involved in the final editing of the manuscript. The corresponding authors attest that all listed authors meet authorship criteria and that no others meeting the criteria have been omitted.

Supplemental material

Supplementary file 1.docx

Download MS Word (1.7 MB)

Disclosure statement

No potential conflict of interest was reported by the author(s).

Supplementary material

Supplemental data for this article can be accessed on the publisher’s website at https://doi.org/10.1080/21645515.2024.2326316

Additional information

Funding

This research was funded by the Research Fund of Jianghan University [Grant No.2021yb135].

References

  • World Health Organization. Coronavirus (COVID-19) Dashboard. [ accessed 2023 Aug 30]. https://covid19.who.int/.
  • Middleton P, Hsu C, Lythgoe MP. Clinical outcomes in COVID-19 and cirrhosis: a systematic review and meta-analysis of observational studies. BMJ Open Gastroenterol. 2021;8(1):e000739. doi:10.1136/bmjgast-2021-000739.
  • Ge J, Pletcher MJ, Lai JC, Consortium NC. Outcomes of SARS-CoV-2 infection in patients with chronic liver disease and cirrhosis: a national COVID cohort collaborative study. Gastroenterology. 2021;161:1487–501 e5. doi:10.1053/j.gastro.2021.07.010.
  • Kovalic AJ, Satapathy SK, Thuluvath PJ. Prevalence of chronic liver disease in patients with COVID-19 and their clinical outcomes: a systematic review and meta-analysis. Hepatol Int. 2020;14(5):612–9. doi:10.1007/s12072-020-10078-2.
  • Fix OK, Blumberg EA, Chang KM, Chu J, Chung RT, Goacher EK, Hameed B, Kaul DR, Kulik LM, Kwok RM, et al. American association for the study of liver diseases expert panel consensus statement: vaccines to prevent coronavirus disease 2019 infection in patients with liver disease. Hepatology. 2021;74(2):1049–64. doi:10.1002/hep.31751.
  • Marjot T, Eberhardt CS, Boettler T, Belli LS, Berenguer M, Buti M, Jalan R, Mondelli MU, Moreau R, Shouval D, et al. Impact of COVID-19 on the liver and on the care of patients with chronic liver disease, hepatobiliary cancer, and liver transplantation: an updated EASL position paper. J Hepatol. 2022;77(4):1161–97. doi:10.1016/j.jhep.2022.07.008.
  • Harmala S, Parisinos CA, Shallcross L, O’Brien A, Hayward A. Effectiveness of influenza vaccines in adults with chronic liver disease: a systematic review and meta-analysis. BMJ Open. 2019;9(9):e031070. doi:10.1136/bmjopen-2019-031070.
  • McCashland TM, Preheim LC, Gentry MJ. Pneumococcal vaccine response in cirrhosis and liver transplantation. J Infect Dis. 2000;181(2):757–60. doi:10.1086/315245.
  • Aggeletopoulou I, Davoulou P, Konstantakis C, Thomopoulos K, Triantos C. Response to hepatitis B vaccination in patients with liver cirrhosis. Rev Med Virol. 2017;27(6):27. doi:10.1002/rmv.1942.
  • Baden LR, El Sahly HM, Essink B, Kotloff K, Frey S, Novak R, Diemert D, Spector SA, Rouphael N, Creech CB, et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med. 2021;384(5):403–16. doi:10.1056/NEJMoa2035389.
  • Polack FP, Thomas SJ, Kitchin N, Absalon J, Gurtman A, Lockhart S, Perez JL, Pérez Marc G, Moreira ED, Zerbini C, et al. Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine. N Engl J Med. 2020;383(27):2603–15. doi:10.1056/NEJMoa2034577.
  • Voysey M, Clemens SAC, Madhi SA, Weckx LY, Folegatti PM, Aley PK, Angus B, Baillie VL, Barnabas SL, Bhorat QE, et al. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet. 2021;397(10269):99–111. doi:10.1016/S0140-6736(20)32661-1.
