Next Article in Journal
Population-Level Immunity for Transient Suppression of COVID-19 Waves in Japan from April 2021 to September 2022
Next Article in Special Issue
Pfizer-BioNTech mRNA Vaccine Protection among Children and Adolescents Aged 12–17 Years against COVID-19 Infection in Qatar
Previous Article in Journal
Heterologous COVID-19 Vaccination and Booster with mRNA Vaccine Provide Enhanced Immune Response in Patients with Cirrhosis: A Prospective Observational Study
Previous Article in Special Issue
Evaluation of Antibody Response and Adverse Effects following Heterologous COVID-19 Vaccine Booster with mRNA Vaccine among Healthcare Workers in Indonesia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Efficacy, Immunogenicity, and Safety of COVID-19 Vaccines in Patients with Autoimmune Diseases: A Systematic Review and Meta-Analysis

by
Alvina Widhani
1,2,*,
Anshari Saifuddin Hasibuan
1,
Retia Rismawati
1,
Suzy Maria
1,
Sukamto Koesnoe
1,
Muhammad Ikrar Hermanadi
1,
Youdiil Ophinni
3,4,5,
Chika Yamada
5,
Kuntjoro Harimurti
6,
Aldean Nadhyia Laela Sari
1,
Evy Yunihastuti
1 and
Samsuridjal Djauzi
1
1
Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia
2
Department of Internal Medicine, Universitas Indonesia Hospital, Depok 16424, Indonesia
3
Division of Clinical Virology, Center for Infectious Diseases, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
4
Department of Host Defense, Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
5
Center for Southeast Asian Studies, Kyoto University, Kyoto 606-8304, Japan
6
Geriatric Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia
*
Author to whom correspondence should be addressed.
Vaccines 2023, 11(9), 1456; https://doi.org/10.3390/vaccines11091456
Submission received: 15 July 2023 / Revised: 30 August 2023 / Accepted: 1 September 2023 / Published: 4 September 2023

Abstract

:
Patients with autoimmune diseases are among the susceptible groups to COVID-19 infection because of the complexity of their conditions and the side effects of the immunosuppressive drugs used to treat them. They might show impaired immunogenicity to COVID-19 vaccines and have a higher risk of developing COVID-19. Using a systematic review and meta-analysis, this research sought to summarize the evidence on COVID-19 vaccine efficacy, immunogenicity, and safety in patients with autoimmune diseases following predefined eligibility criteria. Research articles were obtained from an initial search up to 26 September 2022 from PubMed, Embase, EBSCOhost, ProQuest, MedRxiv, bioRxiv, SSRN, EuroPMC, and the Cochrane Center of Randomized Controlled Trials (CCRCT). Of 76 eligible studies obtained, 29, 54, and 38 studies were included in systematic reviews of efficacy, immunogenicity, and safety, respectively, and 6, 18, and 4 studies were included in meta-analyses for efficacy, immunogenicity, and safety, respectively. From the meta-analyses, patients with autoimmune diseases showed more frequent breakthrough COVID-19 infections and lower total antibody (TAb) titers, IgG seroconversion, and neutralizing antibodies after inactivated COVID-19 vaccination compared with healthy controls. They also had more local and systemic adverse events after the first dose of inactivated vaccination compared with healthy controls. After COVID-19 mRNA vaccination, patients with autoimmune diseases had lower TAb titers and IgG seroconversion compared with healthy controls.

1. Introduction

As of 26 December 2022, there were more than 651 million cases of COVID and more than 6 million deaths reported worldwide [1]. It is important to understand that certain groups in the population are higher-risk groups who are more susceptible to severe COVID-19 infection. These groups consist of people who have comorbidities, such as cancer, chronic kidney disease, underlying lung disorders, diabetes, dementia, cardiac issues, HIV, other immunocompromised conditions, neurological diseases, and pregnancy [2].
One of these susceptible groups is people with autoimmune diseases because of the complexity of these conditions and the mechanisms underlying the therapeutic effects of the drugs used to treat them. Medications play a pivotal role in significantly improving the disease course and outcomes of autoimmune patients. However, the primary disadvantage of these medications is the immunosuppressive effect they have, which can enhance the risk of infections. Therefore, there is an emerging demand to prioritize COVID-19 vaccination for people with autoimmune conditions, as this prevents severe disease outcomes [3,4].
Vaccination is an effort to suppress the case numbers and severity of COVID-19 infections [5,6]. It has been established that vaccines can induce humoral and/or cellular immune responses to build protection against various infectious diseases, which is an ability also known as immunogenicity [5,7]. Not only does COVID-19 vaccination protect healthy individuals from getting infected, it also prevents those who are infected from getting severely ill, or even dying, from COVID-19 [5,6,7]. As of 26 December 2022, 13 billion doses of COVID 19 vaccine had been administered worldwide [1].
An additional cause of concern is that patients with systemic autoimmune diseases might show impaired immunogenicity to COVID-19 vaccines. These patients can have a higher risk of developing COVID-19 [8]. Besides the issue of decreased vaccine efficacy due to the use of immunosuppressive drugs, the safety of the COVID-19 vaccine is also a concern among these patients [9,10]. Certain vaccine antigens and their adjuvants, such as aluminum salts (alum), have been claimed to induce autoimmunity in numerous studies. Adjuvants are usually needed in inactivated and recombinant protein vaccines to boost the immunogenicity induced by the antigen [4]. Patients with autoimmune diseases are more susceptible to vaccination-induced autoimmune/autoinflammatory syndrome induced by adjuvants (ASIA) [11]. SARS-CoV-2 amino acid sequences cross-react with human cell sequences [12]. The antibody to the S1 spike protein of SARS-CoV-2 has a high affinity for transglutaminase 3 protein, transglutaminase 2 protein, anti-extractable nuclear antigen, nuclear antigen, and myelin basic protein [13]. Despite the evidence, this claim should be interpreted cautiously, as the temporal relationship between the vaccine and autoimmune events is still unclear [4]. There is also evidence that non-live vaccines, including those for influenza and pneumococcal virus, do not cause exacerbation of previously diagnosed autoimmune conditions [3,6].
In the third-phase clinical trial of ChAdOx1 nCoV-19 (AstraZeneca), a simian adenovirus-vectored vaccine, there was one case of transverse myelitis reported 14 days after vaccination [14]. A cohort study from the health registry in Denmark and Norway showed an increase in venous thromboembolism cases, including cerebral venous thrombosis, 28 days after ChAdOx1 nCoV-19, and a slight increase in thrombocytopenia and bleeding cases [15]. Another study reported 39 patients with thrombocytopenia and thrombosis 5–24 days after vaccination with ChAdOx1 nCoV-19. These patients were diagnosed with vaccine-induced thrombotic thrombocytopenia (VITT) or thrombosis with thrombocytopenia syndrome (TTS), which were suspected to be caused by platelets activating antibodies to platelet factor 4 [16,17,18]. Brill et al. reported autoimmune hepatitis 6 days after administration of the Pfizer–BioNTech COVID-19 vaccine in a 35-year-old woman. This case report could not conclude whether this was a causal relationship or only coincidence [19]. There were also reports of thrombocytopenia post the mRNA vaccine, which were diagnosed as secondary immune thrombocytopenia (ITP), but again, it could not be determined whether this was a coincidence or vaccine-induced ITP [20].
Despite COVID-19 vaccination being recommended, the efficacy, immunogenicity, and safety of COVID-19 vaccination in people with autoimmune diseases have not been discussed much. In addition, patients with autoimmune conditions and/or people taking immunosuppressants were excluded from clinical trials of approved COVID-19 vaccines [4,21]. Therefore, this systematic review aims to summarize the evidence on COVID-19 vaccine efficacy, immunogenicity, and safety in autoimmune patients.

2. Materials and Methods

The protocol for this study has been registered in PROSPERO with the registration number CRD42022337621. This study was conducted in accordance with the Preferred Reporting Items of the Systematic Review and Meta-Analysis (PRISMA) checklist [22].

2.1. Eligibility Criteria

The specific inclusion criteria for the systematic review and meta-analysis were as follows: (1) all randomized controlled trials (RCTs), non-randomized studies of interventions, cohort studies, case–control studies, and cross-sectional studies; (2) studies with autoimmune patients as the population (with the autoimmune condition existing prior to the intervention); (3) COVID-19 vaccination as the intervention; (4) efficacy, immunogenicity or safety as outcomes; and (5) publication in English. The exclusion criteria were as follows: (1) full text or data that cannot be accessed even though the corresponding author has been contacted.

2.2. Information Sources and Search Strategy

We included all articles on patients with autoimmune diseases published in English from 2020 to 2022. Electronic databases were searched using PubMed, Embase, EBSCOhost, ProQuest, MedRxiv, bioRxiv, SSRN, EuroPMC, and the Cochrane Center of Randomized Controlled Trials (CCRCT) from 6–26 September 2022 for studies evaluating the response to SARS-CoV-2 vaccines using a combination of keywords and medical subject headings. The keywords utilized were “autoimmune”; “vaccine” or “immunization” or “vaccination”; “COVID-19”; “efficacy”; “immunogenicity”; and “safety” or “adverse event” or “adverse effect”, along with their synonyms and related terms incorporated by the appropriate Boolean operators. The detailed search strategy for articles is available in the Supplementary Materials (Table S1).

2.3. Data Extraction

Records were checked for duplicates using Zotero 6.0.19. Two independent researchers screened the literature search and assessed each study for inclusion by reading titles, abstracts, and full texts. Different opinions during data extraction were resolved by discussion and the inclusion of a study was decided by the two researchers. Relevant data were obtained from each eligible study by using an extraction sheet, which was prepared and approved by all the reviewers by reaching a consensus after screening for the eligible studies. Relevant data that were collected included study characteristics (authors, year, country, research setting, study design, study duration, sample size); participant characteristics (autoimmune diagnosis, age, sex, comorbidities); intervention (COVID-19 vaccine platform) and comparison; and outcomes (efficacy, immunogenicity, safety). Two independent researchers collected the data from each research article. The corresponding authors were contacted to obtain any information that was not explicitly available.

2.4. Outcome Measures

All studies describing the efficacy, immunogenicity, or safety of the COVID-19 vaccine in autoimmune patients were evaluated. The main outcomes were (1) breakthrough COVID-19 events, severity of infection, hospitalization, and mortality as markers of efficacy; (2) neutralizing antibodies, antibody titers, and seroconversion as markers of immunogenicity; and (3) flares or autoimmune relapses, local reactions, systemic reactions, and other adverse events as markers of safety.
The pooled efficacy, immunogenicity, and safety data after primary or booster doses of COVID-19 vaccine were evaluated. Efficacy was measured by the number of COVID-19 breakthrough infections, severity of COVID-19 infections, and hospitalizations and mortality related to COVID-19 infection. A COVID-19 breakthrough infection was defined as an infection after receiving the vaccination. Severity was defined by one of three levels of COVID-19 infection after vaccination: mild, moderate, or severe. Hospitalization was defined as the number of people who were taken to hospital as a result of COVID-19 infection. Mortality was defined as the number of people who died as a result of COVID-19 infection. Immunogenicity was defined as the ability of COVID-19 vaccines to stimulate an immune response, which was measured by the proportions of subjects with seroconversion (based on total IgG, as measured by ELISA) and with neutralizing antibodies (based on a plaque reduction neutralization test (PRNT) or surrogate virus neutralization test (sVNT), total IgG antibody titers (following WHO guidelines on translating results from different ELISA manufacturers into standardized binding antibody units (BAU)/mL) [23], and neutralizing activity (based on PRNT or sVNT, calculated as (1-OD value of sample/OD value of control) × 100%). Antibody titers were log-transformed prior to standardized mean difference (SMD) calculation. Where applicable, PRNT50 titer was correlated with sVNT inhibition capacity [24], mean and standard deviation (SD) were estimated from median and interquartile range (IQR) [25], SDs were estimated from 95% confidence interval, and means and SDs were aggregated from multiple subgroups. Safety was measured by the number of autoimmune relapses, local symptoms (pain, erythema, bruising, etc.), systemic symptoms (fever, joint pain, flu like symptoms, fatigue, headache, muscle pain), and other adverse events occurring after receipt of a dose of COVID-19 vaccine.

2.5. Risk of Bias Evaluation

Risk-of-bias and quality-of-study evaluations were carried out by two independent researchers. The Risk of Bias (RoB) and Risk of Bias in Nonrandomized Studies of Interventions (ROBINS-I) tools were used for randomized controlled trials (RCT) and non-randomized studies of interventions, respectively [26]. Cross-sectional and case-series studies were assessed using the Newcastle–Ottawa Quality Assessment Scales and The National Institutes of Health (NIH) quality assessment tool, respectively [27,28]. The certainty of evidence for the primary outcomes was evaluated using the Grades of Recommendation, Assessment, Development, and Evaluation (GRADE) system in eight domains: risk of bias, inconsistency, indirectness, imprecision, publication bias, large effects, plausible confounding, and dose–response gradient [26].

2.6. Data Synthesis

All outcomes were analyzed using Microsoft Excel and RevMan version 5.4 issued by Cochrane. Outcomes were reported as risk ratios for categorical data and standardized mean differences for numerical data, each with a confidence interval. Risk ratio was used to compare the risks of outcomes measured among patients with autoimmune diseases to healthy controls, while standardized mean difference was used to assess and pool continuous data, which was measured in a variety of ways. For analyzing continuous data conversion, guidelines from the Cochrane book were applied [28]. Heterogeneity was assessed using Higgins I2 and considered significant at I2 > 60% [28]. For significantly heterogeneous data, subgroup analysis was performed. Fixed-effects models were used for data with no substantial heterogeneity or which was considered homogeneous, whereas random effect models were used when there was significant heterogeneity. Data was displayed as a forest plot for meta-analysis.

