Next Article in Journal
Insights into the Prognostic Role of Serum Interleukin-6 and Hematobiochemical Alterations in Cattle during Recent Outbreaks of Lumpy Skin Disease in Lodhran District, Pakistan
Previous Article in Journal
Vaccination with Formulation of Nanoparticles Loaded with Leishmania amazonensis Antigens Confers Protection against Experimental Visceral Leishmaniasis in Hamster
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Risks of Cardiac Arrhythmia Associated with COVID-19 Vaccination: A Systematic Review and Meta-Analysis

by
Mohammed H. Abutaleb
1,*,
Hafiz A. Makeen
2,
Abdulkarim M. Meraya
2,
Saad S. Alqahtani
2,
Bayan Ibrahim Al-Mass
3,
Reaam Omar Aljazaeri
4,
Bushra Dhuhayyan Alhazmi
5,
Afnan Mohammed Noor Kalakattawi
6 and
Ahmed Ali Alajam
7
1
Pharmaceutical Care Department, King Fahad Central Hospital, Jazan Health Affairs, Ministry of Health, Jazan 82912, Saudi Arabia
2
Pharmacy Practice Research Unit, Department of Pharmacy Practice, College of Pharmacy, Jazan University, Jazan 82912, Saudi Arabia
3
College of Clinical Pharmacy, King Faisal University, Alahsa 31982, Saudi Arabia
4
College of Pharmacy, Aljouf University, Sakaka 72311, Saudi Arabia
5
College of Clinical Pharmacy, Northern Border University, Arar 91431, Saudi Arabia
6
College of Clinical Pharmacy, Taif University, Taif 21974, Saudi Arabia
7
Pharmaceutical Care Department, Aldarb General Hospital, Jazan 89876, Saudi Arabia
*
Author to whom correspondence should be addressed.
Vaccines 2023, 11(1), 112; https://doi.org/10.3390/vaccines11010112
Submission received: 26 October 2022 / Revised: 12 December 2022 / Accepted: 17 December 2022 / Published: 3 January 2023
(This article belongs to the Section COVID-19 Vaccines and Vaccination)

Abstract

:
This systematic review and meta-analysis aimed to summarize the current evidence regarding the association between coronavirus disease 2019 (COVID-19) vaccination and the risk of cardiac arrhythmia. MEDLINE, via PubMed and OVID, Scopus, CENTRAL, and Web of Science were searched using the relevant keywords to identify the relevant citations. Comprehensive Meta-analysis and Review Manager 5.4.1 were used for all the statistical analyses. Seventeen studies (n = 567,033,087 patients) were included. The pooled analysis showed that the incidence of cardiac arrhythmia post-COVID-19 vaccination with Pfizer, Moderna, AstraZeneca, CoronaVac, and Sinopharm was 0.22%, 95% CI: (0.07% to 0.66%), 0.76%, 95% CI: (0.04% to 12.08%), 0.04%, 95% CI: (0.00% to 0.98%), 0.01%, 95% CI: (0.00% to 0.03%), and 0.03%, 95% CI: (0.00% to 18.48%), respectively. Compared to CoronaVac, Pfizer, Moderna, AstraZeneca, and Sinopharm had a higher incidence ratio rate (IRR; 22-times, 76-times, 4-times, and 3-times higher), respectively. Likewise, Pfizer, Moderna, and AstraZeneca showed a higher IRR than Sinopharm (7.3-times, 25.3-times, and 1.3-times higher). The current evidence shows that the incidence rate (IR) of cardiac arrhythmia post-COVID-19 vaccination is rare and ranges between 1 and 76 per 10,000. mRNA vaccines were associated with a higher IR of arrhythmia compared to vector-based vaccines. Inactivated vaccines showed the lowest IR of arrhythmia.

1. Introduction

Different treatment options have been investigated since the inception of the coronavirus disease 2019 (COVID-19) pandemic in late 2019, including immunotherapy, nucleoside analogs, and others [1,2]. To prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or to lessen the severity of the disease, with the ultimate aim of reducing mortality and morbidity, researchers have investigated a variety of platforms for vaccine development, including protein-based and viral-based vaccines [3,4].
There were 128 vaccines under clinical development as of 22 October 2021, with another 194 in the pre-clinical stage [5]. B-cell response, helper T cells, and cytotoxic cells are prompted by COVID-19 mRNA vaccines such as BNT1626b2 (i.e., Pfizer/BioNTech vaccine) and mRNA1273 (i.e., Moderna vaccine) [6]. Regarding vector vaccination, the COVID-19 virus’s genetic material is inserted into a modified form of another virus, which facilitates the delivery of genetic material from the COVID-19 virus that induces copies of the S protein [7]. The production of antibodies and protective white blood cells is triggered once S proteins are presented on the surface of the body cells. The Johnson & Johnson (Ad26.COV2.S) and Oxford and AstraZeneca (ChAdOx1) COVID-19 vaccines are vectors [8]. Protein subunit vaccines use fragments of protein from the disease-causing virus to trigger protective immunity against it. Studies have revealed that these protein vaccines include S proteins that are not pathogenic. Once the S proteins are recognized by the immune system, antibodies and protective white blood cells are generated [9]. The Novavax COVID-19 vaccine (NVX-CoV2373) is one of these protein-based vaccines. Since the approval of these vaccines, regardless of their type, several reports worldwide have emerged, documenting various adverse events, including local and systemic reactions.
Myalgia, headache, fever, edema, erythema, and pain at the injection site were the most common local or systemic reactions observed [10,11,12]. However, a number of serious and potentially fatal adverse events and complications have been documented in association with COVID-19 vaccinations, including venous thromboembolism, stroke, pulmonary embolism, cardiac arrhythmias, and myocardial infarction [13,14,15]. The unfavorable cardiac outcomes linked to COVID-19 vaccinations can range from inflammations (such as pericarditis or myocarditis) to thrombosis and ischemia [15,16]. Several systematic reviews and meta-analyses have investigated the risk of myocarditis/pericarditis, thrombocytopenia, and thrombosis in vaccinated COVID-19 individuals [17,18,19,20,21]. However, to the best of our knowledge, there is no large-scale study or meta-analysis that investigates the association between COVID-19 vaccination and cardiac arrhythmia. Therefore, in this systematic review and meta-analysis, we aimed to summarize the current evidence regarding the association between COVID-19 vaccination and the risk of arrhythmia.