  • Al-Dury S, Waern J, Waldenström J, Alavanja M, Saed HH, Törnell A, Arabpour M, Wiktorin HG, Einarsdottir S, Ringlander J, et al. Impaired SARS-CoV-2-specific T-cell reactivity in patients with cirrhosis following mRNA COVID-19 vaccination. JHEP Rep. 2022;4(7):100496. doi:10.1016/j.jhepr.2022.100496.
  • Willuweit K, Frey A, Passenberg M, Korth J, Saka N, Anastasiou OE, Möhlendick B, Schütte A, Schmidt H, Rashidi-Alavijeh J, et al. Patients with liver cirrhosis show high immunogenicity upon COVID-19 vaccination but develop premature deterioration of antibody titers. Vaccines (Basel). 2022;10(3):10. doi:10.3390/vaccines10030377.
  • Ai J, Wang J, Liu D, Xiang H, Guo Y, Lv J, Zhang Q, Li J, Zhang X, Li Q, et al. Safety and immunogenicity of SARS-CoV-2 vaccines in patients with chronic liver diseases (CHESS-NMCID 2101): a multicenter study. Clin Gastroenterol Hepatol. 2022;20:1516–24 e2. doi:10.1016/j.cgh.2021.12.022.
  • Biliotti E, Caioli A, Sorace C, Lionetti R, Milozzi E, Taibi C, Visco Comandini U, Maggi F, Puro V, D’Offizi G, et al. Humoral immune response after COVID-19 mRNA vaccination in patients with liver cirrhosis: a prospective real-life single center study. Biomedicines. 2023;11(5):11. doi:10.3390/biomedicines11051320.
  • Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, Shamseer L, Tetzlaff JM, Akl EA, Brennan SE, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. Syst Rev. 2021;10(1):89. doi:10.1186/s13643-021-01626-4.
  • Balduzzi S, Rucker G, Schwarzer G. How to perform a meta-analysis with R: a practical tutorial. Evid Based Ment Health. 2019;22(4):153–60. doi:10.1136/ebmental-2019-300117.
  • Bakasis A-D, Bitzogli K, Mouziouras D, Pouliakis A, Roumpoutsou M, Goules AV, Androutsakos T. Antibody responses after SARS-CoV-2 vaccination in patients with liver diseases. Viruses-Basel. 2022;14:14. doi:10.3390/v14020207.
  • Song R, Yang C, Wang J, Li Q, Chen J, Sun K, Lv H, Yang Y, Liang J, Ye Q, et al. Immune response of primary and booster immunity of SARS-CoV-2 vaccination among patients with chronic liver disease. medRxiv. 2022;14.
  • Goel A, Verma A, Tiwari P, Katiyar H, Aggarwal A, Khetan DM, Kishore RVK, Kumar P, Singh TP, Sheikh S, et al. Serological immune response following ChAdOx1 nCoV-19 vaccine (Covishield®) in patients with liver cirrhosis. Nato Adv Sci Inst Se. 2022;10:10. doi:10.3390/vaccines10111837.
  • Chen Z, Zhang Y, Song R, Wang L, Hu X, Li H, Cai D, Hu P, Shi X, Ren H. Waning humoral immune responses to inactivated SARS-CoV-2 vaccines in patients with severe liver disease. Sig Transduct Target Ther. 2022;7(1). doi:10.1038/s41392-022-01032-9.
  • Wang J, Zhang Q, Ai J, Liu D, Liu C, Xiang H, Gu Y, Guo Y, Lv J, Huang Y, et al. Safety and immunogenicity of SARS-CoV-2 vaccines in Chinese patients with cirrhosis: a prospective multicenter study. Hepatol Int. 2022;16:691–701. doi:10.1007/s12072-022-10332-9.
  • 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):1434–9. doi:10.1016/j.jhep.2021.08.008.
  • Singh A, De A, Singh MP, Rathi S, Verma N, Premkumar M, Taneja S, Duseja A, Singh V. Antibody response and safety of ChAdOx1-nCOV (covishield) in patients with cirrhosis: a cross-sectional, observational study. Dig Dis Sci. 2023;68:676–84. doi:10.1007/s10620-022-07641-2.
  • Giambra V, Piazzolla AV, Cocomazzi G, Squillante MM, De Santis E, Totti B, Cavorsi C, Giuliani F, Serra N, Mangia A, 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):10. doi:10.3390/vaccines10081281.