3. Results

Our search retrieved 1054 records, of which 833 were duplicates and were excluded. The titles and abstracts of the remaining 221 published articles were screened, and 188 were assessed for eligibility via full-text evaluation. One hundred and twelve records did not meet the inclusion criteria after this full-text review, and were excluded. As a result, 76 full-text articles were selected for systematic review. Subsequently, 20 full-text articles were selected for meta-analysis, with 6, 18, and 4 articles included for efficacy, immunogenicity, and safety, respectively. The study flow chart is presented in Figure 1.

3.1. Study Characteristics

Seventy-six studies were included in the qualitative analysis (Table S2). Ten studies were conducted in Israel [29,30,31,32,33,34,35,36,37,38], one study in Denmark [39], eight studies in Italy [8,40,41,42,43,44,45,46], three studies in the USA [47,48,49], nine studies in Germany [50,51,52,53,54,55,56,57,58], one study in New Zealand [59], three studies in Austria [60,61,62], four studies in Spain [63,64,65,66], one study in Japan [67], one study in France [68], one study in Romania [69], one study in Peru [70], one study in Canada [71], six studies in Brazil [72,73,74,75,76,77], two studies in China [78,79], three studies in Thailand [80,81,82], one study in Chile [83], five studies in India [84,85,86,87,88], one study in Greece [89], one study in Turkey [90], four studies in the UK [91,92,93,94], one study in Korea [95], one study in Taiwan [96], two studies in Netherlands [97,98], one study in Switzerland [85], one study in Hungary [99], and one study in the USA and UK [100]. The types of investigated studies encompassed single-blinded [73,96], observer-blinded randomized [31], and non-randomized [8,29,30,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,97,98,99,100,101,102,103] studies. A total of 160,447 participants were involved. All studies concerned adult participants (the majority of participants were >18 years of age), and only one study also involved pediatric participants [91]. Sixty-six studies included participants who had only had a primary dose vaccine [8,29,30,35,36,37,38,39,41,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,82,83,84,85,86,87,88,89,90,91,92,93,97,98,99,100,101,102,103], whereas in ten studies participants had had a booster dose vaccine [31,32,33,34,40,42,81,94,95,96].
The studies in our qualitative analysis were divided into six categories based on the type of vaccine: studies on mRNA vaccines including Pfizer/BioNTech (BNT162b2) and Moderna (mRNA-1273); studies on inactivated virus vaccines including CoronaVac, Covaxin (BBV152), and Sinopharm (BBIBP-CorV); studies on adenovirus vector vaccines including Vaxzevria (ChAdOx1), Janssen (Ad26.COV2.S), Sputnik V (Gam-COVID-Vac), and AstraZeneca (AZD1222); studies on mRNA vaccines and adenovirus vector vaccines; studies on inactivated virus and adenovirus vector vaccines; and studies on mRNA vaccines, inactivated virus vaccines, and adenovirus vector vaccines.
In terms of autoimmune diagnosis, studies included adult-onset Still’s disease [57,61,89,95], antiphospholipid syndrome [33,47,69,72,74,75,88,89,94,96,102], autoimmune encephalitis [40,52,60], autoimmune hepatitis [44,56,69,90,97,101], autoimmune thyroid [69,96], IgG-4-related diseases [47,69,92,94], interstitial lung disease and systemic autoimmune disease/immune pulmonary disease [49,69], inflammatory bowel disease [47,50,62,69,71,91,97], inflammatory myopathies/systemic autoimmune myopathy [33,35,36,61,63,69,72,74,75,76,77,101], immune-mediated thrombocytopenic purpura/immune-mediated thrombotic thrombocytopenic purpura (ITP/iTTP) [32,42,96], juvenile idiopathic arthritis [57,83,88,89,94,98], mixed/undifferentiated connective tissue disease/connective tissue disease [44,47,50,55,57,61,69,86,87,88,89,94,98,103], multiple sclerosis [29,30,31,34,47,50,52,53,54,60,64,67,69,97,98], myasthenia gravis syndrome [52,60,69,97], neuromyelitis optica spectrum disorder [47,52,60,66,97], primary biliary cholangitis [56,69,97,101], psoriasis [50,69,71,80,97], psoriatic arthritis [33,35,50,57,69,71,72,74,75,83,88,94,98,99], rheumatoid arthritis (RA) [33,35,36,39,43,44,47,50,57,58,63,65,72,73,74,75,78,82,83,84,86,87,88,89,93,94,95,96,97,98,99,101,103], systemic lupus erythematosus (SLE) [33,35,36,39,44,45,47,50,57,61,65,66,68,69,70,72,74,75,77,78,81,82,84,86,87,88,89,92,93,94,95,96,97,98,99,101,102,103], sarcoidosis [50,61,88,94,98], spondiloarthritis/spondyloarthropathy [33,35,44,47,49,50,57,69,71,72,74,75,84,86,87,88,89,94,95,96,97,98,99,101,103], sclerosing cholangitis [56,97], Sjogren syndrome/sicca syndrome [33,47,61,65,69,71,74,75,78,88,89,94,96,98,99], systemic sclerosis [33,44,61,63,65,69,72,74,75,86,87,88,89,94,98,99,101,103], and vasculitides/vasculitis [33,35,36,44,47,50,55,57,58,61,66,69,72,74,75,86,87,88,89,92,93,94,95,97,98,99,103].
Autoimmune medications given to the patients included alemtuzumab [29,34,48,66], abatacept [8,33,35,39,47,57,58,63,72,73,74,75,94,98], anti-CD20/-B cell depleting therapy [8,29,32,33,34,35,37,39,40,41,43,45,47,48,49,52,53,54,55,57,60,62,63,64,65,66,72,74,75,87,89,92,94,96,97,98,103], antimalarials including hydroxychloroquine (HCQ) and chloroquine [8,37,39,41,44,45,47,50,55,61,63,65,68,69,70,72,75,77,81,82,83,84,87,88,89,90,93,94,95,96,98,99,103], apremilast [94,103], azathioprine [8,33,39,40,41,44,45,47,50,52,56,57,58,60,63,65,68,69,70,71,72,74,75,76,80,81,82,84,85,87,88,89,90,92,94,95,98,99,101,103], belimumab [8,39,41,44,45,47,50,57,61,65,68,72,74,75,77,89,92,94,95,99], calcineurin inhibitor [33,41,77,95], caplacizumab [42], certolizumab [50,83,101], cladibrine [29,30,34,48,52,66], colchicine [33,89,94,103], corticosteroids [32,33,35,36,37,39,40,41,42,44,45,47,49,50,52,55,56,57,58,60,62,63,64,68,69,70,72,73,75,76,77,80,81,82,83,84,86,87,88,89,90,91,92,93,94,96,98,99,100,101,103], cyclophosphamide (CYP) [45,62,63,64,72,74,75,76,77,87,89,92,95], cyclosporine (CYC) [45,72,74,75,76,80,81,82,83,85,88,89,94,98], denosumab [94], DMF [29,34,48,52,53,66], eculizumab [94], everolimus [85], fampridine [98], fingolimod [29,30,31,34,47,52,53,64,98], glatiramer acetate (GA) [29,34,48,52,53,64,98], ibrutinib [47,100], iguratimod [88], IL-1 inhibitor [89,94], IL-6 inhibitor [8,33,35,37,39,40,44,45,47,50,57,60,61,63,65,72,74,75,83,85,89,94,95,99,101], IL-17 inhibitor [33,35,44,51,57,71,72,74,75,80,83,89,94,98,99,101], IL-12/23 inhibitor [33,47,71,89,99], IL-23 inhibitor [47,71,99], β interferons [29,34,48,52,54,66,98], intravenous immunoglobulin (IVIG) [29,32,34,36,52,71,100], Janus kinase (JAK) inhibitor [8,33,35,39,57,58,62,88,89,94,95,99,101], leflunomide [8,36,39,45,47,50,58,63,69,70,72,73,74,75,76,77,82,83,84,87,88,89,93,94,95,98,99,103], lenalidomide [87,99], mepolizumab [99], methotrexate [8,33,35,36,39,41,43,44,47,57,58,61,63,65,68,69,70,71,72,73,74,75,76,77,80,82,83,84,87,88,89,90,92,93,94,95,96,97,98,99,101,103], mycophenolate mofetil [8,33,35,36,37,39,40,41,43,44,45,47,49,56,57,60,61,63,65,68,69,70,72,74,75,76,77,80,81,82,83,84,85,87,88,89,90,92,94,95,97,100,101,103], natalizumab [29,34,48,52,53,66,98], nintedanib [49], ocrelizumab (OCR) [29,30,34,48,52,60], ofatumumab [48], olumiant [61], omalizumab [80], pembrolizumab [99], plasmapheresis (PLEX) [42,52,64], sphingosine-1-phosphate receptor modulators (S1PRM) [48,66,97], salazopyrin [39], sulfasalazine [8,44,47,51,58,63,69,72,75,83,84,87,93,94,95,96,97,98,98,103], tacrolimus [45,61,72,74,75,81,82,83,85,87,92,94,103], teriflunomide [29,34,48,52,53,66], thalidomide [89,94], tumor necrosis factor alpha inhibitor (TNFi) [8,33,35,39,44,45,47,50,51,57,58,62,63,65,71,72,74,83,87,89,91,94,95,97,98,99,101,103], tofacitinib [37,47,72,74,75,87,103], upadacitinib [37,47], ustekinumab [50,72,74,75,98], and vedolizumab [47,50,91,92].

3.2. Quality of Assessment

Graphical representation of the studies’ quality is illustrated in the Supplementary Materials (Figure S1A–D). Risks of bias in the three RCTs were low; twenty-one non-randomized studies were low-risk, thirty-seven non-randomized studies were moderate-risk, and seven non-randomized studies had serious risk; four case-series studies were defined as good; and four cross-sectional studies were considered fair.

3.3. Qualitative Analysis

3.3.1. Efficacy

In the mRNA vaccine studies group, efficacy after primary vaccination was reported as breakthrough COVID-19 infections [29,35,41,49,63,101], hospitalizations [49,63], and deaths [29,35,49,63]. Efficacy after booster vaccination was also reported as breakthrough COVID-19 infections [33,34] and hospitalizations and deaths [33]. In the inactivated virus vaccine studies, six studies reported breakthrough COVID-19 infections after primary vaccination as outcomes [72,75,76,77,80,83], two studies reported hospitalizations [76,77], and only one study reported death [77]. One study on adenovirus vector vaccines reported breakthrough infections [87]. In the mRNA vaccine and adenovirus vector vaccine studies, efficacy after primary vaccination was reported as breakthrough COVID-19 infections [45,69,97,98], hospitalizations [95,97], and deaths [100]. Efficacy after booster vaccination was reported as breakthrough COVID-19 infections, hospitalizations, deaths [89], and hospitalizations or deaths due to breakthrough infections [90]. In the inactivated virus vaccine and adenovirus vaccine studies, one study reported breakthrough COVID-19 infections after primary vaccination [103]. In the mRNA vaccine, inactivated virus vaccine, and adenovirus vector vaccine studies, efficacy was reported as breakthrough COVID-19 infections and hospitalizations after primary vaccination [88,94]. Breakthrough COVID-19 infections and deaths after booster vaccination were reported in only one study [94].
mRNA vaccination, either primary or booster, has been found to have a protective effect on breakthrough infections where the risk of getting infections after vaccination is lower compared with the unvaccinated group [33]. According to Bieber et al., patients with autoimmune rheumatic disease who received a third booster of mRNA vaccination had lower SARS-CoV-2 infection rates [33]. However, Kim et al. observed both patients and healthy controls to have SARS-CoV-2 omicron breakthrough infections after a third dose of vaccination [95]. Mena-Vázquez et al. also reported that patients who were not infected with SARS-CoV-2 received vaccinations more frequently. Moreover, COVID-19-infected patients took rituximab and glucocorticoids more frequently [63].
Symptomatic breakthrough COVID-19 infections among patients and in a healthy control group were reported in two studies after the participants had had a primary inactivated COVID-19 vaccination [76,77], although only one patient required hospitalization and no patients died [77]. Non-severe infections were reported after a mean period of fourteen weeks from full vaccination, where half of the infected participants were patients with negative total anti-SARS-CoV-2 IgG antibodies and neutralizing antibodies [83].
Studies in which autoimmune patients received an mRNA or adenovirus vector vaccine reported a higher hospitalization rate in the unvaccinated group compared with the vaccinated group, as well as a higher rate of severe COVID-19 cases, which appeared less frequently in third-dose-vaccinated patients than in second-dose-vaccinated patients and an unvaccinated group [89]. Breakthrough infections were also more frequent in patients on strongly impairing immunosuppressants, including anti-CD20 combination therapy, sphingosine 1-phosphate modulators, and mycophenolate mofetil therapy, as opposed to patients on other immunosuppressants [97].
According to the results from a study on inactivated and adenovirus vaccines, the strongest predictor of breakthrough infections is the absence of an antibody response. Vaccine platform and mycophenolate mofetil were found to be the other breakthrough infection predictors [103]. Patients with autoimmune disease receiving Covaxin showed higher rates of breakthrough infection than those receiving the AstraZeneca vaccine [103]. Another result from a study on adenovirus vector vaccines reported that there was no significant difference in the frequency of breakthrough infections between patients who received a second dose of vaccine after 4–6 weeks versus 10–14 weeks [87]. Furthermore, results from a study reporting on autoimmune patients given mRNA, inactivated virus, or adenovirus vector vaccines showed no breakthrough infections in patients vaccinated with mRNA. Meanwhile, inactivated-virus-vaccinated patients had a higher percentage of breakthrough infections after full vaccination than adenovirus-vector-vaccinated patients, although the difference was not significant [88].