2. Methods

The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) checklist and Cochrane handbook for systematic reviews of interventions were followed in the reporting of this study [22,23]. The study protocol (CRD42022371085) was registered on PROSPERO.

2.1. Eligibility Criteria

The studies were included based on the following criteria:
Population: studies that were conducted on the individuals who received any type or dose of the COVID-19 vaccine.
Intervention and comparison: Pfizer-BioNTech, AstraZeneca, CoronaVac, Janssen, Sinopharm, or Moderna vaccines.
Outcomes: studies reported cardiac arrhythmia as an adverse event of one of the aforementioned vaccines.
Studies design: observational studies (case–control, cohort, and cross-sectional).
We excluded case reports, conference abstracts, and non-English studies.

2.2. Information Sources and Search Strategy

On 22 September 2022, we searched the following databases: MEDLINE via PubMed and OVID, Scopus, CENTRAL, and Web of Science, using the relevant keywords to identify the relevant citations. The MeSH keywords used were “Cardiac Arrhythmias, COVID-19, COVID-19 Vaccines, 2019-nCoV Vaccine mRNA-1273, ChAdOx1 nCoV-19, BIBP COVID-19 vaccine, BNT162 Vaccine, Ad26COVS1, sinovac COVID-19 vaccine, CoronaVac vaccine, and BIBP COVID-19 vaccine”. These databases were searched from inception to the date of search. Moreover, the reference lists of all the included citations were searched. The retrieved citations were imported to EndNote X9 software, and duplications were removed.

2.3. Selection Process

Using Microsoft Excel software, a screening sheet was created. Study ID, publication year, title, abstract, keywords, DOI, and URL were all included. The selection process was undertaken using a two-step screening technique by three independent reviewers (B.I.A, R.O.A, and B.D.A). Step one included screening the title and abstract of all the studies found via the literature search to determine which studies might proceed to step two (full-text screening), where reviewers would read and assess whether each study met the eligibility criteria. Any disagreement between the reviewers was resolved by the judgment of the study supervisor (M.A).

2.4. Data Items and Collection Process

Four independent reviewers extracted the following data from the included studies to an offline pre-prepared Excel sheet: demographic data of the included patients (age, gender, and residency), study characteristics (study groups, study duration, total sample size, country, and main findings) and outcomes (incidence rate of arrhythmia following COVID-19 vaccination).

2.5. Risk of Bias and Quality Assessment

Using the National Institutes of Health (NIH) quality assessment tool for observational cohort, case–control, and cross-sectional studies, two authors (A.M.N and A.A.A) independently evaluated the risk of bias and the quality of each included article. Reviewers can critically evaluate the internal validity of research using this tool. Studies were deemed to be “good,” “fair,” or “poor”. When the authors disagreed on a rating, a third author (M.A) resolved any disagreements.

2.6. Data Synthesis

The incidence of adverse events was calculated using the random-effects model with a 95% CI. Using the I2 statistic, we calculated the percentage of heterogeneity and inconsistency between studies, with values of 25%, 50%, and 75% deemed as low, moderate, and high, respectively. The random-effect model was employed if the heterogeneity was considerable and if I2 > 50%; otherwise, the fixed-effect model was utilized. Comprehensive Meta-analysis software was used for all the statistical analyses (Biostat, Inc., CMA; USA: version 3.3.070, Florida, 33618, United States). To assess the difference between Pfizer-BioNTech and AstraZeneca in terms of AEs, we used the Review Manager 5.4 software to calculate the risk ratio (RR) between both groups using the Mantel–Hansel model.

3. Results

3.1. Study Selection

Based on our literature search, we found a total of 980 relevant citations. After removing the duplications, 720 articles underwent title/abstract screening. Then, 690 studies were deemed to be ineligible using our criteria. Full-text screening was performed on 30 articles, and only 17 studies (n = 567,033,087 patients) were included in the qualitative (systematic review) and quantitative synthesis (meta-analysis) [24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40]. Figure 1 shows the PRISMA flow diagram of the included studies.

3.2. Characteristics of Included Studies and Patients

Three studies were cohort studies, three were cross-sectional, eight were retrospective studies, two were controlled case-series studies, and one was a subgroup analysis study. Four studies were multicentric studies, three were from the USA, three were from Israel, one was from Malaysia, one was from Saudi Arabia, one was from Singapore, one was from Turkey, one was from India, one was from China, and one was from England. The Pfizer-BioNTech vaccine was reported in 12 studies, AstraZeneca in 3, CoronaVac in 3, and Moderna in 7. The age of the included patients ranged between 12 and 90 years old. Both males and females were represented in the included studies. Table 1 summarizes the baseline characteristics of the included studies and patients.

3.3. Quality of the Included Studies

Based on the NIH quality assessment tool for observational studies, about 88.2% of the studies were deemed as “Good,” and 11.8% were deemed as “Fair”.