  • Kulkarni AV, Jaggaiahgari S, Iyengar S, Simhadri V, Gujjarlapudi D, Rugwani H, Vemula VK, Gora BA, Shaik S, Sharma M, et al. Poor immune response to coronavirus disease vaccines in decompensated cirrhosis patients and liver transplant recipients. Vaccine. 2022;40(48):6971–8. doi:10.1016/j.vaccine.2022.10.042.
  • Cossiga V, Capasso M, Guarino M, Loperto I, Brusa S, Cutolo FM, Attanasio MR, Lieto R, Portella G, Morisco F. Safety and immunogenicity of anti-SARS-CoV-2 booster dose in patients with chronic liver disease. J Clin Med. 2023;12(6):2281. doi:10.3390/jcm12062281.
  • Ruether DF, Schaub GM, Duengelhoef PM, Haag F, Brehm TT, Fathi A, Wehmeyer M, Jahnke-Triankowski J, Mayer L, Hoffmann A, et al. SARS-CoV2-specific humoral and T-cell immune response after second vaccination in liver cirrhosis and transplant patients. Clin Gastroenterol Hepatol. 2022;20:162–72.e9. doi:10.1016/j.cgh.2021.09.003.
  • Iavarone M, Tosetti G, Facchetti F, Topa M, Er JM, Hang SK, Licari D, Lombardi A, D’Ambrosio R, Degasperi E, et al. Spike-specific humoral and cellular immune responses after COVID-19 mRNA vaccination in patients with cirrhosis: a prospective single center study. Dig Liver Dis. 2023;55(2):160–8. doi:10.1016/j.dld.2022.09.010.
  • Cheung KS, Mok CH, Mao X, Zhang R, Hung IF, Seto WK, Yuen MF. COVID-19 vaccine immunogenicity among chronic liver disease patients and liver transplant recipients: a meta-analysis. Clin Mol Hepatol. 2022;28:890–911. doi:10.3350/cmh.2022.0087.
  • Luo D, Chen X, Du J, Mei B, Wang A, Kuang F, Fang C, Gan Y, Peng F, Yang X, et al. Immunogenicity of COVID-19 vaccines in chronic liver disease patients and liver transplant recipients: a systematic review and meta-analysis. Liver Int. 2023;43(1):34–48. doi:10.1111/liv.15403.
  • Khoury DS, Cromer D, Reynaldi A, Schlub TE, Wheatley AK, Juno JA, Subbarao K, Kent SJ, Triccas JA, Davenport MP, et al. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med. 2021;27(7):1205–11. doi:10.1038/s41591-021-01377-8.
  • Marjot T, Webb GJ, Barritt AS, Gines P, Lohse AW, Moon AM, Pose, E, Trivedi, P, Barnes, E, et al. SARS-CoV-2 vaccination in patients with liver disease: responding to the next big question. Lancet Gastroenterol Hepatol. 2021;6:156–8. doi:10.1016/S2468-1253(21)00008-X.
  • Simao AL, Palma CS, Izquierdo-Sanchez L, Putignano A, Carvalho-Gomes A, Posch A, Zanaga P, Girleanu I, Henrique MM, Araújo C, 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. doi:10.1016/j.jhepr.2023.100697.
  • Ahmed SF, Quadeer AA, McKay MR. SARS-CoV-2 T cell responses elicited by COVID-19 vaccines or infection are expected to remain robust against Omicron. Viruses. 2022;14(1):14. doi:10.3390/v14010079.
  • Le Bert N, Tan AT, Kunasegaran K, Tham CYL, Hafezi M, Chia A, Chng MHY, Lin M, Tan N, Linster M, et al. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature. 2020;584(7821):457–62. doi:10.1038/s41586-020-2550-z.
  • Pearce FA, Lim SH, Bythell M, Lanyon P, Hogg R, Taylor A, Powter G, Cooke GS, Ward H, Chilcot J, et al. Antibody prevalence after three or more COVID-19 vaccine doses in individuals who are immunosuppressed in the UK: a cross-sectional study from MELODY. Lancet Rheumatol. 2023;5:461–73. doi:10.1016/S2665-9913(23)00160-1.
  • Romano C, Esposito S, Donnarumma G, Marrone A. Detection of neutralizing anti-severe acute respiratory syndrome coronavirus 2 antibodies in patients with common variable immunodeficiency after immunization with messenger RNA vaccines. Ann Allergy Asthma Immunol. 2021;127:499–501. doi:10.1016/j.anai.2021.07.026.