3.3.2. Immunogenicity

There were 54 studies reporting immunogenicity: 27 studies on mRNA vaccines (13 studies on Pfizer/BioNTech [30,31,34,36,38,39,43,46,54,59,61,67,68], 1 study on Moderna [101], and 13 studies on Pfizer/BioNTech or Moderna [8,35,41,44,47,50,51,52,53,60,62,65,71]); 9 studies on inactivated virus vaccines using CoronaVac [72,73,74,75,76,77,79,80,83]; 2 studies on adenovirus vector vaccines [84,87]; 12 studies on mRNA and adenovirus vector vaccines (5 studies on Pfizer/BioNTech, Moderna, Vaxzevria, or Janssen [55,57,66,95,98], 2 studies on Pfizer/BioNTech, Moderna, or Vaxzevria [56,58], 3 studies on Pfizer/BioNTech or Vaxzevria [91,92,93], 1 study on Moderna or Vaxzevria [96], and 1 study on Pfizer/BioNTech (BNT162b2), Moderna (mRNA-1273), or Janssen (Ad26.COV2.S)) [85]; 1 study on inactivated virus vaccines and adenovirus vector vaccines using Covaxin or AstraZeneca [103]; 3 studies on mRNA vaccines, inactivated vaccines, and adenovirus vector vaccines using Pfizer, Coronavac, or Vaxzevria [81], Pfizer, Coronavac, Sinopharm or Vaxzevria [82], and Pfizer, Moderna, Sinopharm, Sputnik, and AstraZeneca [99]. Immunogenicity was determined by measuring antibody titers [8,30,31,34,35,36,38,39,41,43,46,47,50,51,52,53,54,56,57,58,59,60,61,62,65,66,67,68,75,76,80,81,82,83,84,85,87,92,93,96,98,99,101,103], seroconversion [8,30,31,35,36,39,41,43,44,46,47,52,53,54,55,56,57,60,61,65,66,71,72,73,74,75,76,77,80,82,84,92,95,97,98,99,101], neutralization antibodies [38,41,47,50,51,53,54,58,62,68,72,73,74,75,76,77,80,81,83,84,95,99,101,103], T-cell response [41,43,46,53,54,55,56,57,60,62,65,66,68,71,81,82,83,91,95,99,101], lymphocyte count [31,93], IgA titer [38,50,58,85,93], IgG avidity [51,54], B-cell counts [43,49,53,56,57,58,93], T-cell counts [55,58,62,93], and IgM titer [93].
Patients with autoimmune diseases who received CoronaVac had neutralizing antibodies and neutralizing activity lower than in the control group [72,74,75] as well as lower seroconversion [74,75]. Factors associated with poor immunogenicity were older age, obesity, and use of prednisone, biologics, and immunosuppressants [74,75]. Another study on patients given CoronaVac also found that mycophenolate and prednisone were related to reduced seroconversion, whereas hydroxychloroquine caused seroconversion to rise [77]. In another study on Pfizer, CoronaVac, Sinopharm, and Vaxzevria vaccination, anti-RBD titers were lower in the inactivated vaccine group, followed by Vaxzevria, then Vaxzevria or Pfizer [82]. The inactivated vaccine was also associated with the lowest humoral response, whereas the adenovirus-vectored/mRNA vaccine was associated with the highest humoral response [82].
Patients with multiple sclerosis who received the Pfizer vaccine while being treated with anti-CD20 therapy [54], fingolimod continuation [31], and other immunosuppressants [34] had lower IgG titers compared with untreated patients or patients who discontinued the therapy. In comparison with healthy controls, patients with autoimmune neurological disorder who had received the Pfizer or Moderna vaccines had decreased seroconversion rates [34] and anti-S1 IgG [53,60] and anti-S(RBD) specific IgG levels [52]. In comparison with healthy controls or patients not receiving immunotherapy, patients receiving anti-CD20 [52,53,60], fingolimod [52,53], azathioprine [52], and steroid therapy [52] exhibited lower levels of anti-S1 IgG and anti-S(RBD) specific IgG. Lower seroconversion rates were observed in multiple sclerosis patients receiving anti-CD20 or sphingosine 1-phosphate receptor modulators who were given the Pfizer, Moderna, or AstraZeneca vaccines compared with other disease-modifying therapies or untreated patients [66].
Additional research on the Pfizer or Vaxzevria vaccine indicated that seroconversion and anti-S IgG levels after the second dose were significantly lower in patients with autoimmune disease than in the control group and that this was associated with B-cell depletion at the time of vaccination [92]. Rituximab was significantly associated with no antibody vaccine response after adjusting for diagnosis and hydroxychloroquine, according to research in patients with SLE and RA who received the Pfizer vaccine [39]. A study of patients with SLE who had been given the Pfizer vaccination found that mycophenolate and methotrexate treatment were associated with a drastically diminished BNT162b2 antibody response [68]. Another study on the Moderna and Vaxzevria vaccines showed that individuals given hydroxychloroquine, low-dose steroid, methotrexate, and/or sulfasalazine therapy had significantly lower anti-SARS-CoV-2 spike IgG titers than those who were not on these therapies [96].
Studies of patients with autoimmune or autoinflammatory diseases, including RA, SLE, Sjogren syndrome, Behcet’s disease, polymyalgia rheumatica, connective tissue disease, vasculitis, adult-onset Still’s disease, and sarcoidosis who received the Pfizer vaccination showed lower seroconversion [46,61], anti S1/S2 IgG [36,38,43,61], neutralization [38,43], total IgA [38,43], and anti-RBD IgG [61] than the control group. The lowest antibody titers were detected in patients with antineutrophil cytoplasmic-antibody-associated vasculitis (AAV) and idiopathic inflammatory myopathy/myositis (IIM), while the highest titers were detected in SLE and RA patients [37]. Another study showed that antibody titers were also reduced with two or more immunosuppressants in combination therapy [61]. Studies in patients with systemic autoimmune disease, RA, SLE, inflammatory bowel disease, Sjogren syndrome, autoimmune hepatitis, psoriatic arthritis, IIM, sarcoidosis, and vasculitis who received the Pfizer and Moderna vaccines found that their anti-S IgG titers were lower than those of the control group, and these differences were particularly significant [8,35,50,51] in those who were receiving B-cell-depleting therapies, prednisone, JAK inhibitors, antimetabolites [47], TNFi [51], mycophenolate, and calcineurin inhibitors [44]. Moreover, compared with the control group, anti-RBD titers were lower in patients [41]. The differences remained significant in individuals receiving treatment with rituximab and belimumab [41]. According to another study, Ab levels and neutralization efficacy against variants of concern in anti-TNF-treated patients were substantially lower than in healthy controls, and by three months following the second dose of the vaccination they were undetectable against Omicron [71].
Seroconversion was considerably higher among Pfizer vaccine recipients when doses were given less than a month apart compared with AstraZeneca recipients, and tendencies towards higher antibody levels in vaccine responders were seen when either vaccine was given using short-interval dosing [93]. A study by Mehta et al. on the AstraZeneca vaccine showed that diabetes mellitus and vaccine interval were significantly associated with anti-RBD antibody titer [87]. A delayed (10–14 weeks) second dose of AstraZeneca vaccine was associated with a higher antibody titer [87]. A study by Ahmed et al. on AstraZeneca and Covaxin revealed that Covaxin and methotrexate treatment were associated with lower antibody titers [103]. Another study that focused on Vaxzevria vaccination in patients with autoimmune inflammatory rheumatic diseases revealed that single-dose-vaccinated patients who had had prior COVID-19 infections showed significantly higher seroconversion and neutralization activity than those who had received a double-dose vaccine [84].
In a study that focused on CoronaVac vaccination, neutralizing antibodies in RA patients on methotrexate therapy were lower than in the control group [73,83], as was the seroconversion rate [73]. Prednisone and mycophenolate usage were both highly linked to a negative NAb [83].
A study on mRNA and inactivated virus vaccines reported that IFN-γ and anti-RBD Abs levels have a slight but significant positive correlation [43]. Another study on mRNA, inactivated virus, and adenovirus vector vaccines reported that neutralizing anti-RBD-specific antibodies and the percentage of positive anti-RBD antibody responses were higher in participants vaccinated with mRNA vaccine compared with inactivated virus and adenovirus vaccines [99]. Additionally, patients who received the adenovirus vector or mRNA vaccines had a higher proportion of TNF-a-producing CD4+ T-cells upon SARS-CoV-2 antigen exposure compared with those who received the inactivated virus vaccine [99].
A third booster dose of mRNA or adenovirus vector vaccine after a primary inactivated vaccine produced a significant humoral and cellular immune response in SLE patients with inactive disease maintaining immunosuppressive treatment [81]. However, another study found that, after booster vaccination, neutralization responses against the Omicron variant were significantly lower in patients than in the healthy control group [92]. Certain medications, such as TNFi, aCD20-BCD- and fingolimod, antimetabolites, and calcineurin inhibitors were able to impair humoral and cellular responses, especially in autoimmune patients [51,53,81]. For instance, Achiron et al. found that a fingolimod continuation group had lower IgG titers than a fingolimod discontinuation group even at 3 months after the third vaccine dose [31]. In addition, anti-BA.2 neutralizing antibodies were not detectable in TNFi-treated patients [51]. Meyer et al. found that patients taking fingolimod failed to develop either humoral or CD4+ T cellular immune responses [53]. In contrast, Meyer et al. also reported that untreated patients showed an increase in anti-S1 IgG, neutralizing capacity, RBD- and S2-specific B cells, and spike-specific T cells after their first booster [53]. Lastly, however, a booster dose, particularly from an mRNA or viral vector vaccine, enhanced strong cellular immune responses, though responses were weaker in patients taking antimetabolites or calcineurin inhibitors [81].

3.3.3. Safety

Following primary vaccination in mRNA vaccine studies, autoimmune relapse was reported as a safety outcome in 12 studies [29,40,60,63,68,70,80,81,82,86,88,99]; local symptoms in 11 studies [8,29,35,36,37,40,50,52,60,67,68]; systemic symptoms in 13 studies [8,34,35,36,37,40,50,52,60,63,67,68,101]; and other symptoms in 12 studies [29,35,36,40,50,52,60,64,88,100,101,102]. Following booster vaccination in mRNA vaccine studies, autoimmune relapse was reported as a safety outcome in three studies [32,34,42] and local and systemic symptoms were also reported in one study [34]. Among the inactivated vaccine studies, following primary vaccination, autoimmune relapse was reported as a safety outcome in two studies [79,80] and local symptoms and systemic symptoms in six studies [72,73,75,76,77,78].
Among the mRNA vaccine and adenovirus vector vaccine studies, following primary vaccination, two studies reported autoimmune relapse after vaccination [20,69]. Local symptoms were reported as a safety outcome in three studies [45,69,72]. Systemic symptoms after primary mRNA and adenovirus vector vaccinations were reported in three studies [45,48,102]. Other symptoms were described in one study [82]. Meanwhile, among the inactivated virus vaccine and adenovirus vector vaccine studies, only one study reported autoimmune relapses and local and systemic symptoms as safety outcomes [86]. Among the mRNA vaccine, inactivated virus vaccine, and adenovirus vector vaccine studies, following primary vaccination, autoimmune relapse was reported as a safety outcome in two studies [88,94]; local symptoms in three studies [88,94]; systemic symptoms in four studies [82,88,94,99]; and other adverse events in two studies [88,94].
Patients who had been vaccinated with an mRNA vaccine reported no difference in relapse incidence before and after vaccination [8,52,68]. De Santis et al. and Ferri et al. also reported that, in the majority of cases, vaccine-related adverse effects were mild, and incidence rates were comparable in autoimmune patients and healthy controls with no differences based on current medications [8,101]. Mild cases, such as headache, occurred more frequently in SLE and cryoglobulinemic vasculitis patients, while pain at the injection site did in systemic vasculitis patients [8]. Moyon et al. found no related serious adverse events caused by vaccination [68]. Most of the relapse cases had significantly higher disease activity scores when compared with patients without post-vaccination relapses [40]. Additionally, De Santis et al. did not find any differences between patients with and without serum responses or in the prevalence of vaccine-related side effects [101]. In terms of booster vaccinations, a study reported more than 10% ITP exacerbations among ITP patients after booster vaccinations [32].
Patients who had been vaccinated with an inactivated virus vaccine were reported to have no moderate or severe adverse events [76,77]. Medeiros-Ribeiro et al. reported that overall reactions, such as arthralgia, back pain, malaise, nausea, and sweating, were more frequently and significantly found to occur in patients with autoimmune rheumatic disease than in a control group [75]. In patients with RA, myalgia and vertigo were significantly more frequent in those patients who were stopping methotrexate therapy at the time of receiving their second vaccination [73]. Headaches had a higher prevalence in patients with systemic autoimmune myopathies compared with healthy controls after a first dose of inactivated vaccine [76]. Autoimmune flare was also detected more frequently in a methotrexate-stopping RA patient group in comparison with a methotrexate-maintaining group at day 69 after vaccine administration [73].
Studies on mRNA and adenovirus vector vaccines reported that there was no difference in self-reported side effects between patients with neuroinflammatory diseases and a control group, whether after first vaccine dose or second vaccine dose, even after adjusting for age, BMI, and comorbidities [48]. Epstein et al. also reported that younger age was associated with an increased rate of reported side effects, whereas patients on high-efficacy therapy were associated with a lower risk of reported side effects [48]. The high-efficacy therapies referred to were therapies using ocrelizumab, rituximab, ofatumumab, alemtuzumab, cladribine, fingolimod, ozanimod, siponimod, and natalizumab [48]. Headaches were more common in patients with neuroinflammatory disease after mRNA vaccination than adenovirus vector vaccination, although no significant differences were observed [48]. Additionally, patients on high-efficacy therapy had a significantly lower rate of reported side effects compared with patients not on medication at the time of vaccination [48]. In terms of flare, there were no differences observed regarding age, comorbidities, number of autoimmune diseases associated, and years from disease diagnosis to the year prior to vaccination [69]. There were no significant differences in flare-up development among Cominarty, Vaxzevria, and Spikevax [69].
Additionally, studies on mRNA, inactivated virus, and adenovirus vector vaccines reported significantly more injection site pain in patients receiving AstraZeneca or Pfizer vaccination than in those who received inactivated vaccination, followed by fatigue and fever [82]. Another study on a third booster dose with an mRNA or viral vector vaccine following inactivated virus vaccination in SLE patients revealed more reactogenicity after the booster dose than the initial CoronaVac vaccination, but this was mild and no SLE flare was reported [81].