3.4. Meta-Analysis

3.4.1. Incidence of Cardiac Arrhythmia

The pooled analysis of 12 studies showed that the incidence of cardiac arrhythmia post-Pfizer COVID-19 vaccine was 0.22%, 95% CI: (0.07% to 0.66%), which means that the incidence rate was 22 per 10,000 persons (Table 2). The random-effect estimate of four studies demonstrated that the pooled incidence of arrhythmia post-Moderna COVID-19 vaccine was 0.76%, 95% CI: (0.04% to 12.08%). Compared to Pfizer, the Moderna vaccine had an incidence rate ratio (IRR) of 3.45, meaning that the group that received the Moderna vaccine had a 3.45-times higher risk of developing cardiac arrhythmia than Pfizer. This finding was supported by the lower RR of cardiac arrhythmia associated with the Pfizer vaccine compared to the Moderna vaccine; however, the difference was not statistically significant (RR = 0.60, 95% CI: 0.28 to 1.29; Figure 2).
The pooled analysis of three studies showed that the incidence of arrhythmia post-AstraZeneca vaccine was 0.04%, 95% CI: (0.00% to 0.98%). The IRR arrhythmia post-Pfizer or Moderna COVID-19 vaccines was 5.5-times and 19-times higher than the IRR of arrhythmia post-AstraZeneca vaccine. Similarly, the pooled RR showed that the Pfizer vaccine had a non-significantly higher RR compared to AstraZeneca (RR = 1.63, 95% CI: 0.77 to 3.46; Figure 3). On the other hand, AstraZeneca and Moderna had a comparable RR (1.28, 95% CI: 0.03 to 48.96; Figure 4).
The random-effect model showed that the incidence of arrhythmia post-CoronaVac and Sinopharm vaccines was 0.01%, 95% CI: (0.00% to 0.03%) and 0.03%, 95% CI: (0.00% to 18.48%), respectively. Compared to CoronaVac, Pfizer, Moderna, AstraZeneca, and Sinopharm had a higher IRR (22-times, 76-times, 4-times, and 3-times higher), respectively. Likewise, Pfizer, Moderna, and AstraZeneca showed a higher IRR than Sinopharm (7.3-times, 25.3-times, and 1.3-times higher). The head-to-head comparison showed that there was no significant difference between the Pfizer and CoronaVac vaccines in terms of RR of arrhythmia (RR = 1.15, 95% CI: 0.43 to 3.05; Figure 5).

3.4.2. Subgroup Analysis

The subgroup analysis according to the study design, sample size, and study setting is presented in Table 3. People from North America had the highest incidence of cardiac arrhythmia post-Pfizer COVID-19 vaccination at 11.95%, 95% CI: (4.92% to 18.98%), followed by Europe at 0.136%, 95% CI: (0.134% to 0.137%), the Middle East at 0.022%, 95% CI: (0.001% to 0.046%), and Asia at 0.006%, 95% CI: (0% to 0.009%). Studies that included a sample size larger than 2000 individuals reported a much lower incidence of cardiac arrhythmia post-Pfizer vaccine than those who included less than 2000. Regarding the study design, cross-sectional studies reported the highest incidence of cardiac arrhythmias compared to cohort studies, case-series studies, and retrospective studies.

4. Discussion

The current systematic review and meta-analysis showed that the IR of cardiac arrhythmia post-COVID-19 vaccination is rare and ranges between 1 and 76 per 10,000. mRNA vaccines, including Moderna and Pfizer, were associated with a higher IR of arrhythmia than vector-based vaccines. On the other hand, inactivated vaccines, such as CoronaVac and Sinopharm, showed the lowest IR of arrhythmia. Due to the absence of head-to-head comparison in the majority of the included studies and the non-significant difference in the RR between the studied vaccines, further studies are required to validate these findings. We did not find enough data about the risk of arrhythmia associated with the Johnson & Johnson vaccine to be pooled in one meta-analysis. However, the results of individual studies demonstrated that arrhythmia was one of the largest relative risks associated with COVID-19 vaccines, besides thromboembolic events, sexual organ reactions, ocular, gastrointestinal, constitutional, hemorrhage, coagulation, and other cardiovascular events. However, arrhythmia was not reported among the ten most frequent cardiovascular events attributed to the Moderna, Pfizer, or Johnson & Johnson vaccines. On the other hand, they showed that the Johnson & Johnson vaccine was associated with the lowest RR of arrhythmia when compared with Moderna and Pfizer (RR = 17.77, 95% CI: 15.48 to 20.39) vs. (RR = 56.87, 95% CI: 49.77 to 64.99 and RR= 70.80, 95% CI: 61.98 to 80.88), respectively [24]. Our findings were limited by the significant heterogeneity between the included studies. There were several causes for this heterogeneity, including the different population characteristics, study setting, study design, and administered vaccine doses. We found that North American studies had the highest incidence of arrhythmia post-Pfizer vaccine, compared to the Asian or Middle Eastern studies. This finding was consistent with the global reports that showed that the American population is more susceptible to cardiac arrhythmia compared to the European and Asian populations [41]. Moreover, we found that the self-reported incidences of cardiac arrhythmia were much higher than those based on clinician diagnosis.
Regarding the comparison between the Pfizer COVID-19 vaccine and other vaccines, AbRahman et al. conducted a controlled case-series study, comparing CoronaVac, Pfizer, and AstraZeneca. Their findings showed that among the patients who developed new-onset arrhythmia, 60.7% received the Pfizer COVID-19 vaccine, 35.3% received CoronaVac, 3.9% received AstraZeneca, and 0.2% received other vaccines. The event rate of arrhythmia per 1 million vaccinated persons was 95.47, 51.76, and 26.92, following Pfizer, CoronaVac, and AstraZeneca vaccines, respectively. However, the IRR results showed that those who received CoronaVac had the lowest IRR of arrhythmia compared to Pfizer and AstraZeneca (1.15, 95% CI: (0.96 to 1.17) vs. 1.16, 95% CI: (1.07 to 1.26) and 1.36, 95% CI: (0.95 to 1.94), respectively). Most of these patients were over 60 years old (66.5%). About 55.1% of the patients received the first dose of the vaccine, and 44.9% experienced arrhythmia after the second dose. Almost half of the patients already had hypertension, 16% had diabetes, and 12% had other heart diseases. They concluded that they could not highlight a significant association between cardiac arrhythmia and COVID-19 vaccines; however, arrhythmia is an AESI of concern [39]. According to Wong et al., there was no significant difference between CoronaVac and Pfizer in terms of the IRR of arrhythmia, CAD, or MI [37]. Compared to Moderna, the Pfizer vaccine induced a higher risk of arrhythmia (7.5 per 10,000 persons, 95% CI: 1.9 to 11.5 events) [27].
Although the numbers may not be indicative of the true situation, given that the Pfizer-BioNTech COVID-19 vaccine was the first to receive emergency approval from the US-FDA, and thus a large number of doses have been administered up until the period of analysis, one possible explanation is a shorter interval between the two doses, as shown by Kaur et al. [32]. The Case Series Drug Analysis Print published by Pfizer-BioNTech for their COVID-19 vaccine as of May 28, 2021, reported a total of 2342 cardiac AEs, among which 1098 events were classified as palpitations, 16 events were classified as acute myocardial infarction, 38 events as arrhythmia, 24 events as cardiac failure, 46 events as angina pectoris, 32 events as sinus tachycardia, 63 events as cardiac flutter, 62 events as cardiac arrest, 108 events as atrial fibrillation, and 466 events as tachycardia [42]. The rhythm disorders, as reported by AstraZeneca for their COVID-19 vaccine, included 1763 events of palpitations, 6 events of unstable angina, 8 events of supraventricular tachycardia, 8 events of tachyarrhythmia, 21 events of extrasystoles, 34 events of sinus tachycardia, 43 events of arrhythmia, 78 events of atrial fibrillation, and 622 events of tachycardia [43].
In terms of ECG changes associated with COVID-19 vaccines, Truong et al. enrolled 139 adolescents and young adults with 140 episodes of suspected myocarditis who received Pfizer, Moderna, and Janssen vaccines. Their findings showed that 97 patients presented with ECG changes and arrhythmias, including ST- or T-wave changes/elevation (97.9%), complete heart block (0.7%), first-degree atrioventricular block (0.7%), premature atrial contractions (0.7%), atrial tachycardia (0.7%), low-voltage QRS (3.6%), and non-sustained ventricular tachycardia (5%) [35].
When compared with non-vaccinated patients, the Pfizer vaccine was associated with non-statistically significant lower RR of arrhythmia (RR = 0.89, 95% CI: 0.74 to 1.04), according to Barda et al. They also demonstrated that vaccination against SARA-CoV-2 was associated with substantially lower RR of arrhythmia compared to infected patients, who showed a significantly high RR of arrhythmia (RR = 3.83, 95% CI: 3.07 to 4.95; 166.1 events per 100,000 persons) [40].
This is the first systematic review and meta-analysis that investigate the incidence rate of arrhythmia following COVID-19 vaccines. However, we acknowledge that our study has some limitations, including the small number of included studies, the significant heterogeneity, the inability to conduct a subgroup analysis for all vaccines, and the lack of head-to-head comparison in the majority of the included studies.
In conclusion, the current evidence shows that the IR of cardiac arrhythmia post-COVID-19 vaccination is rare and ranges between 1 and 76 per 10,000. mRNA vaccines, including Moderna and Pfizer, were associated with a higher IR of arrhythmia compared to vector-based vaccines. On the other hand, inactivated vaccines, such as CoronaVac and Sinopharm, showed the lowest IR of arrhythmia. Due to the absence of head-to-head comparison in the majority of the included studies and the non-significant difference in the RR between the studied vaccines, further studies are required to validate these findings.