  • Zhu Q, Wang L, Hu X, Zhang Y, Huang T, He T, Chen Z, Zhang G, Peng M, Chen M, et al. Dynamic humoral immune response to primary and booster inactivated SARS-CoV-2 vaccination in patients with cirrhosis. J Clin Transl Hepatol. 2023;11(7):1476–84. doi:10.14218/JCTH.2023.00108.
  • Sripongpun P, Pinpathomrat N, Sophonmanee R, Ongarj J, Seepathomnarong P, Seeyankem B, Chamroonkul N, Piratvisuth T, Kaewdech A. Heterologous COVID-19 vaccination and booster with mRNA vaccine provide enhanced immune response in patients with cirrhosis: a prospective observational study. Vaccines (Basel). 2023;11:11. doi:10.3390/vaccines11091455.
  • John BV, Deng Y, Schwartz KB, Taddei TH, Kaplan DE, Martin P, Chao H-H, Dahman B. Postvaccination COVID-19 infection is associated with reduced mortality in patients with cirrhosis. Hepatology. 2022;76:126–38. doi:10.1002/hep.32337.
  • Diaz LA, Fuentes-Lopez E, Lazo M, Kamath PS, Arrese M, Arab JP. Vaccination against COVID-19 decreases hospitalizations in patients with cirrhosis: results from a nationwide analysis. Liver Int. 2022;42(4):942–4. doi:10.1111/liv.15193.
  • Stroffolini T, Ciancio A, Federico A, Benigno RG, Colloredo G, Lombardi A, Niro GA, Verucchi G, Ferrigno L, Gioli F, et al. COVID-19 vaccination among cirrhotics in Italy: high coverage and effectiveness of 3 doses versus 2 in preventing breakthrough infection and hospitalization. Dig Liver Dis. 2023;55(3):316–21. doi:10.1016/j.dld.2022.11.016.
  • John BV, Ferreira RD, Doshi A, Kaplan DE, Taddei TH, Spector SA, Paulus E, Deng Y, Bastaich D, Dahman B, et al. Third dose of COVID-19 mRNA vaccine appears to overcome vaccine hyporesponsiveness in patients with cirrhosis. J Hepatol. 2022;77(5):1349–58. doi:10.1016/j.jhep.2022.07.036.
  • Murray SM, Pose E, Wittner M, Londono MC, Schaub G, Cook J, Dimitriadis S, Meacham G, Irwin S, Lim Z, et al. Immune responses and clinical outcomes after COVID-19 vaccination in patients with liver disease and liver transplant recipients. J Hepatol. 2024;80(1):109–23. doi:10.1016/j.jhep.2023.10.009.
  • John BV, Bastaich DR, Ferreira RD, Doshi A, Taddei TH, Kaplan DE, Spector S, Deng Y, Dahman B. Covid-1 vaccine effectiveness and community prevalence of alpha, delta and omicron variants in patients with cirrhosis. Hepatology. 2022;76(9):573–4. doi:10.1136/gutjnl-2022-327799.
  • Gaborit B, Fernandes S, Loubet P, Ninove L, Dutour A, Cariou B, Coupaye M, Clement K, Czernichow S, Carette C, et al. Early humoral response to COVID-19 vaccination in patients living with obesity and diabetes in France. The COVPOP OBEDIAB study with results from the ANRS0001S COV-POPART cohort. Metabolism. 2023;142:155412. doi:10.1016/j.metabol.2023.155412.
  • Porntharukchareon T, Chartisathian W, Navinpipat M, Samdaengpan C, Cheirsilpa K, Lueprasitsakul A, Worawichawong S, Muangsillapasart V, Pumuthaivirat P, Dechates B, et al. The immunogenicity of the ChAdOx1 nCoV-19 vaccination in participants with underlying comorbidity diseases: a prospective cohort study. Hum Vaccin Immunother. 2023;19:2251850. doi:10.1080/21645515.2023.2251850.
  • Chen JJ, Lee TH, Tian YC, Lee CC, Fan PC, Chang CH. Immunogenicity rates after SARS-CoV-2 vaccination in people with end-stage kidney disease: a systematic review and meta-analysis. JAMA Netw Open. 2021;4:e2131749. doi:10.1001/jamanetworkopen.2021.31749.
  • Li H, Wang Y, Ao L, Ke M, Chen Z, Chen M, Peng M, Ling N, Hu P, Cai D, et al. Association between immunosuppressants and poor antibody responses to SARS-CoV-2 vaccines in patients with autoimmune liver diseases. Front Immunol. 2022;13:988004. doi:10.3389/fimmu.2022.988004.