3.4. Meta-Analysis

For meta-analysis, we included 20 studies that compared the efficacy, immunogenicity, and safety of COVID-19 vaccines between patients with autoimmune diseases and healthy controls. There were six studies for efficacy [72,75,76,77,97,98], 18 studies for immunogenicity [8,35,36,43,50,52,54,61,62,65,72,74,75,76,77,80,83,101], and four studies for safety that could be included [72,75,76,77]. These studies were on inactivated vaccine, mRNA vaccine, and mRNA/adenovirus vector vaccine. All studies were non-randomized studies and on primary doses (two doses) of COVID-19 vaccine. Meta-analysis could not be done from the RCTs because there were only three RCTs [31,73,96] in our systematic review and only one RCT comparing the efficacy, immunogenicity or safety of the COVID-19 vaccine (primary dose) among patients with autoimmune disease (multiple sclerosis) and healthy controls [31].

3.4.1. Efficacy

Six studies were included to evaluate the efficacy of COVID-19 vaccines in patients with autoimmune diseases. Four studies used the inactivated virus vaccine [72,75,76,77], whereas the other two studies used mRNA and adenovirus vector vaccines [97,98] Breakthrough COVID-19 infections were used to assess vaccine efficacy.
Based on Figure 2, the overall effect on breakthrough COVID-19 infection after receipt of a COVID-19 inactivated virus vaccine was in favor of the healthy controls. The combined risk ratio was 1.93 (95% CI: 1.14–3.29, I2 = 0%), and the difference was statistically significant (p = 0.02). According to the GRADE system, the certainty of the evidence on breakthrough COVID-19 infections after inactivated vaccination was moderate (Supplementary Materials, Table S3). Four studies included in this meta-analysis involved patients with various autoimmune diseases: SLE, systemic autoimmune myopathies, and other autoimmune diseases. Patients involved in these four studies received various immunosuppressive treatments: steroids, methotrexate, hydroxychloroquine, mycophenolate mofetil, azathioprine, biologic agents, and others.
We also analyzed the combined risk ratio for breakthrough infections after mRNA or adenovirus vector vaccination, but no statistically significant difference was observed (RR = 0.97; 95% CI: 0.85–1.11; I2 = 0%) (Figure 3). According to the GRADE system, the certainty of the evidence on breakthrough COVID-19 infections after mRNA or adenovirus vector vaccination was moderate (Supplementary Materials). Three studies included in this meta-analysis involved patients with various autoimmune diseases: SLE, rheumatoid arthritis, spondiloarthopathy, vasculitis, and others. Patients involved in these four studies received various immunosuppressive treatments: steroids, methotrexate, hydroxychloroquine, leflunomide, mycophenolate mofetil, azathioprine, biologic agents, and others. Subgroup analysis regarding autoimmune diagnosis and treatment could not be done because of limited studies or a lack of subgroup data.

3.4.2. Immunogenicity

Eighteen studies were included in the meta-analysis to evaluate the immunogenicity of COVID-19 vaccines in patients with autoimmune disease compared with healthy controls [8,35,36,43,50,52,54,61,62,65,72,74,75,76,77,80,83,101]. Studies included in this meta-analysis involved patients with various autoimmune diseases: multiple sclerosis, systemic autoimmune diseases, and other autoimmune diseases. Patients involved in these studies received various immunosuppressive treatments: steroids, methotrexate, hydroxychloroquine, mycophenolate mofetil, azathioprine, biologic agents, and others.
Eleven studies were on mRNA vaccines [8,35,36,43,50,52,54,61,62,65,101] and seven studies [72,74,75,76,77,80,83] on inactivated vaccines. Seroconversion, proportion of neutralizing antibodies (NAb) positive, log total antibody (TAb) titer, and neutralizing activity were analyzed.
As shown in Figure 4, seven studies reported TAb titers after mRNA vaccination. Patients with autoimmune disease showed significantly lower log TAb (log BAU/mL) titers than healthy controls. Heterogeneity was low (SMD = −0.11, 95% CI = −0.2–0.02, I2 = 0%). According to the GRADE system, the certainty of the evidence on TAb after mRNA vaccination was high (Supplementary Materials, Table S3).
As shown in Figure 5, five studies reported Tab titers after inactivated vaccination. Patients with autoimmune disease showed significantly lower log Tab (log BAU/mL) titers compared with healthy controls. Heterogeneity was considerably low (SMD = −0.10, 95% CI = −0.19–0.00, I2 = 43%). According to the GRADE system, the certainty of the evidence on TAb titer after inactivated vaccination was high (Supplementary Materials, Table S3).
As shown in Figure 6, 11 studies reported IgG seroconversion after mRNA vaccination compared with healthy controls. IgG Seroconversion after mRNA vaccination was significantly lower among patients with autoimmune disease than healthy controls. Heterogeneity was high (RR = 0.82, 95% CI = 0.75–0.90, I2 = 97%). According to the GRADE system, the certainty of the evidence on IgG seroconversion after mRNA vaccination was moderate (Supplementary Materials).
As shown in Figure 7, seven studies reported IgG seroconversion after inactivated vaccination compared with healthy controls. IgG seroconversion after inactivated vaccination was significantly lower among patients with autoimmune disease than healthy controls. Heterogeneity was considerably high (RR = 0.77, 95% CI = 0.71–0.84, I2 = 86%). According to the GRADE system, the certainty of the evidence on IgG seroconversion after mRNA vaccination was moderate (Supplementary Materials, Table S3).
As shown in Figure 8, three studies reported neutralizing antibodies after mRNA vaccination. Patients with autoimmune disease showed a lower proportion of positive NAb than healthy controls, but the difference was not statistically significant. Heterogeneity was high (RR = 0.79, 95% CI = 0.54–1.14, I2 = 97%). According to the GRADE system, the certainty of the evidence on neutralizing antibodies after mRNA vaccination was very low (Supplementary Materials, Table S3).
As shown in Figure 9, seven studies reported neutralizing antibodies after inactivated vaccination. Patients with autoimmune disease had a significantly lower proportion of positive NAb than healthy controls. Heterogeneity was considerably low (RR = 0.71, 95% CI = 0.68–0.74, I2 = 37%). According to the GRADE system, the certainty of the evidence on neutralizing antibodies after inactivated vaccination was high (Supplementary Materials, Table S3).
As shown in Figure 10, six studies reported neutralizing activity after inactivated vaccination. Patients with autoimmune disease showed lower mean neutralizing activity after inactivated vaccination than healthy controls, but the result was not statistically significant. Heterogeneity was high (SMD = −0.52, 95% CI = −1.34–0.30, I2 = 98%). According to the GRADE system, the certainty of the evidence on neutralizing antibodies after the first dose of vaccine was very low (Supplementary Materials, Table S3).

3.4.3. Safety

Four studies were eligible for pooling of vaccine-associated adverse events, including local and systemic adverse events. All included studies were on inactivated COVID-19 vaccines [72,75,76,77]. Four studies included in this meta-analysis involved patients with various autoimmune diseases: SLE, systemic autoimmune myopathies, and other autoimmune diseases. Patients involved in these four studies received various immunosuppressive treatments: steroids, methotrexate, hydroxychloroquine, mycophenolate mofetil, azathioprine, biologic agents, and others.
We observed that the combined risk ratio for local adverse events after a first dose of COVID-19 inactivated vaccine was 1.26 (95% CI: 1.05–1.51; I2 = 0%) (Figure 11). Patients with autoimmune diseases had a statistically significant (p = 0.01) risk of local adverse events after receiving a first dose of COVID-19 inactivated vaccine in comparison with healthy controls. According to the GRADE system, the certainty of the evidence on local adverse events after first dose COVID-19 inactivated vaccine was high (Supplementary Materials, Table S3).
We observed that the combined risk ratio for local adverse events after a second dose COVID-19 inactivated vaccine was 1.11 (95% CI: 0.91–1.35; I2 = 1%) (Figure 12). Patients with autoimmune diseases had a higher risk of local adverse events than healthy controls after receiving a second dose of COVID-19 inactivated vaccine, but the difference was not statistically significant (p = 0.31). According to the GRADE system, the certainty of the evidence for local adverse events after a second dose of COVID-19 inactivated vaccine was high (Supplementary Materials, Table S3).
We observed that the combined risk ratio for systemic adverse events after a first dose of COVID-19 inactivated vaccine was 1.31 (95% CI: 1.15–1.48; I2 = 0%). Patients with autoimmune diseases had a statistically significant (p < 0.0001) risk of systemic adverse events after receiving a first dose of COVID-19 inactivated vaccine in comparison with healthy controls (Figure 13). According to the GRADE system, the certainty of the evidence on systemic adverse events after a first dose of COVID-19 inactivated vaccine was high (Supplementary Materials, Table S3).
The combined risk ratio for systemic adverse events after a second dose of COVID-19 inactivated vaccine was 1.13 (Figure 14), but no statistically significant difference was observed (95% CI: 0.88–1.45; I2 = 62%). According to the GRADE system, the certainty of the evidence on local adverse events was moderate (Supplementary Materials, Table S3).

3.5. Publication Bias

We used a funnel plot to assess publication bias for a meta-analysis involving more than 10 studies: IgG seroconversion after mRNA vaccination (Supplementary Materials, Figure S2). The funnel plot was asymmetrical, which could indicate that there was publication bias.

4. Discussion

There are some issues regarding COVID-19 vaccination in autoimmune patients, such as how autoimmune medications might affect the efficacy and immunogenicity of the vaccines and possible adverse reactions following COVID-19 vaccination. Therefore, the efficacy, immunogenicity, and safety of COVID-19 vaccines in autoimmune patients were the primary outcomes in this systematic review and meta-analysis.
Only a few studies were identified that addressed all three outcomes. In the meta-analysis, we compared efficacy, immunogenicity, and safety between patients with autoimmune diseases and healthy controls. Because of the heterogeneity of the studies, we only had non-randomized studies that could be used for this purpose. We also could not conduct a meta-analysis on booster (third-dose) COVID-19 vaccination due to limited studies sharing similar outcomes and interventions.
Regarding the efficacy of COVID-19 vaccination, our meta-analysis showed that the risk of breakthrough COVID-19 infection significantly increased in patients with autoimmune diseases compared with healthy controls after receipt of an inactivated virus vaccine. On the other hand, a meta-analysis with studies using mRNA or adenovirus vectors did not show significant differences in breakthrough infections among patients with autoimmune disease compared with healthy controls. Breakthrough COVID-19 infection can be related to viral profile, host factors (comorbidities, immunosuppressive drugs), and vaccine platform or dose. The mRNA vaccine platform shows stronger neutralizing antibody and T cell responses compared with other vaccine platforms [104].
Ahmed et al. reported that only small numbers of breakthrough infections occurred in patients with autoimmune diseases after they received either an inactivated or adenovirus vector vaccine [103]. Furer et al. observed no symptomatic COVID-19 infections in patients with autoimmune diseases, and only one subject in the healthy control group was diagnosed with a breakthrough COVID-19 infection after a second dose of mRNA vaccine during the study follow-up [35]. Moreover, Stalman et al. reported breakthrough COVID-19 infections after mRNA or adenovirus vector vaccine in both autoimmune patients and healthy controls, with no differences in the trends in the incidence rates [97]. Kim et al. also reported breakthrough infections after booster vaccination with an mRNA vaccine in subjects given an mRNA or adenovirus vector vaccine as their primary COVID-19 vaccination, but the result was not significantly different between patients with autoimmune disease and healthy controls (healthcare workers) [95].
Studies included in a meta-analysis of breakthrough infections after mRNA or adenovirus vector vaccination involved patients with various diagnoses and treatments for autoimmune diseases. Patel et al. and Paik et al. explained that increased breakthrough infections were associated with the use of multiple immunomodulatory therapies, such as methotrexate, mycophenolate mofetil, anti-CD20, and TNF inhibitors [105,106]. A study by Bieber et al. also showed higher doses of steroids and higher proportions of patients given TNF alpha inhibitors, rituximab, and calcineurin inhibitors among cases of breakthrough COVID-19 infection [33].
Regarding the immunogenicity of the vaccine, our meta-analyses showed that patients with autoimmune diseases had reduced total antibody (TAb) titers, IgG seroconversion, and neutralizing antibodies after COVID-19 inactivated vaccination compared with healthy controls. Patients with autoimmune diseases also showed reduced TAb titers and IgG seroconversion after COVID-19 mRNA vaccination compared with healthy controls. A study by Kim et al. on mRNA vaccine boosters showed that limited neutralization of the Omicron variant in the sera of patients with autoimmune disease could contribute to a shorter median time between third-dose vaccination and the time of breakthrough infection compared with a control group [95].
Patients with autoimmune diseases showed noticeably different humoral responses following vaccination, which may be attributed to the use of B-cell-depleting agents, antimetabolites, glucocorticoids, other immunosuppressive drugs, and waning immunity [106]. This was proven by Ferri et al. in a study that showed an increased prevalence of non-responders to vaccines in patients with systemic autoimmune disease treated with glucocorticoids, mycophenolate mofetil, and rituximab [8]. So et al. found that impaired humoral response in SLE patients significantly correlated with the use of mycophenolate and the type of vaccine, especially inactivated virus vaccines in comparison with mRNA vaccines [107]. Paik et al. reported that B-cell-depleting agents, antimetabolites, glucocorticoids, and combination immunosuppressive therapy achieved significantly lower seroconversion, while immunomodulators, such as hydroxychloroquine and intravenous globulin, did not reduce antibody titers [106]. However, patients treated with hydroxychloroquine, combined with other therapies such as methotrexate and/or sulfasalazine, still had significantly lower anti-SARS-CoV spike IgG antibody titers than those who did not receive such a combination [96].
In terms of the safety of vaccination, the overall estimate from the meta-analysis showed a significantly higher risk for patients with autoimmune disease experiencing local and systemic adverse events after a first dose of COVID-19 inactivated vaccine in comparison with healthy controls; however, no statistically significant difference after a second dose of vaccine was observed. Higher frequencies of adverse events were reported among seropositive patients than in seronegative patients and healthy controls [72]. No moderate or severe adverse events related to the vaccine were reported [72,75,76,77]. Vaccine-related adverse events after the inactivated COVID-19 vaccine, especially systemic symptoms, were fewer than those reported with the mRNA vaccine [75].
In our systematic review, flare (worsening of autoimmune disease activity) was observed in more than 10% of patients with SLE after primary mRNA vaccination [70], and in patients with hematologic autoimmune diseases including immune-mediated thrombotic thrombocytopenic purpura and immune thrombocytopenia after a booster mRNA vaccination [32,42]. Meanwhile, this occurred in less than 5% of patients with multiple sclerosis [29] and with systemic autoimmune diseases including cryoglobulinemic vasculitis, rheumatoid arthritis, systemic lupus erythematosus, and systemic sclerosis after primary mRNA vaccination [8]. For the other vaccine types, flare was observed in 7% of autoimmune skin disease patients after primary inactivated virus vaccine and in less than 5% of SLE and autoimmune rheumatic disease patients [77,80]. Other adverse events, such as face tingling, herpes reactivation, bleeding, and urinary tract infection, also occurred in a small number of patients, together with severe adverse events such as high blood pressure, immune thrombocytopenic purpura, myocarditis, and death [29,80,101]. However, causal and temporal relationships between vaccine administration and adverse events or worsening disease activity following vaccination were difficult to determine due to limited data and the lack of a specific analysis of the causal relationship.
Based on our qualitative findings, breakthrough infections occurred less frequently in autoimmune patients after a booster dose. Autoimmune patients still had lower humoral and cellular responses even after having a third vaccine dose. Most of the patients were on immunosuppressant therapy, while untreated patients had better humoral and cellular responses. These findings support some previous evidence regarding the effects of booster vaccination. Regardless of the lower antibody titers in autoimmune patients, a potential increase in titer could be achieved after administering a third dose of vaccine, though the titer was still lower compared with a healthy control group. Evidence from a study by Joudeh et al. indicates that a booster vaccine dose is associated with a higher seroconversion rate, particularly in patients with a history of COVID-19 infection [108]. Further evidence comes from Cardelli, et al., who showed that a time-dependent decrease in protective antibody titer was restored after receipt of a booster dose. After a booster dose, five of nine non-responders developed adequate anti-RBD and neutralizing antibody titers. Three of them reduced their dose of or discontinued mycophenolate mofetil or azathioprine therapy before booster administration [109]. In addition, in terms of efficacy and safety, Dreyer at al. found no relapse activity or breakthrough infections after the third dose of vaccine [34].
This study has several limitations. First, the number of studies used to combine the efficacy, immunogenicity, and safety findings was relatively small. Second, considering that only one RCT was available comparing patients with autoimmune diseases and healthy controls after a primary dose of COVID-19 vaccine, we only included non-randomized studies. Third, since we only included a small number of studies in our meta-analysis, we might have significant publication bias. However, we also included pre-printed studies in our systematic review to reduce the possibility of this bias. Fourth, the variety of autoimmune diagnoses and immunosuppressive treatments could have an impact on the outcome of COVID-19 vaccination. This could affect our meta-analysis, and we could not address this by subgroup analysis due to the limited studies available.