Author Contributions

Conceptualization, M.H.A. and A.A.A.; methodology, M.H.A.; validation, B.I.A.-M. and R.O.A.; formal analysis, H.A.M., B.D.A. and A.M.N.K.; data curation, H.A.M., A.M.M. and S.S.A.; writing—original draft preparation, H.A.M., A.M.M., S.S.A., B.I.A.-M. and R.O.A.; writing—review and editing, B.D.A., A.M.N.K., A.A.A. and M.H.A.; visualization, H.A.M. and R.O.A.; supervision, M.H.A., H.A.M. and A.M.M.; project administration, M.H.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Han, F.; Liu, Y.; Mo, M.; Chen, J.; Wang, C.; Yang, Y.; Wu, J. Current Treatment Strategies for COVID-19 (Review). Mol. Med. Rep. 2021, 24, 858. [Google Scholar] [CrossRef] [PubMed]
  2. Baral, P.K.; Yin, J.; James, M.N.G. Treatment and Prevention Strategies for the COVID 19 Pandemic: A Review of Immunotherapeutic Approaches for Neutralizing SARS-CoV-2. Int. J. Biol. Macromol. 2021, 186, 490–500. [Google Scholar] [CrossRef] [PubMed]
  3. Huang, H.-Y.; Wang, S.-H.; Tang, Y.; Sheng, W.; Zuo, C.-J.; Wu, D.-W.; Fang, H.; Du, Q.; Li, N. Landscape and Progress of Global COVID-19 Vaccine Development. Hum. Vaccin. Immunother. 2021, 17, 3276–3280. [Google Scholar] [CrossRef] [PubMed]
  4. Ebada, M.A.; Allah, A.W.; Bahbah, E.; Negida, A. An Updated Review on COVID-19. Infect. Disord. Drug Targets 2021, 21, e160921189190. [Google Scholar] [CrossRef]
  5. COVID-19 Vaccine Tracker and Landscape. Available online: https://www.who.int/publications/m/item/draft-landscape-of-covid-19-candidate-vaccines (accessed on 12 August 2022).
  6. Verbeke, R.; Lentacker, I.; De Smedt, S.C.; Dewitte, H. The Dawn of MRNA Vaccines: The COVID-19 Case. J. Control. Release 2021, 333, 511–520. [Google Scholar] [CrossRef]
  7. Tregoning, J.S.; Brown, E.S.; Cheeseman, H.M.; Flight, K.E.; Higham, S.L.; Lemm, N.-M.; Pierce, B.F.; Stirling, D.C.; Wang, Z.; Pollock, K.M. Vaccines for COVID-19. Clin. Exp. Immunol. 2020, 202, 162–192. [Google Scholar] [CrossRef]
  8. Kyriakidis, N.C.; López-Cortés, A.; González, E.V.; Grimaldos, A.B.; Prado, E.O. SARS-CoV-2 Vaccines Strategies: A Comprehensive Review of Phase 3 Candidates. npj Vaccines 2021, 6, 28. [Google Scholar] [CrossRef]
  9. Heinz, F.X.; Stiasny, K. Distinguishing Features of Current COVID-19 Vaccines: Knowns and Unknowns of Antigen Presentation and Modes of Action. npj Vaccines 2021, 6, 104. [Google Scholar] [CrossRef]
  10. Polack, F.P.; Thomas, S.J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J.L.; Pérez Marc, G.; Moreira, E.D.; Zerbini, C.; et al. Safety and Efficacy of the BNT162b2 MRNA Covid-19 Vaccine. N. Engl. J. Med. 2020, 383, 2603–2615. [Google Scholar] [CrossRef]
  11. Omran, D.; Al Soda, M.; Bahbah, E.; Esmat, G.; Shousha, H.; Elgebaly, A.; Abdel Ghaffar, M.; Alsheikh, M.; El Sayed, E.; Afify, S.; et al. Predictors of Severity and Development of Critical Illness of Egyptian COVID-19 Patients: A Multicenter Study. PLoS ONE 2021, 16, e0256203. [Google Scholar] [CrossRef]
  12. AbdelGhaffar, M.M.; Omran, D.; Elgebaly, A.; Bahbah, E.I.; Afify, S.; AlSoda, M.; El-Shiekh, M.; ElSayed, E.S.; Shaaban, S.S.; AbdelHafez, S.; et al. Prediction of Mortality in Hospitalized Egyptian Patients with Coronavirus Disease-2019: A Multicenter Retrospective Study. PLoS ONE 2022, 17, e0262348. [Google Scholar] [CrossRef] [PubMed]
  13. García-Grimshaw, M.; Ceballos-Liceaga, S.E.; Hernández-Vanegas, L.E.; Núñez, I.; Hernández-Valdivia, N.; Carrillo-García, D.A.; Michel-Chávez, A.; Galnares-Olalde, J.A.; Carbajal-Sandoval, G.; Del Mar Saniger-Alba, M.; et al. Neurologic Adverse Events among 704,003 First-Dose Recipients of the BNT162b2 MRNA COVID-19 Vaccine in Mexico: A Nationwide Descriptive Study. Clin. Immunol. 2021, 229, 108786. [Google Scholar] [CrossRef] [PubMed]
  14. Bozkurt, B.; Kamat, I.; Hotez, P.J. Myocarditis With COVID-19 MRNA Vaccines. Circulation 2021, 144, 471–484. [Google Scholar] [CrossRef] [PubMed]
  15. Klein, N.P.; Lewis, N.; Goddard, K.; Fireman, B.; Zerbo, O.; Hanson, K.E.; Donahue, J.G.; Kharbanda, E.O.; Naleway, A.; Nelson, J.C.; et al. Surveillance for Adverse Events After COVID-19 MRNA Vaccination. JAMA 2021, 326, 1390–1399. [Google Scholar] [CrossRef] [PubMed]
  16. Abdelgawad, H.A.H.; Sayed, A.; Munir, M.; Elberry, M.H.; Sayed, I.M.; Kamal, M.A.; Negida, A.; Ebada, M.A.; Bahbah, E.I. Clinical Review of COVID-19; Pathogenesis, Diagnosis, and Management. Curr. Pharm. Des. 2021, 27, 4232–4244. [Google Scholar] [CrossRef]
  17. Matar, R.H.; Mansour, R.; Nakanishi, H.; Smayra, K.; El Haddad, J.; Vankayalapati, D.K.; Daniel, R.S.; Tosovic, D.; Than, C.A.; Yamani, M.H. Clinical Characteristics of Patients with Myocarditis Following COVID-19 MRNA Vaccination: A Systematic Review and Meta-Analysis. J. Clin. Med. 2022, 11, 4521. [Google Scholar] [CrossRef]
  18. Chou, O.H.I.; Mui, J.; Chung, C.T.; Radford, D.; Ranjithkumar, S.; Evbayekha, E.; Nam, R.; Pay, L.; Satti, D.I.; Garcia-Zamora, S.; et al. COVID-19 Vaccination and Carditis in Children and Adolescents: A Systematic Review and Meta-Analysis. Clin. Res. Cardiol. 2022, 111, 1161–1173. [Google Scholar] [CrossRef]
  19. Parmar, K.; Subramanyam, S.; Del Rio-Pertuz, G.; Sethi, P.; Argueta-Sosa, E. Cardiac Adverse Events after Vaccination-A Systematic Review. Vaccines 2022, 10, 700. [Google Scholar] [CrossRef]
  20. Samimisedeh, P.; Jafari Afshar, E.; Shafiabadi Hassani, N.; Rastad, H. Cardiac MRI Findings in COVID-19 Vaccine-Related Myocarditis: A Pooled Analysis of 468 Patients. J. Magn. Reson. Imaging 2022, 56, 971–982. [Google Scholar] [CrossRef]
  21. Ling, R.R.; Ramanathan, K.; Tan, F.L.; Tai, B.C.; Somani, J.; Fisher, D.; MacLaren, G. Myopericarditis Following COVID-19 Vaccination and Non-COVID-19 Vaccination: A Systematic Review and Meta-Analysis. Lancet. Respir. Med. 2022, 10, 679–688. [Google Scholar] [CrossRef]
  22. Higgins, J.P.; Green, S. Cochrane Handbook for Systematic Reviews of Interventions, version 5; Wiley online library: Hoboken, NJ, USA, 2008; ISBN 9780470699515. [Google Scholar]
  23. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 Statement: An Updated Guideline for Reporting Systematic Reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
  24. Montano, D. Frequency and Associations of Adverse Reactions of COVID-19 Vaccines Reported to Pharmacovigilance Systems in the European Union and the United States. Front. Public Health 2022, 9, 756633. [Google Scholar] [CrossRef] [PubMed]
  25. Patone, M.; Mei, X.W.; Handunnetthi, L.; Dixon, S.; Zaccardi, F.; Shankar-Hari, M.; Watkinson, P.; Khunti, K.; Harnden, A.; Coupland, C.A.C.; et al. Risks of Myocarditis, Pericarditis, and Cardiac Arrhythmias Associated with COVID-19 Vaccination or SARS-CoV-2 Infection. Nat. Med. 2022, 28, 410–422. [Google Scholar] [CrossRef] [PubMed]
  26. Dagan, N.; Barda, N.; Balicer, R. Adverse Effects after BNT162b2 Vaccine and SARS-CoV-2 Infection, According to Age and Sex. N. Engl. J. Med. 2021, 27, 2115045. [Google Scholar] [CrossRef]
  27. Dickerman, B.A.; Madenci, A.L.; Gerlovin, H.; Kurgansky, K.E.; Wise, J.K.; Figueroa Muñiz, M.J.; Ferolito, B.R.; Gagnon, D.R.; Gaziano, J.M.; Cho, K.; et al. Comparative Safety of BNT162b2 and MRNA-1273 Vaccines in a Nationwide Cohort of US Veterans. JAMA Intern. Med. 2022, 182, 739. [Google Scholar] [CrossRef] [PubMed]
  28. El-shitany, N.A.; Bagher, A.M.; Binmahfouz, L.S.; Eid, B.G.; Almukadi, H.; Badr-eldin, S.M.; El-hamamsy, M.; Mohammedsaleh, Z.M.; Saleh, F.M.; Almuhayawi, M.S.; et al. The Adverse Reactions of Pfizer BioNTech COVID-19 Vaccine Booster Dose Are Mild and Similar to the Second Dose Responses: A Retrospective Cross-Sectional Study. Int. J. Gen. Med. 2022, 15, 6821–6836. [Google Scholar] [CrossRef]
  29. Kadali, R.A.K.; Janagama, R.; Peruru, S.; Malayala, S.V. Side Effects of BNT162b2 MRNA COVID-19 Vaccine: A Randomized, Cross-Sectional Study with Detailed Self-Reported Symptoms from Healthcare Workers. Int. J. Infect. Dis. 2021, 106, 376–381. [Google Scholar] [CrossRef]
  30. Kadali, R.A.K.; Janagama, R.; Peruru, S.; Gajula, V.; Madathala, R.R.; Chennaiahgari, N.; Malayala, S.V. Non-life-threatening Adverse Effects with COVID-19 MRNA-1273 Vaccine: A Randomized, Cross-sectional Study on Healthcare Workers with Detailed Self-reported Symptoms. J. Med. Virol. 2021, 93, 4420–4429. [Google Scholar] [CrossRef]
  31. Kaur, U.; Ojha, B.; Kumar, B.; Singh, A.; Giri, K.R.; Singh, A.; Das, A.; Misra, A.; Kumar, A.; Kansal, S.; et al. EClinicalMedicine A Prospective Observational Safety Study on ChAdOx1 NCoV-19 Corona Virus Vaccine (Recombinant) Use in Healthcare Workers- Fi Rst Results from India. EClinicalMedicine 2021, 38, 101038. [Google Scholar] [CrossRef]
  32. Kaur, R.; Dutta, S.; Charan, J.; Bhardwaj, P.; Tandon, A.; Yadav, D.; Islam, S.; Haque, M. Cardiovascular Adverse Events Reported from COVID-19 Vaccines: A Study Based on WHO Database. Int. J. Gen. Med. 2021, 14, 3909–3927. [Google Scholar] [CrossRef]
  33. Tan, J.T.C.; Tan, C.; Teoh, J.; Mt, W.; Zhong, G.; Mbbs, T.; Yew, R.; Chin, Z.; Lee, A.; Mutalib, A.; et al. Adverse Reactions and Safety Profile of the MRNA COVID-19 Vaccines among Asian Military Personnel. Ann. Acad. Med. Singap. 2021, 50, 827–837. [Google Scholar] [CrossRef] [PubMed]