5. Conclusions

In conclusion, from this meta-analysis, we found that patients with autoimmune diseases showed significantly more breakthrough COVID-19 infections and lower total antibody (TAb) titers, IgG seroconversion, and neutralizing antibodies after inactivated COVID-19 vaccination compared with healthy controls. They also had more local and systemic adverse events after a first dose of inactivated vaccination compared with healthy controls, but this result was not seen after a second dose. Patients with autoimmune diseases also showed significantly lower TAb titers and IgG seroconversion after COVID-19 mRNA vaccination compared with healthy controls.
A second dose of vaccine was, however, found to be important, since it is associated with improved antibody titers and seroconversion. It is important to consult a healthcare provider before taking a vaccine, since immunosuppressants might affect the immunogenicity of vaccines. Additionally, the administration of third doses of COVID-19 vaccines should be considered due to improved seroprotection in these patients.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/vaccines11091456/s1, Table S1: Search strategy; Table S2: Summarize of research articles; Table S3: GRADE asessement; Figure S1: (A): Quality assessment of RCT studies, (B): Quality assessment of non-RCT studies, (C): Quality assessment of case-series studies, (D): Quality assessment of cross-sectional studies; Figure S2: Funnel plot of IgG seroconversion after mRNA vaccination.

Author Contributions

Conceptualization, A.W., A.S.H., S.M., Y.O., C.Y., S.K., E.Y. and S.D.; methodology, R.R., Y.O., C.Y. and K.H.; software, R.R. and C.Y.; validation, A.S.H., K.H, C.Y., S.K. and E.Y.; formal analysis, A.W., A.S.H., R.R., S.M., M.I.H., Y.O., C.Y. and K.H.; investigation, A.W., A.S.H., R.R., S.M., M.I.H., Y.O., C.Y. and A.N.L.S.; resources, Y.O., C.Y., S.K., E.Y. and S.D.; data curation, A.W., R.R. and M.I.H.; writing—original draft preparation, A.W., A.S.H., R.R., S.M. and M.I.H.; writing—review and editing, Y.O., C.Y., K.H, S.K., E.Y. and S.D.; visualization, R.R.; supervision, S.K., E.Y. and S.D.; project administration, R.R.; funding acquisition, A.W., A.S.H., Y.O. and C.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This systematic review received study funding from the University of Indonesia, grant number NKB-745/UN2.RST/HKP.05.00/2022.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Acknowledgments