  34. Tobaiqy, M.; Maclure, K.; Elkout, H. Thrombotic Adverse Events Reported for Moderna, Pfizer and Oxford-AstraZeneca COVID-19 Vaccines: Comparison of Occurrence and Clinical Outcomes in the EudraVigilance Database. Vaccines 2021, 9, 1326. [Google Scholar] [CrossRef] [PubMed]
  35. Truong, D.T.; Dionne, A.; Muniz, J.C.; McHugh, K.E.; Portman, M.A.; Lambert, L.M.; Thacker, D.; Elias, M.D.; Li, J.S.; Toro-Salazar, O.H.; et al. Clinically Suspected Myocarditis Temporally Related to COVID-19 Vaccination in Adolescents and Young Adults: Suspected Myocarditis After COVID-19 Vaccination. Circulation 2022, 145, 345–356. [Google Scholar] [CrossRef] [PubMed]
  36. Witberg, G.; Barda, N.; Hoss, S.; Richter, I.; Wiessman, M.; Aviv, Y.; Grinberg, T.; Auster, O.; Dagan, N.; Balicer, R.D.; et al. Myocarditis after Covid-19 Vaccination in a Large Health Care Organization. N. Engl. J. Med. 2021, 385, 2132–2139. [Google Scholar] [CrossRef] [PubMed]
  37. Wong, C.K.H.; Lau, K.T.K.; Xiong, X.; Au, I.C.H.; Lai, F.T.T.; Wan, E.Y.F.; Chui, C.S.L.; Li, X.; Chan, E.W.Y.; Gao, L.; et al. Adverse Events of Special Interest and Mortality Following Vaccination with MRNA (BNT162b2) and Inactivated (CoronaVac) SARS-CoV-2 Vaccines in Hong Kong: A Retrospective Study. PLOS Med. 2022, 19, e1004018. [Google Scholar] [CrossRef]
  38. Dizbay, M.; Şenol, E. Evaluation of the Frequency of Side Effects after Coronovirus Inactive Vero Cell Vaccine in Healthcare Workers of Gazi University Hospital. Gazi Med. J. 2021, 32, 651–654. [Google Scholar]
  39. Ab Rahman, N.; Tsuey, M.; Yee, F.; Chet, S.; Ramli, A. Risk of Serious Adverse Events after the BNT162b2, CoronaVac, and ChAdOx1 Vaccines in Malaysia: A Self-Controlled Case Series Study. Vaccine 2022, 40, 4394–4402. [Google Scholar] [CrossRef]
  40. Barda, N.; Dagan, N.; Ben-Shlomo, Y.; Kepten, E.; Waxman, J.; Ohana, R.; Hernán, M.A.; Lipsitch, M.; Kohane, I.; Netzer, D.; et al. Safety of the BNT162b2 MRNA Covid-19 Vaccine in a Nationwide Setting. N. Engl. J. Med. 2021, 385, 1078–1090. [Google Scholar] [CrossRef]
  41. Dai, H.; Zhang, Q.; Much, A.A.; Maor, E.; Segev, A.; Beinart, R.; Adawi, S.; Lu, Y.; Bragazzi, N.L.; Wu, J. Global, Regional, and National Prevalence, Incidence, Mortality, and Risk Factors for Atrial Fibrillation, 1990–2017: Results from the Global Burden of Disease Study 2017. Eur. Heart J.—Qual. Care Clin. Outcomes 2021, 7, 574–582. [Google Scholar] [CrossRef]
  42. Publishing Service. Government of UK COVID-19 MRNA Pfizer- BioNTech Vaccine Analysis Print. Available online: https://assets.publishing.service.gov.uk/government/uploads/system/uploads/attachment_data/file/989996/COVID-19_mRNA_Pfizer-_BioNTech_Vaccine_Analysis_Print.pdf (accessed on 20 July 2022).
  43. AstraZeneca COVID-19 Vaccine AstraZeneca Analysis Print; 30 May 2021. Available online: https://assets.publishing.service.gov.uk/government/uploads/system/uploads/attachment_data/file/1121352/COVID-19_Vaccine_AstraZeneca_VAP.pdf (accessed on 12 August 2022).
Figure 1. PRISMA flow diagram.
Figure 1. PRISMA flow diagram.
Vaccines 11 00112 g001
Figure 2. The RR of arrhythmia post-Pfizer COVID-19 vaccine compared to Moderna.
Figure 2. The RR of arrhythmia post-Pfizer COVID-19 vaccine compared to Moderna.
Vaccines 11 00112 g002
Figure 3. The RR of arrhythmia post-Pfizer COVID-19 vaccine compared to AstraZeneca.
Figure 3. The RR of arrhythmia post-Pfizer COVID-19 vaccine compared to AstraZeneca.
Vaccines 11 00112 g003
Figure 4. The RR of arrhythmia post-AstraZeneca COVID-19 vaccine compared to Moderna.
Figure 4. The RR of arrhythmia post-AstraZeneca COVID-19 vaccine compared to Moderna.
Vaccines 11 00112 g004
Figure 5. The RR of arrhythmia post-Pfizer COVID-19 vaccine compared to CoronaVac.
Figure 5. The RR of arrhythmia post-Pfizer COVID-19 vaccine compared to CoronaVac.
Vaccines 11 00112 g005
Table 1. Summary of included studies.
Table 1. Summary of included studies.
Study IDCountry Study DesignGroupsNumber of Each GroupSample SizeAgeMale, n (%)Any Other Heart DiseasesHypertensionCoronary Artery DiseaseLeft Ventricular DysfunctionMI
AbRahman et al., 2022 [39]MalaysiaSelf-controlled case-series studyBNT162b215,387,58535,201,509 *From 12 to 60 years and above4,485,174844 (13.83) d3061 (50.16) dNANANA
CoronaVac17,030,2434,376,900
ChAdOx12,744,5071,008,040
Barda et al., 2021 [40]IsraelRetrospective registry-based analysisBNT162b2884,8281,769,65639.333 (19.274) a461,590 (52)31,836 (4)94,819 (11)NANANA
Control884,82839.333 (19.274) a461,590 (52)31,596 (4)93,357 (11)