We express our appreciation to Annisa Layalia Widjanarko, who helped with data collection.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO. WHO Coronavirus (COVID-19) Dashboard|WHO Coronavirus (COVID-19) Dashboard with Vaccination Data. 2022. Available online: https://covid19.who.int/ (accessed on 3 May 2022).
  2. Centers for Disease Control Prevention (CDC) People with Certain Medical, Conditions|CDC. Available online: https://www.cdc.gov/coronavirus/2019-ncov/need-extra-precautions/people-with-medical-conditions.html (accessed on 2 July 2022).
  3. Velikova, T.; Georgiev, T. SARS-CoV-2 vaccines and autoimmune diseases amidst the COVID-19 crisis. Rheumatol. Int. 2021, 41, 509–518. [Google Scholar] [CrossRef]
  4. Soy, M.; Keser, G.; Atagunduz, P.; Mutlu, M.Y.; Gunduz, A.; Koybaşi, G.; Bes, C. A practical approach for vaccinations including COVID-19 in autoimmune/autoinflammatory rheumatic diseases: A non-systematic review. Clin. Rheumatol. 2021, 40, 3533–3545. [Google Scholar] [CrossRef] [PubMed]
  5. Kaur, S.P.; Gupta, V. COVID-19 Vaccine: A comprehensive status report. Virus Res. 2020, 288, 198114. [Google Scholar] [CrossRef]
  6. World Health Organization (WHO). COVID-19 Vaccines. 2021. Available online: https://www.who.int/emergencies/diseases/novel-coronavirus-2019/covid-19-vaccines (accessed on 2 July 2021).
  7. Furer, V.; Rondaan, C.; Heijstek, M.W.; Agmon-Levin, N.; Assen, S.; Van Bijl, M.; Breedveld, F.C.; D’Amelio, R.; Dougados, M.; Kapetanovic, M.C.; et al. 2019 update of EULAR recommendations for vaccination in adult patients with autoimmune inflammatory rheumatic diseases. Ann. Rheum. Dis. 2020, 79, 39–52. [Google Scholar] [CrossRef] [PubMed]
  8. Ferri, C.; Ursini, F.; Gragnani, L.; Raimondo, V.; Giuggioli, D.; Foti, R.; Caminiti, M.; Olivo, D.; Cuomo, G.; Visentini, N.; et al. Impaired immunogenicity to COVID-19 vaccines in autoimmune systemic diseases. High prevalence of non-response in different patients’ subgroups. J. Autoimmun. 2021, 125, 102744. [Google Scholar] [CrossRef] [PubMed]
  9. Widhani, A.; Sukmana, N. Vaksinasi pada penyakit autoimun. In Dalam; Djauzi, S., Rengganis, I., Sundoro, J., Koesnoe, S., Soegiarto, G., Maria, S., Eds.; Pedoman Imunisasi Pada Orang Dewasa; Satgas Imunisasi Dewasa PAPDI: Jakarta, Indonesia, 2017. [Google Scholar]
  10. Toussirot, É.; Bereau, M. Vaccination and induction of autoimmune diseases. Inflamm. Allergy Drug Targets 2015, 14, 94–98. [Google Scholar] [CrossRef]
  11. Soriano, A.; Nesher, G.; Shoenfeld, Y. Predicting post-vaccination autoimmunity: Who might be at risk? Pharmacol. Res. 2015, 92, 18–22. [Google Scholar] [CrossRef]
  12. Shoenfeld, Y. Corona (COVID-19) time musings: Our involvement in COVID-19 pathogenesis, diagnosis, treatment and vaccine planning. Autoimmun. Rev. 2020, 19, 102538. [Google Scholar] [PubMed]
  13. Vojdani, A.; Kharrazian, D. Potential antigenic cross-reactivity between SARS-CoV-2 and human tissue with a possible link to an increase in autoimmune diseases. Clin. Immunol. 2020, 217, 108480. [Google Scholar] [CrossRef]
  14. Voysey, M.; Clemens, S.A.C.; Madhi, S.A.; Weckx, L.Y.; Folegatti, P.M.; Aley, P.K.; Angus, B.; Baillie, V.L.; Barnabas, S.L.; Bharat, Q.E.; 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, 99–111. [Google Scholar] [CrossRef]
  15. Pottegård, A.; Lund, L.C.; Karlstad, Ø.; Dahl, J.; Andersen, M.; Hallas, J.; Lidegaard, Ø.; Tapi, G.; Gulseth, H.L.; Ruiz, P.L.; et al. Arterial events, venous thromboembolism, thrombocytopenia, and bleeding after vaccination with Oxford-AstraZeneca ChAdOx1-S in Denmark and Norway: Population based cohort study. BMJ 2021, 373, n1114. [Google Scholar] [CrossRef]
  16. Schultz, N.H.; Sørvoll, I.H.; Michelsen, A.E.; Munthe, L.A.; Lund-Johansen, F.; Ahlen, M.T.; Wiedmann, M.; Aamodt, A.; Skattør, T.H.; Tjønnfjord, G.E.; et al. Thrombosis and Thrombocytopenia after ChAdOx1 nCoV-19 Vaccination. N. Engl. J. Med. 2021, 384, 2124–21230. [Google Scholar] [CrossRef]
  17. Greinacher, A.; Thiele, T.; Warkentin, T.E.; Weisser, K.; Kyrle, P.A.; Eichinger, S. Thrombotic Thrombocytopenia after ChAdOx1 nCov-19 Vaccination. N. Eng. J. Med. 2021, 384, 2092–2101. [Google Scholar] [CrossRef]
  18. Scully, M.; Singh, D.; Lown, R.; Poles, A.; Solomon, T.; Levi, M.; Goldblatt, D.; Kotoucek, P.; Thomas, W.; Lester, W. Pathologic Antibodies to Platelet Factor 4 after ChAdOx1 nCoV-19 Vaccination. N. Eng. J. Med. 2021, 384, 2202–2211. [Google Scholar] [CrossRef] [PubMed]
  19. Bril, F.; Diffalha SAl Dean, M.; Fettig, D.M. Autoimmune hepatitis developing after coronavirus disease 2019 (COVID-19) vaccine: Causality or casualty? J. Hepatol. 2021, 75, 222–224. [Google Scholar] [CrossRef] [PubMed]
  20. Lee, E.J.; Cines, D.B.; Gernsheimer, T.; Kessler, C.; Michel, M.; Tarantino, M.D.; Semple, J.W.; Arnold, D.M.; Godeau, B.; Lambert, M.P.; et al. Thrombocytopenia following Pfizer and Moderna SARS-CoV-2 vaccination. Am. J. Hematol. 2021, 96, 534–537. [Google Scholar] [CrossRef]
  21. Hazlewood, G.S.; Pardo, J.P.; Barnabe, C.; Schieir, O.; Barber, C.E.H.; Bernatsky, S.; Colmegna, I.; Hitcon, C.; Loeb, M.; Mertz, D.; et al. Canadian rheumatology association recommendation for the use of COVID-19 vaccination for patients with autoimmune rheumatic diseases. J. Rheumatol. 2021, 48, 1330–1339. [Google Scholar] [CrossRef] [PubMed]
  22. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred Reporting Items for Systematic Reviews and Meta-Analyses: The PRISMA Statement. Ann. Intern. Med. 2009, 151, 264–269. [Google Scholar] [CrossRef]
  23. Infantino, M.; Pieri, M.; Nuccetelli, M.; Grossi, V.; Lari, B.; Tomassetti, F.; Calugi, G.; Pancani, S.; Benucci, M.; Casprini, P.; et al. The WHO International Standard for COVID-19 serological tests: Towards harmonization of anti-spike assays. Int. Immunopharmacol. 2021, 100, 108095. [Google Scholar] [CrossRef] [PubMed]
  24. Valcourt, E.J.; Manguiat, K.; Robinson, A.; Chen, J.C.; Dimitrova, K.; Philipson, C.; Lamoreux, L.; McLachlan, E.; Schiffman, Z.; Drebot, M.A.; et al. Evaluation of a commercially-available surrogate virus neutralization test for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Diagn. Microbiol. Infect. Dis. 2021, 99, 115294. [Google Scholar] [CrossRef] [PubMed]
  25. Wan, X.; Wang, W.; Liu, J.; Tong, T. Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC Med. Res. Methodol. 2014, 14, 135. [Google Scholar] [CrossRef]
  26. Higgins, J.; Thomas, J.; Chandler, J.; Cumpston, M.; Li, T.; Page, M.; Welch, V.A. Cochrane Handbook for Systematic Reviews of Interventions, Version 6.3; Updated February 2022; Cochrane: 2022. Available online: https://training.cochrane.org/handbook (accessed on 23 February 2023).
  27. Wells, G.A.; Shea, B.; O’Connel, D.; Peterson, J.; Welch, V.; Losos, M.; Tugwell, P. The Newcastle-Ottawa Scale (NOS) for Assessing the Quality of Nonrandomised Studies in Meta-Analyses. Available online: https://www.ohri.ca/programs/clinical_epidemiology/oxford.asp (accessed on 1 April 2023).
  28. The National Institutes of Health. Study Quality Assessment Tools. 2013. Available online: https://www.nhlbi.nih.gov/health-topics/study-quality-assessment-tools (accessed on 1 April 2023).
  29. Achiron, A.; Dolev, M.; Menascu, S.; Zohar, D.N.; Dreyer-Alster, S.; Miron, S.; Shirbint, E.; Megalashvili, D.; Fletcher, S.; Givon, U.; et al. COVID-19 vaccination in patients with multiple sclerosis: What we have learnt by February 2021. Mult. Scler. 2021, 27, 864–870. [Google Scholar] [CrossRef]
  30. Achiron, A.; Mandel, M.; Dreyer-Alster, S.; Harari, G.; Magalashvili, D.; Sonis, P.; Dolev, M.; Menascu, S.; Fletcher, S.; Falb, R.; et al. Humoral immune response to COVID-19 mRNA vaccine in patients with multiple sclerosis treated with high-efficacy disease-modifying therapies. Ther. Adv. Neurol. Disord. 2021, 14, 17562864211022581. [Google Scholar] [CrossRef] [PubMed]
  31. Achiron, A.; Mandel, M.; Gurevich, M.; Dreyer-Alster, S.; Magalashvili, D.; Sonis, P.; Dolev, M.; Menascu, S.; Harari, G.; Fletcher, S.; et al. Immune response to the third COVID-19 vaccine dose is related to lymphocyte count in multiple sclerosis patients treated with fingolimod. J. Neurol. 2021, 269, 2286–2292. [Google Scholar] [CrossRef] [PubMed]
  32. Aharoni, M.; Leader, A.; Shochat, T.; Raanani, P.; Spectre, G. Exacerbation of immune thrombocytopenia following initial and booster vaccination with Pfizer-BioNTech COVID-19 vaccine. Platelets 2022, 33, 781–786. [Google Scholar] [CrossRef] [PubMed]
  33. Bieber, A.; Sagy, I.; Novack, L.; Brikman, S.; Abuhasira, R.; Ayalon, S.; Novofastovski, I.; Abu-Shakra, M.; Mader, R. BNT162b2 mRNA COVID-19 vaccine and booster in patients with autoimmune rheumatic diseases: A national cohort study. Ann. Rheum. Dis. 2022, 81, 1028–1035. [Google Scholar] [CrossRef] [PubMed]
  34. Dreyer-Alster, S.; Menascu, S.; Mandel, M.; Shirbint, E.; Magalashvili, D.; Dolev, M.; Fletcher, S.; Givon, U.; Guber, D.; Stern, Y.; et al. COVID-19 vaccination in patients with multiple sclerosis: Safety and humoral efficacy of the third booster dose. J. Neurol. Sci. 2022, 434, 120155. [Google Scholar] [CrossRef]
  35. Furer, V.; Eviatar, T.; Zisman, D.; Peleg, H.; Paran, D.; Levartovsky, D.; Zisapel, M.; Elalouf, O.; Kaufman, I.; Meidan, R.; et al. Immunogenicity and safety of the BNT162b2 mRNA COVID-19 vaccine in adult patients with autoimmune inflammatory rheumatic diseases and in the general population: A multicentre study. Ann. Rheum. Dis. 2021, 80, 1330–1338. [Google Scholar] [CrossRef] [PubMed]
  36. Furer, V.; Eviatar, T.; Zisman, D.; Peleg, H.; Braun-Moscovici, Y.; Balbir-Gurman, A.; Paran, D.; Levartovsky, D.; Zisapel, M.; Elalouf, O.; et al. Predictors of Immunogenic Response to the BNT162b2 mRNA COVID-19 Vaccination in Patients with Autoimmune Inflammatory Rheumatic Diseases Treated with Rituximab. Vaccines 2022, 10, 901. [Google Scholar] [CrossRef]
  37. Furer, V.; Zisman, D.; Kibari, A.; Rimar, D.; Paran, Y.; Elkayam, O. Herpes zoster following BNT162b2 mRNA COVID-19 vaccination in patients with autoimmune inflammatory rheumatic diseases: A case series. Rheumatology 2021, 60, SI90–SI95. [Google Scholar] [CrossRef] [PubMed]
  38. Lustig, Y.; Sapir, E.; Regev-Yochay, G.; Cohen, C.; Fluss, R.; Olmer, L.; Indebaum, V.; Mandelboim, M.; Doolman, R.; Amit, S.; et al. BNT162b2 COVID-19 vaccine and correlates of humoral immune responses and dynamics: A prospective, single-centre, longitudinal cohort study in health-care workers. Lancet Respir. Med. 2021, 9, 999–1009. [Google Scholar] [CrossRef] [PubMed]
  39. Ammitzbøll, C.; Bartels, L.E.; Andersen, J.B.; Vils, S.R.; Mistegård, C.E.; Johannsen, A.D.; Hermansen, M.F.; Thomsen, M.K.; Erikstrup, C.; Hauge, E.; et al. Impaired Antibody Response to the BNT162b2 Messenger RNA Coronavirus Disease 2019 Vaccine in Patients With Systemic Lupus Erythematosus and Rheumatoid Arthritis. ACR Open Rheumatol. 2021, 3, 622–628. [Google Scholar] [CrossRef] [PubMed]
  40. Dinoto, A.; Gastaldi, M.; Iorio, R.; Marini, S.; Damato, V.; Farina, A.; Zoccarato, M.; Sechi, E.; Pinna, F.; Maniscalco, G.T.; et al. Safety profile of SARS-CoV-2 vaccination in patients with antibody-mediated CNS disorders. Mult. Scler. Relat. Disord. 2022, 63, 103827. [Google Scholar] [CrossRef] [PubMed]
  41. Fabris, M.; de Marchi, G.; Domenis, R.; Caponnetto, F.; Guella, S.; Secco, C.D.; Cabas, N.; De Vita, S.; Beltrami, A.P.; Curcio, F.; et al. High T-cell response rate after COVID-19 vaccination in belimumab and rituximab recipients. J. Autoimmun. 2022, 129, 102827. [Google Scholar] [CrossRef] [PubMed]
  42. Giuffrida, G.; Markovic, U.; Condorelli, A.; Calagna, M.; Grasso, S.; Duminuco, A.; Riccobene, C.; Pelle, A.C.; Zanghi, G.; Di Raimondo, F. Relapse of immune-mediated thrombotic thrombocytopenic purpura following mRNA COVID-19 vaccination: A prospective cohort study. Haematologica 2022, 107, 2661–2666. [Google Scholar] [CrossRef]
  43. Malipiero, G.; Moratto, A.; Infantino, M.; D’Agaro, P.; Piscianz, E.; Manfredi, M.; Grossi, V.; Benvenuti, E.; Bulgaresi, M.; Benucci, M.; et al. Assessment of humoral and cellular immunity induced by the BNT162b2 SARS-CoV-2 vaccine in healthcare workers, elderly people, and immunosuppressed patients with autoimmune disease. Immunol. Res. 2021, 69, 576–583. [Google Scholar] [CrossRef]
  44. Zecca, E.; Rizzi, M.; Tonello, S.; Matino, E.; Costanzo, M.; Rizzi, E.; Casciaro, G.F.; Manfredi, G.F.; Acquaviva, A.; Gagliardi, I.; et al. Ongoing Mycophenolate Treatment Impairs Anti-SARS-CoV-2 Vaccination Response in Patients Affected by Chronic Inflammatory Autoimmune Diseases or Liver Transplantation Recipients: Results of the RIVALSA Prospective Cohort. Viruses 2022, 14, 1766. [Google Scholar] [CrossRef]
  45. Gerosa, M.; Schioppo, T.; Argolini, L.M.; Sciascia, S.; Ramirez, G.A.; Moroni, G.; Sinico, R.A.; Bonelli, G.; Alberici, F.; Mescia, F.; et al. The Impact of Anti-SARS-CoV-2 Vaccine in Patients with Systemic Lupus Erythematosus: A Multicentre Cohort Study. Vaccines 2022, 10, 663. [Google Scholar] [CrossRef]
  46. Costa, C.; Scozzari, G.; Migliore, E.; Galassi, C.; Ciccone, G.; Ricciardelli, G.; Scarmozzino, A.; Angeloe, L.; Cassoni, P.; Cavallo, R. Cellular Immune Response to BNT162b2 mRNA COVID-19 Vaccine in a Large Cohort of Healthcare Workers in a Tertiary Care University Hospital. Vaccines 2022, 10, 1031. [Google Scholar] [CrossRef]
  47. Deepak, P.; Kim, W.; Paley, M.A.; Yang, M.; Carvidi, A.B.; El-Qunni, A.A. Glucocorticoids and B Cell Depleting Agents Substantially Impair Immunogenicity of mRNA Vaccines to SARS-CoV-2. medRXiv 2021. [Google Scholar] [CrossRef]
  48. Epstein, S.; Xia, Z.; Lee, A.J.; Dahl, M.; Edwards, K.; Levit, E.; Longbrake, E.E.; Perrone, C.; Kavak, K.; Weinstock-Guttman, B.; et al. Vaccination Against SARS-CoV-2 in Neuroinflammatory Disease: Early Safety/Tolerability Data. Mult. Scler. Relat. Disord. 2022, 57, 103433. [Google Scholar] [CrossRef] [PubMed]
  49. Sakano, T.; Bittner, E.A.; Chang, M.G. Severe COVID pneumonia and undetectable B cells after vaccination in patients previously treated with rituximab: A case series. Postgrad. Med. 2022, 134, 239–243. [Google Scholar] [CrossRef]
  50. Geisen, U.M.; Berner, D.K.; Tran, F.; Sümbül, M.; Vullriede, L.; Ciripoi, M.; Reid, H.M.; Schaffarzyk, A.; Longardt, A.C.; Franzenburg, J.; et al. Immunogenicity and safety of anti-SARS-CoV-2 mRNA vaccines in patients with chronic inflammatory conditions and immunosuppressive therapy in a monocentric cohort. Ann. Rheum. Dis. 2021, 80, 1306–1311. [Google Scholar] [CrossRef]
  51. Geisen, U.M.; Rose, R.; Neumann, F.; Ciripoi, M.; Vullriede, L.; Reid, H.M.; Berner, D.K.; Bertoglio, F.; Hoff, P.; Hust, M.; et al. The long term vaccine-induced anti-SARS-CoV-2 immune response is impaired in quantity and quality under TNFα blockade. J. Med. Virol. 2022, 94, 5780–5789. [Google Scholar] [CrossRef] [PubMed]
  52. Giannoccaro, M.P.; Vacchiano, V.; Leone, M.; Camilli, F.; Zenesini, C.; Panzera, I.; Balboni, A.; Tappata, M.; Borghi, A.; Salvi, F.; et al. Difference in safety and humoral response to mRNA SARS-CoV-2 vaccines in patients with autoimmune neurological disorders: The ANCOVAX study. J. Neurol. 2022, 269, 4000–4012. [Google Scholar] [CrossRef] [PubMed]
  53. Meyer-Arndt, L.; Braun, J.; Fauchere, F.; Vanshylla, K.; Loyal, L.; Henze, L.; Kruse, B.; Dingeldey, M.; Jürchott, K.; Mangold, M.; et al. SARS-CoV-2 mRNA vaccinations fail to elicit humoral and cellular immune responses in patients with multiple sclerosis receiving fingolimod. J. Neurol. Neurosurg. Psychiatry 2022, 93, 960–971. [Google Scholar] [CrossRef]
  54. Schwarz, T.; Otto, C.; Jones, T.C.; Pache, F.; Schindler, P.; Niederschweiberer, M.; Schmidt, F.A.; Drosten, C.; Corman, V.M.; Ruprecht, K. Preserved T cell responses to SARS-CoV-2 in anti-CD20 treated multiple sclerosis. medRxiv 2021, 15, 17562864221141505. [Google Scholar] [CrossRef] [PubMed]
  55. Arnold, F.; Huzly, D.; Tanriver, Y.; Welte, T. Response to SARS-CoV-2 vaccines in patients receiving B-cell modulating antibodies for renal autoimmune disease. BMC Infect. Dis. 2021, 22, 734. [Google Scholar] [CrossRef]
  56. Duengelhoef, P.; Hartl, J.; Rüther, D.; Steinmann, S.; Brehm, T.T.; Weltzsch, J.P.; Glaser, F.; Schaub, G.M.; Sterneck, M.; Sebode, M.; et al. SARS-CoV-2 vaccination response in patients with autoimmune hepatitis and autoimmune cholestatic liver disease. United Eur. Gastroenterol. J. 2022, 10, 319–329. [Google Scholar] [CrossRef]
  57. Krasselt, M.; Wagner, U.; Nguyen, P.; Pietsch, C.; Boldt, A.; Baerwald, C.; Pierer, M.; Seifert, O. Humoral and cellular response to COVID-19 vaccination in patients with autoimmune inflammatory rheumatic diseases under real-life conditions. Rheumatology 2022, 61, 180–188. [Google Scholar] [CrossRef]
  58. Stefanski, A.L.; Rincon-Arevalo, H.; Schrezenmeier, E.; Karberg, K.; Szelinski, F.; Ritter, J.; Jahrsdorfer, B.; Schrenzenmeier, H.; Ludwig, C.; Sattler, A.; et al. B Cell Numbers Predict Humoral and Cellular Response Upon SARS–CoV-2 Vaccination Among Patients Treated With Rituximab. Arthritis Rheumatol. 2022, 74, 934–947. [Google Scholar] [CrossRef] [PubMed]
  59. Hills, T.; Paterson, A.; Woodward, R.; Middleton, F.; Carlton, L.H.; McGregor, R.; Barfoot, S.; Ramiah, C.; Whitcombe, A.L.; Zimbron, V.M.; et al. The effect of needle length and skin to deltoid muscle distance in adults receiving an mRNA COVID-19 vaccine. Vaccine 2022, 40, 4827–4834. [Google Scholar] [CrossRef]
  60. Kornek, B.; Leutmezer, F.; Rommer, P.S.; Koblischke, M.; Schneider, L.; Haslacher, H.; Thalhammer, R.; Zimprich, F.; Zulanher, G.; Bsteh, G.; et al. B Cell Depletion and SARS-CoV-2 Vaccine Responses in Neuroimmunologic Patients. Ann. Neurol. 2022, 91, 342–352. [Google Scholar] [CrossRef]
  61. Mandl, P.; Tobudic, S.; Haslacher, H.; Karonitsch, T.; Mrak, D.; Nothnagl, T.; Perkmann, T.; Radner, H.; Sautner, J.; Simader, E.; et al. Response to SARS-CoV-2 vaccination in systemic autoimmune rheumatic disease depends on immunosuppressive regimen: A matched, prospective cohort study. Ann. Rheum. Dis. 2022, 81, 1017–1022. [Google Scholar] [CrossRef]
  62. Wagner, A.; Garner-Spitzer, E.; Schötta, A.M.; Orola, M.; Wessely, A.; Zwazl, I.; Ohradanova-Repic, A.; Weseslindtner, L.; Tatji, G.; Gebetsberger, L.; 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. [Google Scholar] [CrossRef] [PubMed]
  63. Mena-Vázquez, N.; García-Studer, A.; Rojas-Gimenez, M.; Romero-Barco, C.M.; Manrique-Arija, S.; Mucientes, A.; Velloso-Feijoo, M.L.; Godoy-Navarette, F.J.; Morales-Garrido, P.; Redondo-Rodriguez, R.; et al. Importance of Vaccination against SARS-CoV-2 in Patients with Interstitial Lung Disease Associated with Systemic Autoimmune Disease. J. Clin. Med. 2022, 11, 2437. [Google Scholar] [CrossRef]
  64. Quintanilla-Bordás, C.; Gascón-Gimenez, F.; Alcalá, C.; Payá, M.; Mallada, J.; Silla, R.; Carratalá-Bosca, S.; Gasque-Rubio, R.; Castillo, J.; Casanova, B. Case Report: Exacerbation of Relapses Following mRNA COVID-19 Vaccination in Multiple Sclerosis: A Case Series. Front. Neurol. 2022, 13, 897275. [Google Scholar] [CrossRef] [PubMed]
  65. Santos, C.S.; Antolin, S.C.; Morales, C.M.; Herrero, J.G.; Alvarez, E.D.; Ortega, F.R.; de Morales, J.G.R. Immune responses to mRNA vaccines against SARS-CoV-2 in patients with immune-mediated inflammatory rheumatic diseases. RMD Open. 2022, 8, e001898. [Google Scholar] [CrossRef]
  66. Zabalza, A.; Arrambide, G.; Otero-Romero, S.; Pappolla, A.; Tagliani, P.; López-Maza, S.; Cárdenas-Robledo, S.; Esperalba, J.; Fernández-Naval, C.; Martínez-Gallo, M.; et al. Is humoral and cellular response to SARS-CoV-2 vaccine modified by DMT in patients with multiple sclerosis and other autoimmune diseases? Mult. Scler. 2022, 28, 1138–1145. [Google Scholar] [CrossRef]
  67. Mitsunaga, T.; Ohtaki, Y.; Seki, Y.; Yoshioka, M.; Mori, H.; Suzuka, M.; Mashiko, S.; Takeda, S.; Mashiko, K. Evaluation of the antibody response and adverse reactions of the BNT162b2 vaccine of participants with prior COVID-19 infection in Japan. medRxiv 2021. [Google Scholar] [CrossRef]
  68. Moyon, Q.; Sterlin, D.; Miyara, M.; Anna, F.; Mathian, A.; Lhote, R.; Ghillani-Dalbin, P.; Breillat, P.; Mudumba, S.; de Alba, S.; et al. BNT162b2 vaccine-induced humoral and cellular responses against SARS-CoV-2 variants in Systemic Lupus Erythematosus. Ann. Rheum. Dis. 2021, 81, 575–583. [Google Scholar] [CrossRef] [PubMed]
  69. Pinte, L.; Negoi, F.; Ionescu, G.D.; Caraiola, S.; Balaban, D.V.; Badea, C.; Mazilu, D.; Dumitrescu, B.; Mateescu, B.; Lonescu, R.; et al. COVID-19 Vaccine Does Not Increase the Risk of Disease Flare-Ups among Patients with Autoimmune and Immune-Mediated Diseases. J. Pers. Med. 2021, 11, 1283. [Google Scholar] [CrossRef]
  70. Zavala-Flores, E.; Salcedo-Matienzo, J.; Quiroz-Alva, A.; Berrocal-Kasay, A. Side effects and flares risk after SARS-CoV-2 vaccination in patients with systemic lupus erythematosus. Clin. Rheumatol. 2022, 41, 1349–1357. [Google Scholar] [CrossRef] [PubMed]
  71. Dayam, R.M.; Law, J.C.; Goetgebuer, R.L.; Chao, G.Y.C.; Abe, K.T.; Sutton, M.; Finkelstein, N.; Stempak, J.M.; Pereira, D.; Croitoru, D.; et al. Accelerated waning of immunity to SARS-CoV-2 mRNA vaccines in patients with immune-mediated inflammatory diseases. JCI Insight 2022, 7, e159721. [Google Scholar] [CrossRef]
  72. Aikawa, N.E.; Kupa, L.V.K.; Pasoto, S.G.; Medeiros-Ribeiro, A.C.; Yuki, E.F.N.; Saad, C.G.S.; Pedrosa, T.; Fuller, R.; Shinjo, S.K.; Sampaio-Barros, P.; et al. Immunogenicity and safety of two doses of the CoronaVac SARS-CoV-2 vaccine in SARS-CoV-2 seropositive and seronegative patients with autoimmune rheumatic diseases in Brazil: A subgroup analysis of a phase 4 prospective study. Lancet Rheumatol. 2022, 4, 113–124. [Google Scholar] [CrossRef]
  73. Araujo, C.S.R.; Medeiros-Ribeiro, A.C.; Saad, C.G.S.; Bonfiglioli, K.R.; Domiciano, D.S.; Shimabuco, A.Y.; Silvo, M.S.R.; Yuki, E.F.N.; Pasoto, S.G.; Pedrosa, T.; et al. Two-week methotrexate discontinuation in patients with rheumatoid arthritis vaccinated with inactivated SARS-CoV-2 vaccine: A randomised clinical trial. Ann. Rheum. Dis. 2022, 81, 889–897. [Google Scholar] [CrossRef]
  74. Gualano, B.; Lemes, I.R.; Silva, R.P.; Pinto, A.J.; Mazzolani, B.C.; Smaira, F.I.; Sieczkowska, S.M.; Aikawa, N.E.; Pasoto, S.G.; Medeiros-Ribeiro, A.C.; et al. Association between physical activity and immunogenicity of an inactivated virus vaccine against SARS-CoV-2 in patients with autoimmune rheumatic diseases. Brain Behav. Immun. 2022, 101, 49–56. [Google Scholar] [CrossRef] [PubMed]
  75. Medeiros-Ribeiro, A.C.; Aikawa, N.E.; Saad, C.G.S.; Yuki, E.F.N.; Pedrosa, T.; Fusco, S.R.G.; Rojo, P.T.; Pereira, R.M.R.; Shinjo, S.K.; Andrade, D.C.O.; et al. Immunogenicity and safety of the CoronaVac inactivated vaccine in patients with autoimmune rheumatic diseases: A phase 4 trial. Nat. Med. 2021, 27, 1744–1751. [Google Scholar] [CrossRef]
  76. Shinjo, S.K.; de Souza, F.H.C.; Borges, I.B.P.; Dos Santos, A.M.; Miossi, R.; Misse, R.G.; Medeiros-Ribeiro, A.C.; Saad, C.G.S.; Yuki, E.F.N.; Pasoto, S.G.; et al. Systemic autoimmune myopathies: A prospective phase 4 controlled trial of an inactivated virus vaccine against SARS-CoV-2. Rheumatology 2022, 61, 3351–3561. [Google Scholar] [CrossRef]
  77. Yuki, E.F.N.; Borba, E.F.; Pasoto, S.G.; Seguro, L.P.; Lopes, M.; Saad, C.G.S.; Medeiros-Ribeiro, A.C.; Silva, C.A.; de Andrade, D.C.O.; Kupa, L.D.K.; et al. Impact of Distinct Therapies on Antibody Response to SARS-CoV-2 Vaccine in Systemic Lupus Erythematosus. Arthritis Care Res. 2022, 74, 562–571. [Google Scholar] [CrossRef]
  78. Chen, J.; Cai, W.; Liu, T.; Zhou, Y.; Jin, Y.; Yang, Y.; Chen, S.; Tang, K.; Li, C. The COVID-19 vaccine: Attitudes and vaccination in patients with autoimmune inflammatory rheumatic diseases. Rheumatol. Autoimmun. 2022, 2, 82–91. [Google Scholar] [CrossRef]
  79. Huang, L.; Jiang, Z.; Zhou, J.; Chen, Y.; Huang, H. The Effect of Inactivated SARS-CoV-2 Vaccines on TRAB in Graves’ Disease. Front. Endocrinol. 2022, 13, 835880. [Google Scholar] [CrossRef]
  80. Seree-Aphinan, C.; Chanprapaph, K.; Rattanakaemakorn, P.; Setthaudom, C.; Suangtamai, T.; Pomsoong, C.; Ratanapokasatit, Y.; Suchonwanit, P. Inactivated COVID-19 Vaccine Induces a Low Humoral Immune Response in a Subset of Dermatological Patients Receiving Immunosuppressants. Front. Med. 2021, 8, 769845. [Google Scholar] [CrossRef]
  81. Assawasaksakul, T.; Sathitratanacheewin, S.; Vichaiwattana, P.; Wanlapakorn, N.; Poovorawan, Y.; Kittanamongkolchai, W. Immunogenicity, safety and reactogenicity of a heterogeneous booster following the CoronaVac inactivated SARS-CoV-2 vaccine in patients with SLE: A case series. RMD Open 2021, 7, e002019. [Google Scholar] [CrossRef] [PubMed]
  82. Assawasaksakul, T.; Lertussavavivat, T.; Sathitratanacheewin, S.; Oudomying, N.; Vichaiwattana, P.; Wanlapakorn, N.; Poorvorawan, Y.; Avihingsanon, Y.; Assawasaksakul, N.; Buranapraditkun, S.; et al. Comparison of Immunogenicity and Safety of Inactivated, Adenovirus-Vectored, and Heterologous Adenovirus-Vectored/mRNA Vaccines in Patients with Systemic Lupus Erythematosus and Rheumatoid Arthritis: A Prospective Cohort Study. Vaccines 2022, 10, 853. [Google Scholar] [CrossRef]
  83. Balcells, M.E.; Le Corre, N.; Durán, J.; Ceballos, M.E.; Vizcaya, C.; Mondaca, S.; Dib, M.; Rabagliati, R.; Sarmiento, M.; Burgos, P.I.; et al. Reduced Immune Response to Inactivated Severe Acute Respiratory Syndrome Coronavirus 2 Vaccine in a Cohort of Immunocompromised Patients in Chile. Clin. Infect. Dis. 2022, 75, e594–e602. [Google Scholar] [CrossRef] [PubMed]
  84. Shenoy, P.; Ahmed, S.; Paul, A.; Cherian, S.; Umesh, R.; Shenoy, V.; Vijayan, A.; Babu, S.; Nivin, S.; Thambi, A. Hybrid immunity versus vaccine-induced immunity against SARS CoV2 in Patients with Autoimmune Rheumatic Diseases. medRxiv 2021, 4, e80–e82. [Google Scholar] [CrossRef]
  85. Timmermann, L.; Globke, B.; Lurje, G.; Schmelzle, M.; Schöning, W.; Öllinger, R.; Pratschke, J.; Bettina, E.; Drosten, C.; Hofmann, J.; et al. Humoral Immune Response following SARS-CoV-2 Vaccination in Liver Transplant Recipients. Vaccines 2021, 9, 1422. [Google Scholar] [CrossRef]
  86. Cherian, S.; Paul, A.; Ahmed, S.; Alias, B.; Manoj, M.; Santhosh, A.K.; Varghese, D.R.; Krishnan, N.; Shenoy, P. Safety of the ChAdOx1 nCoV-19 and the BBV152 vaccines in 724 patients with rheumatic diseases: A post-vaccination cross-sectional survey. Rheumatol. Int. 2021, 41, 1441–1445. [Google Scholar] [CrossRef]