Dagan et al., 2021 [26]IsraelSubgroup analysis of Barda 2021 studyBNT162b2884,8281,769,65639.333 (19.274) a461,590 (52)31,836 (4)94,819 (11)NANANA
Control884,82839.333 (19.274) a461,590 (52)31,596 (4)93,357 (11)
Dickerman et al., 2022 [27]USCohort StudyBNT162b2216,836433,67267.667 (10.378) a200,908 (93)59,153 (27) b137,265 (63)NANANA
mRNA-1273216,83667.667 (10.378) a200,908 (93)59,266 (27) b140,774 (65)
El-Shitany et al., 2022 [28]Saudi ArabiaCross-SectionalBNT162b2422422Any age groups c123 (27.8)NA94 (58.4)NANANA
Kadali et al., 2021 [29]USCross-SectionalBNT162b2803803From 18 to 90 years (Average 43 years)108 (13.45)NANANANANA
Kadali et al., 2021a (cross) [30]USCross-SectionalmRNA-1273432432From 18 to 80 years (Average 43.76 years)45 (10.42)8 (1.85)55 (12.73)NANANA
Kaur et al., 2021 [31]IndiaCohort studyChAdOx180480438.44 (11.47) e573 (71.27)NA73 (9)5 (0.6)NANA
Kaur et al., 2021b f [32]GlobalRetrospective registry-based analysisBNT162b242014863NANANANANANA
ChAdOx1262
mRNA-1273400
Montano et al., 2022 [24]GlobalRetrospective registry-based analysisChAdOx134,643,783452,016,608From 18 to 65 years and above266,008,304 (58.85)NANANANANA
Ad26.COV2.S32,233,117
mRNA-1273105,518,547
BNT162b2279,620,827
Patone et al., 2022 g [25]EnglandSelf-controlled case-seriesChAdOx1 (1st) 20,615,9111st dose at least = 38,615,49155.2 (14.8) e7191428 (34.9)NANANANANA
ChAdOx1 (2nd)19,754,22455.4 (14.7) e6900964 (34.9)
BNt162b2 (1st)16,993,38947.8 (21.7) e5401842 (31.8)
BNt162b2 (2nd)11,972,733Two doses = 32,095,748 55.5 (20.4) e3,906,666 (32.6)
mRNA-1273 (1st)1,006,19132.3 (9.4) e286,893 (28.5)
mRNA-1273 (2nd)368,79139.6 (7.3) e97,524 (26.4)
Tan et al., 2021 g [33]SingaporeCohortBNT162b2 (1st)37,3671st dose at least = 64,66127.33 (11.119) a33,913 (90.8)NANANANANA
BNT162b2 (2nd)37,16227 (11.119) a33,766 (90.9)
mRNA-1273 (1st)27,294Two doses = 62,42021 (2.9653) a25,661 (94.0)
mRNA-1273 (2nd)25,25821 (2.9653) a23,710 (93.9)
Tobaiqy et al., 2021 [34]GlobalRetrospective registry-based analysismRNA-12733363420From 18 to 85 years and above171 (50.9)NANANANANA
BNT162b21096494 (45.1)
ChAdOx11988952 (47.9)
Truong et al., 2022 [35]US and CanadaRetrospective studyBNT162b213113917.567 (18.728) e126 (90.6)NANANANANA
mRNA-12735
Ad26.COV2.S1
Unknown2
Witberg et al., 2022 [36] IsraelRetrospective registry-based analysisBNT162b22,558,4212,558,42145.667 (24.463) e1,248,433 (49)NA7 (13) h1 (2) h1 (2) hNA
Wong et al., 2022 [37]ChinaRetrospective studyBNT162b2 (1st)1,308,8201st dose at least = 2,264,67945.7 (16.0) e 584,158 (44.6) 657 (0.05) i 177,913 (13.6) 253,883 (19.4)NA1662 (0.1)
BNT162b2 (2nd)1,116,67745.9 (15.7) e502,740 (45)513 (0.04) i148,978 (13.3)210,140 (18.8)NA1347 (0.1)
CoronaVac (1st)955,859Two doses = 1,938,237 55.3 (14.1) e439,928
(46)
881 (0.09) i225,107
(23.6)
343,413
(35.9)
NA2238 (0.2)
CoronaVac (2nd)821,56054.8 (13.9) e383,164
(46.6)
653 (0.07) i 182,379
(22.2)
274,353
(33.4)
NA1688 (0.2)
Dizbay et al., 2021 [38]TurkeyRetrospective studyCoronaVac1102110237.667 (12.619) a352 (31.9)13 (1.2)48 (4.4)NANANA
* Cumulative numbers include all types of vaccines administered to the population. Numbers may not add up to the total due to missing values. a. Mean, and SD were calculated from median and IQR. b. Cardiovascular disease included acute myocardial infarction, cardiomyopathy, cerebrovascular disease, coronary heart disease, heart failure, and peripheral vascular disease. c. Not reported range or mean and SD. d. In events groups. e. Mean and SD. f. Report data only in AE populations. g. 1st = one dose at least; 2nd = two doses. h. In the myocarditis group only. i. Peripheral vascular disease.
Table 2. Meta-analysis outcomes.
Table 2. Meta-analysis outcomes.
VaccineStudiesPooled IncidenceLower CIUpper CIIRHeterogeneity (I2)p-Value
Pfizer120.22%0.07%0.66%22 per 10,00099.96%<0.001
Sinopharm20.03%0.00%18.48%3 per 10,00099.96%<0.001
Moderna40.76%0.04%12.08%76 per 10,00099.92%<0.001
CoronaVac20.01%0.00%0.03%1 per 10,00099.59%<0.001
AstraZeneca30.04%0.00%0.98%4 per 10,00099.78%<0.001
Table 3. Subgroup analysis.
Table 3. Subgroup analysis.
Subgroup AnalysisStudiesPooled IncidenceLower CIUpper CIHeterogeneity (I2)p-Value
CountryAsia30.006%0%0.009%92.8%<0.001
North America311.95%4.92%18.98%98.6%<0.001
Middle East40.022%0.001%0.046%97%<0.001
Europe10.136%0.134%0.137%--
Study DesignCohort20.692%0%2.04%99.97%<0.001
Retrospective registry-based50.023%0%0.053%98.9%<0.001
Cross-sectional25.25%3.42%7.07%66%0.086
Case-series20.071%0%0.198%100%<0.001
Sample Size >200060.238%0.183%0.294%99.99%<0.001
<200058.51%3.58%13.43%98.05%<0.001
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

Abutaleb, M.H.; Makeen, H.A.; Meraya, A.M.; Alqahtani, S.S.; Al-Mass, B.I.; Aljazaeri, R.O.; Alhazmi, B.D.; Kalakattawi, A.M.N.; Alajam, A.A. Risks of Cardiac Arrhythmia Associated with COVID-19 Vaccination: A Systematic Review and Meta-Analysis. Vaccines 2023, 11, 112. https://doi.org/10.3390/vaccines11010112

AMA Style

Abutaleb MH, Makeen HA, Meraya AM, Alqahtani SS, Al-Mass BI, Aljazaeri RO, Alhazmi BD, Kalakattawi AMN, Alajam AA. Risks of Cardiac Arrhythmia Associated with COVID-19 Vaccination: A Systematic Review and Meta-Analysis. Vaccines. 2023; 11(1):112. https://doi.org/10.3390/vaccines11010112

Chicago/Turabian Style

Abutaleb, Mohammed H., Hafiz A. Makeen, Abdulkarim M. Meraya, Saad S. Alqahtani, Bayan Ibrahim Al-Mass, Reaam Omar Aljazaeri, Bushra Dhuhayyan Alhazmi, Afnan Mohammed Noor Kalakattawi, and Ahmed Ali Alajam. 2023. "Risks of Cardiac Arrhythmia Associated with COVID-19 Vaccination: A Systematic Review and Meta-Analysis" Vaccines 11, no. 1: 112. https://doi.org/10.3390/vaccines11010112

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