  87. Mehta, P.; Paul, A.; Ahmed, S.; Cherian, S.; Panthak, A.; Benny, J.; Shenoy, P. Effectiveness of delayed second dose of AZD1222 vaccine in patients with autoimmune rheumatic disease. Clin. Rheumatol. 2022, 41, 3537–3542. [Google Scholar] [CrossRef] [PubMed]
  88. Mohanasundaram, K.; Santhanam, S.; Natarajan, R.; Murugesan, H.; Nambi, T.; Chilikuri, B.; Nallasivan, S. Covid-19 vaccination in autoimmune rheumatic diseases: A multi-center survey from southern India. Int. J. Rheum. Dis. 2022, 25, 1046–1052. [Google Scholar] [CrossRef] [PubMed]
  89. Bakasis, A.D.; Mavragani, C.P.; Voulgari, P.V.; Gerolymatou, N.; Argyropoulou, O.D.; Vlachoyiannopoulos, P.G.; Skopuli, F.N.; Tzioufas, A.G.; Moutsopoulos, H.M. COVID-19: Clinical features and outcomes in unvaccinated 2-dose and 3-dose vaccinated against SARS-CoV-2 patients with systemic autoimmune and autoinflammatory rheumatic diseases. J. Autoimmun. 2022, 131, 102846. [Google Scholar] [CrossRef]
  90. Efe, C.; Taşçılar, K.; Gerussi, A.; Bolis, F.; Lammert, C.; Ebik, B.; Stättermayer, A.F.; Cengiz, M.; Gökçe, D.T.; Cristoferi, L.; et al. SARS-CoV-2 vaccination and risk of severe COVID-19 outcomes in patients with autoimmune hepatitis. J. Autoimmun. 2022, 132, 102906. [Google Scholar] [CrossRef]
  91. Kennedy, N.A.; Lin, S.; Goodhand, J.R.; Chanchlani, N.; Hamilton, B.; Bewshea, C.; Nice, R.; Chee, D.; Cummings, J.F.; Fraser, A.; et al. Infliximab is associated with attenuated immunogenicity to BNT162b2 and ChAdOx1 nCoV-19 SARS-CoV-2 vaccines in patients with IBD. Gut 2021, 70, 1884–1893. [Google Scholar] [CrossRef]
  92. Prendecki, M.; Clarke, C.; Edwards, H.; McIntyre, S.; Mortimer, P.; Gleeson, S.; Martin, P.; Thomson, T.; Randell, P.; Shah, A.; et al. Humoral and T-cell responses to SARS-CoV-2 vaccination in patients receiving immunosuppression. Ann. Rheum. Dis. 2021, 80, 1322–1329. [Google Scholar] [CrossRef]
  93. Shields, A.M.; Venkatachalam, S.; Shafeek, S.; Paneesha, S.; Ford, M.; Sheeran, T.; Kelly, M.; Qureshi, I.; Salhan, B.; Karim, F.; et al. SARS-CoV-2 vaccine responses following CD20-depletion treatment in patients with haematological and rheumatological disease: A West Midlands Research Consortium study. Clin. Exp. Immunol. 2022, 207, 3–10. [Google Scholar] [CrossRef] [PubMed]
  94. Machado, P.M.; Lawson-Tovey, S.; Strangfeld, A.; Mateus, E.F.; Hyrich, K.L.; Gossec, L.; Carmona, L.; Rodrigues, A.; Raffeiner, B.; Duarte, C.; et al. Safety of vaccination against SARS-CoV-2 in people with rheumatic and musculoskeletal diseases: Results from the EULAR Coronavirus Vaccine (COVAX) physician-reported registry. Ann. Rheum. Dis. 2022, 81, 695–709. [Google Scholar] [CrossRef]
  95. Kim, W.J.; Choi, S.H.; Park, J.Y.; Song, J.S.; Chung, J.W.; Choi, S.T. SARS-CoV-2 Omicron escapes mRNA vaccine booster-induced antibody neutralisation in patients with autoimmune rheumatic diseases: An observational cohort study. Ann. Rheum. Dis. 2022, 81, 1585–1593. [Google Scholar] [CrossRef] [PubMed]
  96. Lin, K.Y.; Hsieh, M.J.; Chang, S.Y.; Ieong, S.M.; Cheng, C.Y.; Sheng, W.H.; Chang, S. Serological response after COVID-19 mRNA-1273 booster dose in immunocompromised patients, Taiwan, July to August 2021. J. Formos. Med. Assoc. 2022, 121, 2438–2445. [Google Scholar] [CrossRef] [PubMed]
  97. Stalman, E.W.; Wieske, L.; van Dam, K.P.J.; Kummer, L.Y.; Kempen, Z.L.E.; Killestein, J.; Volkers, A.G.; Tas, S.W.; Boekel, L.; Wolbink, G.J.; et al. Breakthrough infections with the SARS-CoV-2 omicron (B.1.1.529) variant in patients with immune-mediated inflammatory diseases. Ann. Rheum. Dis. 2022, 81, 1757–1766. [Google Scholar] [CrossRef]
  98. Boekel, L.; Steenhuis, M.; Hooijberg, F.; Besten, Y.R.; van Kempen, Z.L.E.; Kummer, L.Y.; van Dam, K.P.J.; Stalman, E.W.; Vogelzang, E.H.; Cristianawati, O.; et al. Antibody development after COVID-19 vaccination in patients with autoimmune diseases in the Netherlands: A substudy of data from two prospective cohort studies. Lancet Rheumatol. 2021, 3, 778–788. [Google Scholar] [CrossRef]
  99. Szebeni, G.J.; Gémes, N.; Honfi, D.; Szabó, E.; Neuperger, P.; Balog, J.; Nagy, L.I.; Szekanecz, Z.; Puskás, L.G.; Toldi, G.; et al. Humoral and Cellular Immunogenicity and Safety of Five Different SARS-CoV-2 Vaccines in Patients with Autoimmune Rheumatic and Musculoskeletal Diseases in Remission or With Low Disease Activity and in Healthy Controls: A Single Center Study. Front. Immunol. 2022, 13, 846248. [Google Scholar] [CrossRef]
  100. Lee, E.J.; Beltrami-Moreira, M.; Al-Samkari, H.; Cuker, A.; Diraimo, J.; Gernsheimer, T.; Kruse, A.; Kessler, C.; Kruse, C.; Leavitt, A.D.; et al. SARS-CoV-2 vaccination and ITP in patients with de novo or preexisting ITP. Blood 2022, 139, 1564–1574. [Google Scholar] [CrossRef] [PubMed]
  101. De Santis, M.; Motta, F.; Isailovic, N.; Clementi, M.; Criscuolo, E.; Clementi, N.; Tonutti, A.; Rodolfi, S.; Barone, E.; Colapietro, F.; et al. Dose-Dependent Impairment of the Immune Response to the Moderna-1273 mRNA Vaccine by Mycophenolate Mofetil in Patients with Rheumatic and Autoimmune Liver Diseases. Vaccines 2022, 10, 801. [Google Scholar] [CrossRef]
  102. Lonati, P.A.; Bodio, C.; Scavone, M.; Martini, G.; Pesce, E.; Bandera, A.; Lombardi, A.; Gerosa, M.; Franceschini, F.; Tincani, A.; et al. Production of anti-PF4 antibodies in antiphospholipid antibody-positive patients is not affected by COVID-19 vaccination. RMD Open 2022, 8, 1902. [Google Scholar] [CrossRef]
  103. Ahmed, S.; Mehta, P.; Paul, A.; Anu, S.; Cherian, S.; Shenoy, V.; Nalianda, N.K.; Joseph, S.; Poulose, A.; Shenoy, P. Postvaccination antibody titres predict protection against COVID-19 in patients with autoimmune diseases: Survival analysis in a prospective cohort. Ann. Rheum. Dis. 2022, 81, 868–874. [Google Scholar] [CrossRef]
  104. Amanatidou, E.; Gkiouliava, A.; Fella, E.; Serafidi, M.; Tsilingiris, D.; Vallianou, N.G.; Karampela, I.; Dalamaga, M. Breakthrough infections after COVID-19 vaccination: Insights, perspectives and challenges. Metabol. Open 2022, 14, 100180. [Google Scholar] [CrossRef]
  105. Patel, N.J.; Wang, X.; Fu, X.; Kawano, Y.; Cook, C.; Vanni, K.M.M.; Qian, G.; Banasiak, E.; Kowalski, E.; Zhang, Y.; et al. Factors associated with COVID-19 breakthrough infection among vaccinated patients with rheumatic diseases: A cohort study. Semin. Arthritis Rheum. 2023, 58, 152108. [Google Scholar] [CrossRef]
  106. Paik, J.J.; Sparks, J.A.; Kim, A.H.J. Immunogenicity, breakthrough infection, and underlying disease flare after SARS-CoV-2 vaccination among individuals with systemic autoimmune rheumatic diseases. Curr. Opin. Pharmacol. 2022, 65, 102243. [Google Scholar] [CrossRef] [PubMed]
  107. So, H.; Li, T.; Chan, V.; Tam, L.S.; Chan, P.K.S. Immunogenicity and safety of inactivated and mRNA COVID-19 vaccines in patients with systemic lupus erythematosus. Ther. Adv. Musculoskelet. Dis. 2022, 14, 1759720X221089586. [Google Scholar] [CrossRef] [PubMed]
  108. Joudeh, A.I.; Lutf, A.Q.; Mahdi, S.; Tran, G. Efficacy and safety of mRNA and AstraZeneca COVID-19 vaccines in patients with autoimmune rheumatic diseases: A systematic review. Vaccine 2023, 41, 3812. [Google Scholar] [CrossRef] [PubMed]
  109. Cardelli, C.; Caruso, T.; Tani, C.; Pratesi, F.; Talarico, R.; Di Cianni, F.; Italiano, N.; Laurino, E.; Moretti, M.; Cascarano, G.; et al. COVID-19 mRNA vaccine booster in patients with autoimmune rheumatic diseases. Rheumatology 2022, 61, 328–330. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA flow chart.
Figure 1. PRISMA flow chart.
Vaccines 11 01456 g001
Figure 2. Breakthrough COVID-19 infections after receiving primary doses (two doses) of COVID-19 inactivated vaccine. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,77].
Figure 2. Breakthrough COVID-19 infections after receiving primary doses (two doses) of COVID-19 inactivated vaccine. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,77].
Vaccines 11 01456 g002
Figure 3. Breakthrough COVID-19 infections from studies using mRNA and adenovirus viral vector COVID 19 vaccines. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [97,98].
Figure 3. Breakthrough COVID-19 infections from studies using mRNA and adenovirus viral vector COVID 19 vaccines. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [97,98].
Vaccines 11 01456 g003
Figure 4. Log TAb titer after mRNA vaccination. Green squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [8,35,36,43,50,61,101].
Figure 4. Log TAb titer after mRNA vaccination. Green squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [8,35,36,43,50,61,101].
Vaccines 11 01456 g004
Figure 5. Log TAb titer after inactivated vaccination. Green squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,80,83].
Figure 5. Log TAb titer after inactivated vaccination. Green squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,80,83].
Vaccines 11 01456 g005
Figure 6. IgG seroconversion after mRNA vaccination. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [8,35,36,43,50,52,54,61,62,65,101].
Figure 6. IgG seroconversion after mRNA vaccination. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [8,35,36,43,50,52,54,61,62,65,101].
Vaccines 11 01456 g006
Figure 7. IgG seroconversion after inactivated vaccination. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,74,75,76,77,80,83].
Figure 7. IgG seroconversion after inactivated vaccination. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,74,75,76,77,80,83].
Vaccines 11 01456 g007
Figure 8. Proportion of neutralizing antibodies positive after mRNA vaccination. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [50,54,62].
Figure 8. Proportion of neutralizing antibodies positive after mRNA vaccination. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [50,54,62].
Vaccines 11 01456 g008
Figure 9. Proportion of neutralizing antibodies positive after inactivated vaccination. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,74,75,76,77,80,83].
Figure 9. Proportion of neutralizing antibodies positive after inactivated vaccination. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,74,75,76,77,80,83].
Vaccines 11 01456 g009
Figure 10. Neutralizing activity after inactivated vaccination. Green squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,77,80,83].
Figure 10. Neutralizing activity after inactivated vaccination. Green squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,77,80,83].
Vaccines 11 01456 g010
Figure 11. Local adverse events after receiving a first dose of COVID-19 inactivated vaccine. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,77].
Figure 11. Local adverse events after receiving a first dose of COVID-19 inactivated vaccine. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,77].
Vaccines 11 01456 g011
Figure 12. Local adverse events after receiving a second dose of COVID-19 inactivated vaccine. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,77].
Figure 12. Local adverse events after receiving a second dose of COVID-19 inactivated vaccine. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,77].
Vaccines 11 01456 g012
Figure 13. Systemic adverse events after receiving a first dose of COVID-19 inactivated vaccine. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,77].
Figure 13. Systemic adverse events after receiving a first dose of COVID-19 inactivated vaccine. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,77].
Vaccines 11 01456 g013
Figure 14. Systemic adverse events after receiving a second dose of COVID-19 inactivated vaccine. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,77].
Figure 14. Systemic adverse events after receiving a second dose of COVID-19 inactivated vaccine. Blue squares represent effect sizes for a single study, and black rhombus represent pooled results for all studies [72,75,76,77].
Vaccines 11 01456 g014
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Widhani, A.; Hasibuan, A.S.; Rismawati, R.; Maria, S.; Koesnoe, S.; Hermanadi, M.I.; Ophinni, Y.; Yamada, C.; Harimurti, K.; Sari, A.N.L.; et al. Efficacy, Immunogenicity, and Safety of COVID-19 Vaccines in Patients with Autoimmune Diseases: A Systematic Review and Meta-Analysis. Vaccines 2023, 11, 1456. https://doi.org/10.3390/vaccines11091456

AMA Style

Widhani A, Hasibuan AS, Rismawati R, Maria S, Koesnoe S, Hermanadi MI, Ophinni Y, Yamada C, Harimurti K, Sari ANL, et al. Efficacy, Immunogenicity, and Safety of COVID-19 Vaccines in Patients with Autoimmune Diseases: A Systematic Review and Meta-Analysis. Vaccines. 2023; 11(9):1456. https://doi.org/10.3390/vaccines11091456

Chicago/Turabian Style

Widhani, Alvina, Anshari Saifuddin Hasibuan, Retia Rismawati, Suzy Maria, Sukamto Koesnoe, Muhammad Ikrar Hermanadi, Youdiil Ophinni, Chika Yamada, Kuntjoro Harimurti, Aldean Nadhyia Laela Sari, and et al. 2023. "Efficacy, Immunogenicity, and Safety of COVID-19 Vaccines in Patients with Autoimmune Diseases: A Systematic Review and Meta-Analysis" Vaccines 11, no. 9: 1456. https://doi.org/10.3390/vaccines11091456

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop