Next Article in Journal
Factors Associated with Pneumonia in Patients Hospitalized with COVID-19 and the Role of Vaccination
Next Article in Special Issue
SARS-CoV-2 BA.2.86 (“Pirola”): Is it Pi or Just Another Omicron Sublineage?
Previous Article in Journal
Multimorbidity and Serological Response to SARS-CoV-2 Nine Months after 1st Vaccine Dose: European Cohort of Healthcare Workers—Orchestra Project
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Antibiotic Use Prior to COVID-19 Vaccine Is Associated with Higher Risk of COVID-19 and Adverse Outcomes: A Propensity-Scored Matched Territory-Wide Cohort

1
Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
2
Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
3
Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong
4
Department of Pharmacy, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
5
Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen 518057, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Vaccines 2023, 11(8), 1341; https://doi.org/10.3390/vaccines11081341
Submission received: 11 July 2023 / Revised: 31 July 2023 / Accepted: 4 August 2023 / Published: 8 August 2023
(This article belongs to the Special Issue COVID Vaccines: Design, Development, and Immune Response Studies)

Abstract

:
Background: Antibiotics may increase the risk of COVID-19 among non-vaccinated subjects via probable gut dysbiosis. We aimed to investigate whether antibiotics also affect the clinical outcomes of COVID-19 vaccine recipients. Methods: This was a territory-wide cohort study of 3,821,302 COVID-19 vaccine recipients (aged ≥ 18 years) with ≥2 doses of either BNT162b2 or CoronaVac. Exclusion criteria included prior COVID-19, prior gastrointestinal surgery, and immunocompromised status. The primary outcome was COVID-19 infection and secondary outcomes included COVID-19-related hospitalization and severe infection (composite of intensive care unit admission, ventilatory support, and/or death). Exposure was pre-vaccination antibiotic use (within 180 days of first vaccine dose). Covariates included age, sex, Charlson Comorbidity Index, and concomitant medication use. Subjects were followed from the index date (first dose vaccination) until outcome occurrence, death, an additional dose of vaccination, or 15 November 2022. Propensity score (PS) matching and a Poisson regression model were used to estimate the adjusted incidence rate ratio (aIRR) of outcomes with antibiotic use. Results: Among 342,338 PS matched three-dose vaccine recipients (mean age: 57.4 years; male: 45.1%) with a median follow-up of 13.6 months (IQR: 9.2–16.3), antibiotics were associated with a higher risk of COVID-19 infection (aIRR: 1.16;95% CI: 1.14–1.19), hospitalization (aIRR: 1.75;95% CI: 1.65–1.86), and severe infection (aIRR: 1.60; 95% CI: 1.21–2.11). Notably, antibiotic use was associated with a higher risk of severe infection and death among CoronaVac recipients (aIRR: 1.62 95% CI: 1.18–2.22 and aIRR: 2.70, 95% CI: 1.54–4.73 for the two secondary outcomes, respectively), but not BNT162b2 recipients. Conclusions: Pre-vaccination use of antibiotics was associated with a higher risk of COVID-19 infection, hospitalization, and severe disease outcomes.

1. Introduction

According to the World Health Organization, COVID-19 has led to nearly 7 million deaths globally as of May 2023. Vaccination is one of the most important measures in preventing severe symptoms and death [1]. There is increasing evidence of the role of the gut microbiome on the impact of COVID-19 disease as well as vaccine immunogenicity [2,3]. Gut microbiota perturbation induced by antibiotics could impair antibody production and affinity among those with low pre-existing antibody titers against influenza virus [4]. Similarly, the early vaccine immunogenicity of BNT162b2 may be impaired by antibiotic usage [5]. The mechanisms underlying these observations include the expression of viral entry receptor angiotensin-converting enzyme 2 (ACE2) [6], the effects of microbiota-derived short chain fatty acids [7], the modulation of B and T cell immune responses [3], and the mediation of the “gut-lung axis”. Gut microbiota alteration by COVID-19 has been associated with inflammatory bowel disease and COVID-19 severity. Intuitively, it has been postulated that gut dysbiosis induced by antibiotics leads to a higher viral load in the gastrointestinal (GI) tract, resulting in a cytokine storm [8] and the impairment of leukocyte function [9]—which is similar to the effects of proton-pump inhibitors (PPIs) in increasing the risk of COVID-19 and severe clinical outcomes in observational studies [10,11].
Worldwide antibiotic use increased by 65% between 2000 and 2015 [12]. Antibiotic misuse was exemplified in a recent study evaluating antibiotic use among 11,047 COVID-19 cases in Hong Kong, carried out between January 2018 to March 2021, that revealed that nearly 30% of cases were given antibiotics—but only less than 2% had confirmed bacterial co-infections [13]. It has been shown that recent usage of antibiotics affects COVID-19 vaccine immunogenicity [5]. However, there is a lack of population-based studies investigating whether antibiotics could increase the risk of COVID-19 and serious clinical outcomes after vaccination. A differential effect may also exist depending on the number of vaccine doses received and the vaccine platform (messenger RNA (mRNA) vs. inactivated virus).
Here, we conducted a territory-wide cohort study on COVID-19 vaccine recipients to determine the effects of antibiotics on the development of COVID-19 and adverse clinical outcomes, with stratification.

2. Methods

2.1. Data Source

A retrospective cohort study was conducted using the electronic medical record database from the Hospital Authority (HA; including all individuals who ever used HA services), linked with territory-wide vaccination records and COVID-19 confirmed case records from the Department of Health using anonymous unique patient identifiers. Medical history and comorbidities were identified by International Classification of Diseases, Ninth Revision (ICD-9) diagnosis codes, looking back to 2018 at the earliest. Medication use was identified by prescription records using British National Formulary (BNF) codes.

2.2. Setting and Study Population

The Hong Kong government provides the public with COVID-19 vaccines (BNT162b2 and CoronaVac) free of charge. Local residents can choose either vaccine according to their own preference. They can receive vaccination according to the recommended vaccination schedule in designated clinics and community vaccination centers.
Individuals aged ≥18 who had received 3 doses of either BNT162b2 or CoronaVac vaccine during the study period (23 February 2021 to 15 November 2022) were included (n = 3,299,819). Figure S1 shows the study subject selection process. Subsequently, patients with a history of GI surgery (n = 17,372), who were immunocompromised (cancer; transplant receipient; primary immunodeficiency; immune-related inflammatory disorders including rheumatoid arthritis, inflammatory bowel disease, and others; splenectomy; end-stage renal disease/dialysis; n = 129,984), who had a history of COVID-19 before the first dose of the vaccination (n = 18,017), or who used any antibiotic between the first and third dose of vaccination (n = 140,060) were excluded. The cohort size of our study was 2,994,386 after exclusion.
Patients were followed from the index date (date of first-dose vaccination) until the earliest of the following: outcome occurrence, death, date of vaccination of an additional dose, or end of data availability (15 November 2022).

2.3. Exposures of Interest

The primary exposure of interest is the pre-vaccination use of antibiotics. Table S2 shows the list of antibiotics included and their classification in terms of their anti-aerobic/anti-anaerobic effects and anti-bacterial spectrum. Pre-vaccination antibiotic users were those who received any prescription of any antibiotics within 180 days before their first-dose vaccination [5]. Antibiotic non-users were those who did not receive any prescription of antibiotics within 180 days from the first dose of vaccination. Figure 1 shows the study timeframe.

2.4. Outcomes of Interest

Outcomes of interest included: (i) COVID-19, defined by a positive polymerase chain reaction (PCR) test (voluntary reporting of rapid antigen test (RAT) positive test was not included) as confirmed by the Department of Health of Hong Kong Special Administrative Region Government; (ii) COVID-19-related hospitalization, defined as hospital admission within 28 days following a positive PCR test; (iii) COVID-19-related mortality, defined as all-cause mortality within 28 days following a positive PCR test; (iv) severe COVID-19, defined as a composite of COVID-19-related mortality or intensive care unit (ICU) admission or ventilatory support (ICD-9 procedure codes: 39.65, 89.18, 93.90, 93.95, 93.96, 96.7, 96.04) within 28 days following a positive PCR test.

2.4.1. Statistical Analyses

R software (version 3.2.3) was used for all statistical analyses. Continuous variables were expressed as mean with standard deviation (SD) or median with interquartile range (IQR). Discrete variables were expressed as counts with percentage.
Covariates included age, sex, Charlson Comorbidity Index (CCI) score [14], index date, presence of comorbidities (hypertension [15], diabetes mellitus [16], dyslipidemia [17], cardiovascular disease [18], respiratory disease, obesity diagnosis [19], smoking [20], alcohol use disorders [21], ulcers, moderate-to-severe liver disease [22,23,24], chronic renal failure [25]), and medication use within the past 90 days (angiotensin-converting enzyme inhibitors (ACEIs), angiotensin receptor blockers (ARBs) [26], metformin, lipid-modifying drugs, antiplatelets, nonsteroidal anti-inflammatory drugs (NSAIDs), oral anticoagulants, oral corticosteroids, antidepressants, antiviral drugs, PPIs [10,11], or H2 receptor antagonists (H2RAs) [27].
Propensity-score matching was conducted to minimize potential confounding between pre-vaccination antibiotic users versus non-users at a 1:1 ratio using the nearest-neighbor algorithm with a caliper of 0.2, where the propensity score was estimated using logistic regression for the covariates listed above. A standardized mean difference (SMD) of 0.2 or less was considered negligible.
Poisson regression adjusted for covariates was used to estimate the incidence rate ratio (IRR) of outcomes among pre-vaccination antibiotic users versus non-users in the propensity-score (PS)-matched cohort. Stratified analyses by age (<60 and ≥60), sex, Charlson Comorbidity Index categories (0 and ≥1), and vaccine platform were conducted. Effects of the nature of various antibiotics (anti-aerobic vs. anti-anaerobic, narrow- vs. broad-spectrum, and intravenous vs. oral) and different antibiotic classes used pre-vaccination on vaccine effectiveness were also estimated.
To increase the robustness of the study results by demonstrating a biological gradient, the association between pre-vaccination cumulative antibiotic exposure and outcomes were estimated using Poisson regression adjusted for the covariates listed above. Cumulative antibiotic exposure in the past 180 days before first dose vaccination were categorized by their cumulative duration (0 (non-users), 1–7, and ≥8 days). Incidence rate ratios (IRRs) and 95% confidence intervals were reported.
A two-sided p-value of less than 0.05 was considered statistically significant.

2.4.2. Statements of Ethics

The study protocol conformed to the ethical guidelines of the World Medical Association Declaration of Helsinki and was approved by the Institutional Review Board of the University of Hong Kong and the Hong Kong West Cluster of Hospital Authority.

3. Results

3.1. Patient Characteristics

In total, 342,338 three-dose vaccine recipients (171,169 antibiotic users and 171,169 antibiotic non-users) were PS matched. The mean age was 57.2 years (SD: 18.2) in antibiotic non-users and 57.6 (SD: 19.1) in antibiotic users. The median follow-up was 13.6 months (IQR: 9.2–16.3), and the median duration of pre-vaccination antibiotic use was 7.0 days (IQR: 6.0–9.0). The difference in baseline characteristics between antibiotic users and non-users before PS matching and after PS matching (all with SMD < 0.2) is shown in Table S1 and Table 1, respectively.

3.2. Association between Pre-Vaccination Antibiotic Use and COVID-19 Outcomes

Among the PS-matched vaccine recipients, 45,173 (13.2%) developed COVID-19. The median time from the index date to development of infection was 7.9 months (IQR: 5.7–10.8), respectively. Pre-vaccination antibiotic use was associated with a higher risk of COVID-19 (aIRR: 1.16; 95% CI: 1.14–1.18), COVID-19-related hospitalization (aIRR: 1.75; 95% CI: 1.65–1.86), and severe disease outcomes (composite of ICU admission/ventilatory support/death—aIRR: 1.60; 95% CI: 1.21–2.11) and death (aIRR: 2.56; 95% CI: 1.56–4.20) (Table 2).
Antibiotic use was associated with a similarly higher risk of infection regardless of the duration of use (Table 3). A biological gradient existed for COVID-19 hospitalization and severe COVID-19. Compared with antibiotic non-use, the use of antibiotics for 7 days or less was associated with an approximately 1.5-fold higher risk of these outcomes (hospitalization aIRR: 1.57 (95% CI: 1.47–1.69); severe outcomes aIRR: 1.49 (95% CI: 1.08–2.04), respectively); while use of antibiotics for ≥8 days was associated with a 1.8- to 2.0-fold higher risk of these outcomes (hospitalization aIRR: 2.07 (95% CI: 1.92–2.23); severe outcomes aIRR: 1.77 (95% CI: 1.26–2.50), respectively; Table 3). However, a longer duration of antibiotic use did not pose a higher risk of COVID-19 mortality compared with shorter durations.

3.3. Stratified Analysis

3.3.1. Stratified by Vaccine Platform, Age, Sex, and Charlson Comorbidity Index

The magnitude of the higher infection risk and hospitalization associated with antibiotic use was similar regardless of vaccine platform, age, sex, or CCI (Table 4).
However, antibiotic use was associated with a higher risk of severe disease outcomes and death with CoronaVac only (aIRR: 1.62 95% CI: 1.18–2.22; aIRR: 2.70, 95% CI: 1.54–4.73 for the two secondary outcomes, respectively), aged ≥60 years (aIRR: 1.57 95% CI: 1.17–2.11; aIRR: 2.53, 95% CI: 1.54–4.15 for the two secondary outcomes, respectively), and those with CCI > 1 (aIRR: 1.94 95% CI: 1.38–2.74; aIRR: 2.66, 95% CI: 1.49–4.76 for the two secondary outcomes, respectively). On the other hand, the magnitude of risk for the secondary outcomes was similar for males and females.

3.3.2. Stratified by Nature and Class of Antibiotics

Compared with antibiotic non-use, anti-anaerobic, broad-spectrum, and oral antibiotics were associated with a higher infection risk (aIRR: 1.16, 95% CI: 1.14–1.19; aIRR: 1.16, 95% CI: 1.14–1.18; aIRR: 1.16, 95% CI: 1.14–1.18, respectively), hospitalization (aIRR: 1.71, 95% CI: 1.60–1.83; aIRR: 1.77, 95% CI: 1.67–1.89; aIRR: 1.66, 95% CI: 1.60–1.77, respectively) and severe infection outcomes (aIRR: 1.70, 95% CI: 1.26–2.29; aIRR: 1.68, 95% CI: 1.26–2.22; aIRR: 1.53, 95% CI: 1.14–2.04, respectively; Table 5). Broad-spectrum antibiotics were associated with a higher infection risk (aIRR: 1.16; 95% CI: 1.14–1.18) and severe infection outcomes (aIRR: 1.67; 95% CI: 1.26–2.22). Anti-aerobic, narrow-spectrum, and intravenous antibiotics were associated with a higher infection risk (aIRR: 1.11, 95% CI: 1.07–1.15; aIRR: 1.14, 95% CI: 1.09–1.20; aIRR: 1.15, 95% CI: 1.02–1.29, respectively), but not severe infection outcomes (aIRR: 1.49, 95% CI: 0.88–2.53; aIRR: 0.79, 95% CI: 0.25–2.51; aIRR: 1.92, 95% CI: 0.77–4.76, respectively).
Regarding antibiotic class (Table S3), compared with antibiotic non-use, penicillins (aIRR: 1.17; 95% CI: 1.15–1.19), cephalosporins (aIRR: 1.12; 95% CI: 1.04–1.21), macrolides (aIRR: 1.12; 95% CI: 1.05–1.19), carbapenems (aIRR: 1.35; 95% CI: 1.16–1.58), quinolones (aIRR: 1.16; 95% CI: 1.11–1.21), tetracyclines (aIRR: 1.20; 95% CI: 1.13–1.28), and aminoglycosides (aIRR: 1.45; 95% CI: 1.11–1.89) were associated with a higher risk of COVID-19—but this was not the case with nitroimidazoles (aIRR: 1.04; 95% CI: 0.98–1.10) or glycopeptides (aIRR: 1.14; 95% CI: 0.92–1.42). All antibiotic classes were associated with a higher risk of COVID-19-related hospitalization. Penicillins and quinolones were associated with a higher risk of severe COVID-19 (aIRR: 1.62, 95% CI: 1.22–2.16 and aIRR: 1.75, 95% CI: 1.06–2.91, respectively).

4. Discussion

This is the first study to demonstrate the potential undesirable effects of pre-vaccination antibiotics on the COVID-19 disease course, with a higher risk of infection, hospitalization, and severe disease outcomes being demonstrated in a territory-wide cohort study involving >300,000 PS-matched three-dose vaccine recipients, with a biological gradient being demonstrated. Notably, severe infection and death associated with pre-vaccination antibiotic use was only observed in CoronaVac recipients, but not in BNT162b2 recipients.
Antibiotics can cause significant changes in gut microbiota that have both short- and long-term health consequences [28]. Antibiotics reduce the overall diversity of gut microbiota species—including the loss of some important taxa—which causes metabolic shifts, increases gut susceptibility to colonization, and stimulates the development of bacterial antibiotic resistance [29]. Given the high efficacy of two doses of BNT162b2 and CoronaVac (protection from symptomatic COVID-19 is 95% [30] and 70% [31], respectively), it is hardly surprising that the effects of antibiotics on increasing infection risk was only slightly elevated (5–7%) in two- or three-dose vaccine recipients in the current study.
Although booster doses can enhance antibody production [32,33] and reduce the severity of infection outcomes [34], we showed that antibiotics were still associated with 1.8-fold and 1.6-fold higher risks of hospitalization and severe clinical outcomes (including ICU admission, ventilatory support, and death), respectively, in the three-dose vaccine recipients. Bacterial co-infection causes life-threatening complications in patients with severe viral infections [35], including COVID-19. Studies show that early antibiotic exposure poses risks for childhood asthma, allergies, and airway illnesses [36]. Indiscriminate administration of broad-spectrum antibiotics may increase nosocomial bloodstream infection rates in immunocompromised cancer patients [37]. While the baseline composition of the gut microbiota is mostly restored within 1.5 months, several common species (e.g., Bifidobacterium adolescentis, Coprococcus eutactus, Coliinsella aerofaciens, Methanobrevibacter smithii, Bididobacterium catenulatum, Bifidobacterium angulatum, and Eubacterium ventriosum) remain undetectable for at least 180 days [38]. With the combination of the immunocompromising effects of the viral infection and the antibiotic-driven depletion of commensal gut microbes, it has been proven that microorganisms from the dysbiotic gut microbiome translocate into the blood of COVID-19 patients, leading to dangerous blood-streamed infection during COVID-19 [39,40].
Subgroup analysis showed that antibiotic use was associated with a higher risk of adverse outcomes for COVID-19 with CoronaVac, but not BNT162b2, among the three-dose recipients. BNT162b2 mounts a higher neutralizing antibody level of nearly 10-fold more than CoronaVac after two doses of vaccine [40]. Owing to the waning effects of serum antibody levels, vaccine effectiveness against infection progressively diminishes [41]. This corroborates the hypothesis that a higher humoral response induced by BNT162b2 may minimize the unfavorable effects of antibiotics on vaccine immunogenicity and adverse clinical outcomes. A similar phenomenon was observed among older individuals and those with comorbidities, but not among younger age groups and those without comorbidities. The presence of comorbidities is linked to decreased immunity, reduced functional status, and increased healthcare utilization [42]. Advanced age and the number of concurrent comorbidities could also have a synergistic adverse effect on the health status of the elderly, explaining the higher COVID-19 mortality observed among elderly comorbid patients [43].
It was observed that broad-spectrum antibiotics were associated with a higher infection risk in the three-dose vaccine recipients, but not narrow-spectrum antibiotics. Furthermore, the intravenous route of the antibiotics had an overall higher risk of adverse infection outcomes than the oral route. Both gram-positive and gram-negative bacteria are disrupted by broad-spectrum antibiotics, as opposed to narrow spectrum antibiotics [44]. Broad-spectrum antibiotics decrease the Firmicutes to Bacteroidetes (F/B) ratio [45], which is associated with maintaining homeostasis, and changes in this ratio are regarded as dysbiosis. Models have shown that broad-spectrum antimicrobials (specifically vancomycin, metronidazole, and lacticin) cause population shifts from Firmicutes to Proteobacteria in the distal colon. In contrast, the narrow-spectrum bacteriocin (thuricin CD) seems to cause negligible changes in gut flora [46]. One study revealed a significant difference in the antibiotic resistance gene pools in mice treated with tetracycline between intravenous injection and oral administration of the drug, suggesting that drug administration routes influence the level of antibiotic resistance in gut microbiota [47]. The exact mechanism between the type, dosage, duration, administration, and pharmacokinetic and pharmacodynamics properties of antibiotics with vaccine immunogenicity and disease outcomes requires further investigation.
This study had several strengths: First, the population-based study with a large sample size allowed us to detect a statistically significant, albeit small in magnitude, increase in risk of infection—which is important for verifying the hypothesis of antibiotic-induced gut microbiota dysbiosis leading to impaired vaccine immunogenicity. Second, the demonstration of a biological gradient for the majority of outcomes increased the robustness of the study results and strengthened our underlying hypothesis. Third, reverse causality (i.e., symptomatic COVID-19 leading to antibiotic use before infection diagnosis) was minimized, as antibiotic use was defined before the first dose of vaccination while infection outcomes were only counted after the second dose of vaccination. With such a study design, the minimal interval between the commencement of antibiotics was at least three and four weeks for BNT162b2 and CoronaVac recipients, respectively. Fourth, since this was a cohort study, there was less recall and interviewer bias in terms of antibiotics as compared with a case–control study. To minimize the effects of confounding factors on the causality, PS matching ensured the similarity of the baseline characteristics between antibiotic users and non-users in order to reach a quasi-experimental design that may not be otherwise have been achieved by a randomized clinical trial (RCT).
Several limitations of this study should be noted: First, unmeasured confounding may still exist due to the observational study design, although all major confounding factors affecting vaccine immunogenicity and disease outcomes were minimized by the large sample size and PS matching design (Table 1). Second, gut microbiota data was not available; thus, the exact mechanisms of antibiotic-induced gut dysbiosis on vaccine immunogenicity and COVID-19 disease outcomes require further study. Third, asymptomatic infection cannot be ruled out, as this study included only PCR-confirmed positive case, which may overestimate the effects of antibiotics. Fourth, data on further booster doses and bivalent vaccines were not available before the study end date. Fifth, over-the-counter antibiotic usage information was not available, which may bias the study results. For example, self administered antibiotics may be taken incorrectly for too short a time, with wrong doses or timing with food intake. Lastly, COVID-19 vaccination effectiveness may vary in different waves of the epidemic due to the difference in the number of infected cases and prevention protocol/testing criteria from government policies. A prior study showed different frequencies of COVID-19 cases in different waves before the rollout of the vaccination program [48].

5. Conclusions

Antibiotics were associated with a higher COVID-19 risk, hospitalization, and severe infection in three-dose vaccine recipients. Further studies are warranted to investigate the interactions between gut microbiota and antibiotics and their effects on the COVID-19 vaccine’s immunogenicity and clinical outcomes. These results further reinforce the importance of the judicious use of antibiotics to minimize their direct and indirect impact on health.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/vaccines11081341/s1, Figure S1: Patient selection flow chart; Table S1: Baseline characteristics of pre-vaccination antibiotic users and non-users before propensity score matching; Table S2: Anti-anaerobic/anti-aerobic activity and anti-bacterial spectrum of various classes of antibiotics; Table S3: Association between pre-vaccination use of different classes of antibiotics and COVID-19 outcomes after vaccination with three doses of BNT162b2/CoronaVac.

Author Contributions

Conceptualization, K.S.C. and W.K.L.; methodology, K.S.C., V.K.C.Y., E.W.C. and W.K.L.; data collection, V.K.C.Y. and E.W.C.; statistical analysis, V.K.C.Y. and X.Y.; drafting of manuscript, K.S.C., V.K.C.Y. and L.K.L.; supervision and revision of manuscript; I.F.N.H., E.W.C. and W.K.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by a research grant from the the Food and Health Bureau; HMRF Research on COVID-19, The Government of the Hong Kong Special Administrative Region (Principal Investigator (WP2): EWC; Ref: COVID1903011).

Institutional Review Board Statement

This study was approved by the Central Institutional Review Board of the Hospital Authority of Hong Kong (CIRB-2021-005-4) and the DH Ethics Committee (LM171/2021).

Informed Consent Statement

Informed consent was not needed as all subjects were anonymized in the electronic healthcare database system.

Data Availability Statement

Data sharing will be available upon reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Adjusted incidence rate ratio, aIRR; angiotensin-converting enzyme 2, ACE2; angiotensin-converting enzyme inhibitors, ACEIs; angiotensin receptor blockers, ARBs; British National Formulary, BNF; Charlson Comorbidity Index, CCI; gastrointestinal, GI; H2 receptor antagonists, H2RAs; Hospital Authority, HA; incidence rate ratio, IRR; intensive care unit, ICU; International Classification of Diseases, Ninth Revision, ICD-9; interquartile range, IQR; messenger RNA, mRNA; nonsteroidal anti-inflammatory drugs, NSAIDs; polymerase chain reaction, PCR; propensity score, PS; proton-pump inhibitors, PPIs; rapid antigen test, RAT; standard deviation, SD; standardized mean difference, SMD.

References

  1. Haas, E.J.; Angulo, F.J.; McLaughlin, J.M.; Anis, E.; Singer, S.R.; Khan, F.; Brooks, N.; Smaja, M.; Mircus, G.; Pan, K.; et al. Impact and effectiveness of mRNA BNT162b2 vaccine against SARS-CoV-2 infections and COVID-19 cases, hospitalisations, and deaths following a nationwide vaccination campaign in Israel: An observational study using national surveillance data. Lancet 2021, 397, 1819–1829. [Google Scholar] [CrossRef] [PubMed]
  2. Ng, H.Y.; Leung, W.K.; Cheung, K.S. Association between Gut Microbiota and SARS-CoV-2 Infection and Vaccine Immunogenicity. Microorganisms 2023, 11, 452. [Google Scholar] [CrossRef] [PubMed]
  3. Lynn, D.J.; Benson, S.C.; Lynn, M.A.; Pulendran, B. Modulation of immune responses to vaccination by the microbiota: Implications and potential mechanisms. Nat. Rev. Immunol. 2022, 22, 33–46. [Google Scholar] [CrossRef] [PubMed]
  4. Hagan, T.; Cortese, M.; Rouphael, N.; Boudreau, C.; Linde, C.; Maddur, M.S.; Das, J.; Wang, H.; Guthmiller, J.; Zheng, N.Y.; et al. Antibiotics-Driven Gut Microbiome Perturbation Alters Immunity to Vaccines in Humans. Cell 2019, 178, 1313–1328.e1313. [Google Scholar] [CrossRef] [PubMed]
  5. Cheung, K.S.; Lam, L.K.; Zhang, R.; Ooi, P.H.; Tan, J.T.; To, W.P.; Hui, C.H.; Chan, K.H.; Seto, W.K.; Hung, I.F.N.; et al. Association between Recent Usage of Antibiotics and Immunogenicity within Six Months after COVID-19 Vaccination. Vaccines 2022, 10, 1122. [Google Scholar] [CrossRef] [PubMed]
  6. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Kruger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e278. [Google Scholar] [CrossRef]
  7. Portincasa, P.; Bonfrate, L.; Vacca, M.; De Angelis, M.; Farella, I.; Lanza, E.; Khalil, M.; Wang, D.Q.; Sperandio, M.; Di Ciaula, A. Gut Microbiota and Short Chain Fatty Acids: Implications in Glucose Homeostasis. Int. J. Mol. Sci. 2022, 23, 1105. [Google Scholar] [CrossRef]
  8. Liu, Y.; Kuang, D.; Li, D.; Yang, J.; Yan, J.; Xia, Y.; Zhang, F.; Cao, H. Roles of the gut microbiota in severe SARS-CoV-2 infection. Cytokine Growth Factor Rev. 2022, 63, 98–107. [Google Scholar] [CrossRef]
  9. Ghazanfar, H.; Kandhi, S.; Shin, D.; Muthumanickam, A.; Gurjar, H.; Qureshi, Z.A.; Shaban, M.; Farag, M.; Haider, A.; Budhathoki, P.; et al. Impact of COVID-19 on the Gastrointestinal Tract: A Clinical Review. Cureus 2022, 14, e23333. [Google Scholar] [CrossRef]
  10. Almario, C.V.; Chey, W.D.; Spiegel, B.M.R. Increased Risk of COVID-19 Among Users of Proton Pump Inhibitors. Am. J. Gastroenterol. 2020, 115, 1707–1715. [Google Scholar] [CrossRef]
  11. Lee, S.W.; Ha, E.K.; Yeniova, A.; Moon, S.Y.; Kim, S.Y.; Koh, H.Y.; Yang, J.M.; Jeong, S.J.; Moon, S.J.; Cho, J.Y.; et al. Severe clinical outcomes of COVID-19 associated with proton pump inhibitors: A nationwide cohort study with propensity score matching. Gut 2021, 70, 76–84. [Google Scholar] [CrossRef] [PubMed]
  12. Klein, E.Y.; Van Boeckel, T.P.; Martinez, E.M.; Pant, S.; Gandra, S.; Levin, S.A.; Goossens, H.; Laxminarayan, R. Global increase and geographic convergence in antibiotic consumption between 2000 and 2015. Proc. Natl. Acad. Sci. USA 2018, 115, E3463–E3470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kwok, K.O.; Wei, W.I.; Ma, B.H.M.; Ip, M.; Cheung, H.; Hui, E.; Tang, A.; McNeil, E.B.; Wong, S.Y.S.; Yeoh, E.K. Antibiotic use among COVID-19 patients in Hong Kong, January 2018 to March 2021. J. Infect. 2022, 84, e129–e132. [Google Scholar] [CrossRef]
  14. Charlson, M.E.; Pompei, P.; Ales, K.L.; MacKenzie, C.R. A new method of classifying prognostic comorbidity in longitudinal studies: Development and validation. J. Chronic. Dis. 1987, 40, 373–383. [Google Scholar] [CrossRef] [PubMed]
  15. Soegiarto, G.; Wulandari, L.; Purnomosari, D.; Dhia Fahmita, K.; Ikhwan Gautama, H.; Tri Hadmoko, S.; Edwin Prasetyo, M.; Aulia Mahdi, B.; Arafah, N.; Prasetyaningtyas, D.; et al. Hypertension is associated with antibody response and breakthrough infection in health care workers following vaccination with inactivated SARS-CoV-2. Vaccine 2022, 40, 4046–4056. [Google Scholar] [CrossRef]
  16. Boroumand, A.B.; Forouhi, M.; Karimi, F.; Moghadam, A.S.; Naeini, L.G.; Kokabian, P.; Naderi, D. Immunogenicity of COVID-19 vaccines in patients with diabetes mellitus: A systematic review. Front. Immunol. 2022, 13, 940357. [Google Scholar] [CrossRef] [PubMed]
  17. Guo, Q.; Yang, L.; Peng, R.; Gao, T.; Chu, X.; Jiang, D.; Ke, D.; Ren, H. Safety and immunogenicity of inactivated COVID-19 vaccine in patients with metabolic syndrome: A cross-sectional observational study. Front. Public Health 2022, 10, 1067342. [Google Scholar] [CrossRef]
  18. Naruse, H.; Ito, H.; Izawa, H.; Sarai, M.; Ishii, J.; Sakaguchi, E.; Murakami, R.; Ando, T.; Fujigaki, H.; Saito, K. Immunogenicity of BNT162b2 mRNA COVID-19 Vaccine in Patients with Cardiovascular Disease. J. Clin. Med. 2021, 10, 5498. [Google Scholar] [CrossRef] [PubMed]
  19. Ou, X.; Jiang, J.; Lin, B.; Liu, Q.; Lin, W.; Chen, G.; Wen, J. Antibody responses to COVID-19 vaccination in people with obesity: A systematic review and meta-analysis. Influenza Other Respir. Viruses 2023, 17, e13078. [Google Scholar] [CrossRef]
  20. Ferrara, P.; Gianfredi, V.; Tomaselli, V.; Polosa, R. The Effect of Smoking on Humoral Response to COVID-19 Vaccines: A Systematic Review of Epidemiological Studies. Vaccines 2022, 10, 303. [Google Scholar] [CrossRef]
  21. Yamamoto, S.; Tanaka, A.; Ohmagari, N.; Yamaguchi, K.; Ishitsuka, K.; Morisaki, N.; Kojima, M.; Nishikimi, A.; Tokuda, H.; Inoue, M.; et al. Use of heated tobacco products, moderate alcohol drinking, and anti-SARS-CoV-2 IgG antibody titers after BNT162b2 vaccination among Japanese healthcare workers. Prev. Med. 2022, 161, 107123. [Google Scholar] [CrossRef] [PubMed]
  22. Cheung, K.S.; Lam, L.K.; Mao, X.; Tan, J.T.; Ooi, P.H.; Zhang, R.; Chan, K.H.; Hung, I.F.N.; Seto, W.K.; Yuen, M.F. Effect of Moderate to Severe Hepatic Steatosis on Vaccine Immunogenicity against Wild-Type and Mutant Virus and COVID-19 Infection among BNT162b2 Recipients. Vaccines 2023, 11, 497. [Google Scholar] [CrossRef] [PubMed]
  23. Cheung, K.S.; Lam, L.K.; Hui, R.W.H.; Mao, X.; Zhang, R.R.; Chan, K.H.; Hung, I.F.; Seto, W.K.; Yuen, M.F. Effect of moderate-to-severe hepatic steatosis on neutralising antibody response among BNT162b2 and CoronaVac recipients. Clin. Mol. Hepatol. 2022, 28, 553–564. [Google Scholar] [CrossRef] [PubMed]
  24. Cheung, K.S.; Mok, C.H.; Mao, X.; Zhang, R.; Hung, I.F.; Seto, W.K.; Yuen, M.F. COVID-19 vaccine immunogenicity among chronic liver disease patients and liver transplant recipients: A meta-analysis. Clin. Mol. Hepatol. 2022, 28, 890–911. [Google Scholar] [CrossRef]
  25. Ma, B.M.; Tam, A.R.; Chan, K.W.; Ma, M.K.M.; Hung, I.F.N.; Yap, D.Y.H.; Chan, T.M. Immunogenicity and Safety of COVID-19 Vaccines in Patients Receiving Renal Replacement Therapy: A Systematic Review and Meta-Analysis. Front. Med. 2022, 9, 827859. [Google Scholar] [CrossRef]
  26. Cheung, K.S.; Hung, I.F.N.; Leung, W.K. Association between angiotensin blockade and COVID-19 severity in Hong Kong. Can. Med. Assoc. J. 2020, 192, E635. [Google Scholar] [CrossRef]
  27. Cheung, K.S.; Hung, I.F.N.; Leung, W.K. Association Between Famotidine Use and COVID-19 Severity in Hong Kong: A Territory-wide Study. Gastroenterology 2021, 160, 1898–1899. [Google Scholar] [CrossRef]
  28. Ramirez, J.; Guarner, F.; Bustos Fernandez, L.; Maruy, A.; Sdepanian, V.L.; Cohen, H. Antibiotics as Major Disruptors of Gut Microbiota. Front. Cell. Infect. Microbiol. 2020, 10, 572912. [Google Scholar] [CrossRef]
  29. Lange, K.; Buerger, M.; Stallmach, A.; Bruns, T. Effects of Antibiotics on Gut Microbiota. Dig. Dis. 2016, 34, 260–268. [Google Scholar] [CrossRef]
  30. 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]
  31. Jara, A.; Undurraga, E.A.; González, C.; Paredes, F.; Fontecilla, T.; Jara, G.; Pizarro, A.; Acevedo, J.; Leo, K.; Leon, F.; et al. Effectiveness of an Inactivated SARS-CoV-2 Vaccine in Chile. N. Engl. J. Med. 2021, 385, 875–884. [Google Scholar] [CrossRef] [PubMed]
  32. Munro, A.P.S.; Janani, L.; Cornelius, V.; Aley, P.K.; Babbage, G.; Baxter, D.; Bula, M.; Cathie, K.; Chatterjee, K.; Dodd, K.; et al. Safety and immunogenicity of seven COVID-19 vaccines as a third dose (booster) following two doses of ChAdOx1 nCov-19 or BNT162b2 in the UK (COV-BOOST): A blinded, multicentre, randomised, controlled, phase 2 trial. Lancet 2021, 398, 2258–2276. [Google Scholar] [CrossRef] [PubMed]
  33. Nemet, I.; Kliker, L.; Lustig, Y.; Zuckerman, N.; Erster, O.; Cohen, C.; Kreiss, Y.; Alroy-Preis, S.; Regev-Yochay, G.; Mendelson, E.; et al. Third BNT162b2 Vaccination Neutralization of SARS-CoV-2 Omicron Infection. N. Engl. J. Med. 2022, 386, 492–494. [Google Scholar] [CrossRef]
  34. Barda, N.; Dagan, N.; Cohen, C.; Hernán, M.A.; Lipsitch, M.; Kohane, I.S.; Reis, B.Y.; Balicer, R.D. Effectiveness of a third dose of the BNT162b2 mRNA COVID-19 vaccine for preventing severe outcomes in Israel: An observational study. Lancet 2021, 398, 2093–2100. [Google Scholar] [CrossRef] [PubMed]
  35. Yu, D.; Ininbergs, K.; Hedman, K.; Giske, C.G.; Stralin, K.; Ozenci, V. Low prevalence of bloodstream infection and high blood culture contamination rates in patients with COVID-19. PLoS ONE 2020, 15, e0242533. [Google Scholar] [CrossRef]
  36. Zou, Z.; Liu, W.; Huang, C.; Sun, C.; Zhang, J. First-Year Antibiotics Exposure in Relation to Childhood Asthma, Allergies, and Airway Illnesses. Int. J. Environ. Res. Public Health 2020, 17, 5700. [Google Scholar] [CrossRef]
  37. Taur, Y.; Xavier, J.B.; Lipuma, L.; Ubeda, C.; Goldberg, J.; Gobourne, A.; Lee, Y.J.; Dubin, K.A.; Socci, N.D.; Viale, A.; et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin. Infect. Dis. 2012, 55, 905–914. [Google Scholar] [CrossRef]
  38. Palleja, A.; Mikkelsen, K.H.; Forslund, S.K.; Kashani, A.; Allin, K.H.; Nielsen, T.; Hansen, T.H.; Liang, S.; Feng, Q.; Zhang, C.; et al. Recovery of gut microbiota of healthy adults following antibiotic exposure. Nat. Microbiol. 2018, 3, 1255–1265. [Google Scholar] [CrossRef]
  39. Venzon, M.; Bernard-Raichon, L.; Klein, J.; Axelrad, J.E.; Hussey, G.A.; Sullivan, A.P.; Casanovas-Massana, A.; Noval, M.G.; Valero-Jimenez, A.M.; Gago, J.; et al. Gut microbiome dysbiosis during COVID-19 is associated with increased risk for bacteremia and microbial translocation. Res. Sq. 2021. [Google Scholar] [CrossRef]
  40. Lim, W.W.; Mak, L.; Leung, G.M.; Cowling, B.J.; Peiris, M. Comparative immunogenicity of mRNA and inactivated vaccines against COVID-19. Lancet Microbe 2021, 2, e423. [Google Scholar] [CrossRef]
  41. Nordström, P.; Ballin, M.; Nordström, A. Risk of infection, hospitalisation, and death up to 9 months after a second dose of COVID-19 vaccine: A retrospective, total population cohort study in Sweden. Lancet 2022, 399, 814–823. [Google Scholar] [CrossRef] [PubMed]
  42. Divo, M.J.; Martinez, C.H.; Mannino, D.M. Ageing and the epidemiology of multimorbidity. Eur. Respir. J. 2014, 44, 1055–1068. [Google Scholar] [CrossRef] [Green Version]
  43. Mahmoud, M.; Carmisciano, L.; Tagliafico, L.; Muzyka, M.; Rosa, G.; Signori, A.; Bassetti, M.; Nencioni, A.; Monacelli, F.; Group, G.S. Patterns of Comorbidity and In-Hospital Mortality in Older Patients With COVID-19 Infection. Front. Med. 2021, 8, 726837. [Google Scholar] [CrossRef] [PubMed]
  44. Yang, L.; Bajinka, O.; Jarju, P.O.; Tan, Y.; Taal, A.M.; Ozdemir, G. The varying effects of antibiotics on gut microbiota. AMB Express 2021, 11, 116. [Google Scholar] [CrossRef] [PubMed]
  45. Panda, S.; El Khader, I.; Casellas, F.; Lopez Vivancos, J.; Garcia Cors, M.; Santiago, A.; Cuenca, S.; Guarner, F.; Manichanh, C. Short-term effect of antibiotics on human gut microbiota. PLoS ONE 2014, 9, e95476. [Google Scholar] [CrossRef]
  46. Rea, M.C.; Dobson, A.; O’Sullivan, O.; Crispie, F.; Fouhy, F.; Cotter, P.D.; Shanahan, F.; Kiely, B.; Hill, C.; Ross, R.P. Effect of broad- and narrow-spectrum antimicrobials on Clostridium difficile and microbial diversity in a model of the distal colon. Proc. Natl. Acad. Sci. USA 2011, 108, 4639–4644. [Google Scholar] [CrossRef]
  47. Zhang, L.; Huang, Y.; Zhou, Y.; Buckley, T.; Wang, H.H. Antibiotic administration routes significantly influence the levels of antibiotic resistance in gut microbiota. Antimicrob. Agents Chemother. 2013, 57, 3659–3666. [Google Scholar] [CrossRef] [Green Version]
  48. De Maria, L.; Sponselli, S.; Caputi, A.; Pipoli, A.; Giannelli, G.; Delvecchio, G.; Zagaria, S.; Cavone, D.; Stefanizzi, P.; Bianchi, F.P.; et al. Comparison of Three Different Waves in Healthcare Workers during the COVID-19 Pandemic: A Retrospective Observational Study in an Italian University Hospital. J. Clin. Med. 2022, 11, 3074. [Google Scholar] [CrossRef]
Figure 1. Study timeframe.
Figure 1. Study timeframe.
Vaccines 11 01341 g001
Table 1. Baseline characteristics of pre-vaccination antibiotic users and non-users after propensity score matching.
Table 1. Baseline characteristics of pre-vaccination antibiotic users and non-users after propensity score matching.
Pre-Vaccination
Antibiotic Non-Users
(n = 171,169)
Pre-Vaccination
Antibiotic Users
(n = 171,169)
SMD
Age, years (mean (SD))57.21 (18.2)57.60 (19.1)0.021
Sex, male (%)76,808 (44.9)77,638 (45.4)0.010
Charlson Comorbidity Index (mean (SD))0.40 (0.8)0.41 (0.8)0.019
Vaccine platform—CoronaVac (%)91,576 (53.5)93,168 (54.4)0.019
Received 3rd dose (%)1.00 (0.0)1.00 (0.0)<0.001
Comorbidities—no. (%)
Hypertension 51,436 (30.0)51,294 (30.0)0.002
Diabetes mellitus 26,124 (15.3)26,414 (15.4)0.005
Dyslipidemia 29,851 (17.4)29,351 (17.1)0.008
Cardiovascular diseases 57,263 (33.5)57,375 (33.5)0.001
Respiratory diseases 8634 (5.0)9169 (5.4)0.014
Obesity diagnosis 8377 (4.9)8626 (5.0)0.007
Smoking 2100 (1.2)2279 (1.3)0.009
Alcohol use disorders 1047 (0.6)1066 (0.6)0.001
Ulcers 3569 (2.1)3921 (2.3)0.014
Moderate-severe liver disease 334 (0.2)372 (0.2)0.005
Chronic renal failure 3358 (2.0)3459 (2.0)0.004
Medication use in past 6 months—no. (%)
ACEIs15,317 (8.9)15,693 (9.2)0.008
ARBs18,010 (10.5)18,074 (10.6)0.001
Metformin21,302 (12.4)21,316 (12.5)<0.001
Lipid lowering agents 46,982 (27.4)46,076 (26.9)0.012
Antiplatelets 23,053 (13.5)23,134 (13.5)0.001
NSAIDs 28,861 (16.9)26,385 (15.4)0.039
Oral anticoagulants 4739 (2.8)4897 (2.9)0.006
Steroids 3478 (2.0)4104 (2.4)0.025
Antidepressants 12,873 (7.5)12,939 (7.6)0.001
Antiviral drugs3214 (1.9)3231 (1.9)0.001
PPIs36,374 (21.3)36,571 (21.4)0.003
H2RAs41,112 (24.0)38,394 (22.4)0.038
SMD: standardized mean difference, SD: standard deviation, ACEIs: angiotensin-converting enzyme inhibitors, ARBs: angiotensin receptor blockers, NSAIDs: non-steroidal anti-inflammatory drugs, PPIs: proton pump inhibitors, H2RAs: H2 receptor antagonists.
Table 2. Association between pre-vaccination antibiotic use and COVID-19 outcomes after three doses of BNT162b2/CoronaVac.
Table 2. Association between pre-vaccination antibiotic use and COVID-19 outcomes after three doses of BNT162b2/CoronaVac.
EventsPerson-DaysNo. of PersonsIncidence Rate
(per 100,000 Person-Days)
Adjusted IRR (95% CI)
COVID-19
Never users 21,11066,365,213171,16931.80883-
Pre-vaccination antibiotics users24,06365,191,587171,16936.91121.16 (1.14–1.18)
COVID-19-related hospitalization
Never users 155070,426,545171,1692.200875-
Pre-vaccination antibiotics users288969,621,220171,1694.1495971.75 (1.65–1.86)
Severe COVID-19 (ICU admission/ventilatory support/death)
Never users 7870,655,136171,1690.110395-
Pre-vaccination antibiotics users13870,067,227171,1690.1969541.60 (1.21–2.11)
COVID-19-related death
Never users 2170,665,654171,1690.029717-
Pre-vaccination antibiotics users6670,080,467171,1690.0941772.56 (1.56–4.20)
IRR: incidence rate ratio; CI: confidence interval.
Table 3. Association between cumulative duration of pre-vaccination antibiotic exposure and COVID-19, hospitalization and severe COVID-19.
Table 3. Association between cumulative duration of pre-vaccination antibiotic exposure and COVID-19, hospitalization and severe COVID-19.
Cumulative Duration
(within 180 Days
Pre-Vaccination)
EventsPerson-DaysPersonsIncidence Rate
(per 100,000 Person-Days)
Adjusted IRR
(95% CI)
COVID-19 infection
0 days (non-users)21,11066,365,213171,16931.80883-
1–7 days16,91147,596,958122,11235.529581.140 (1.117–1.164)
≥8 days715217,594,62949,05740.648771.212 (1.180–1.246)
COVID-19 hospitalisation
0 days (non-users)155070,426,545171,1692.200875-
1–7 days164150,791,737122,1123.230841.572 (1.466–1.685)
≥8 days124818,829,48349,0576.6279042.070 (1.919–2.232)
Severe COVID-19
0 days (non-users)7870,655,136171,1690.110395-
1–7 days7851,044,140122,1120.1528091.485 (1.083–2.035)
≥8 days6019,023,08749,0570.3154061.772 (1.258–2.496)
COVID-19 mortality
0 days (non-users)2170,665,654171,1690.029717-
1–7 days4051,051,180122,1120.0783532.626 (1.543–4.469)
≥8 days2619,029,28749,0570.1366312.459 (1.373–4.403)
IRR: incidence rate ratio; CI: confidence interval.
Table 4. Association between pre-vaccination antibiotic use and COVID-19 outcomes, stratified by vaccine platform, age, sex, and Charlson Comorbidity Index.
Table 4. Association between pre-vaccination antibiotic use and COVID-19 outcomes, stratified by vaccine platform, age, sex, and Charlson Comorbidity Index.
Pre-Vaccination Antibiotic UseAdjusted Incidence Rate Ratio (95% CI)
BNT162b2CoronaVacAge < 60Age ≥ 60MaleFemaleCCI 0CCI ≥ 1
COVID-19
1.17
(1.13–1.20)
1.15
(1.13–1.18)
1.15
(1.12–1.18)
1.18
(1.15–1.21)
1.15
(1.12–1.19)
1.17
(1.14–1.20)
1.14
(1.12–1.16)
1.22
(1.17–1.26)
COVID-19-related hospitalization
1.69
(1.48–1.92)
1.76
(1.64–1.89)
1.63
(1.40–1.89)
1.76
(1.64–1.88)
1.74
(1.60–1.90)
1.76
(1.61–1.93)
1.73
(1.57–1.90)
1.75
(1.61–1.89)
Severe COVID-19
1.50
(0.83–2.72)
1.62
(1.18–2.22)
1.80
(0.71–4.54)
1.57
(1.17–2.11)
1.65
(1.14–2.38)
1.56
(1.01–2.40)
0.98
(0.59–1.62)
1.94
(1.38–2.74)
COVID-19-related mortality
2.02
(0.70–5.85)
2.70
(1.54–4.73)
-2.53
(1.54–4.15)
2.71
(1.41–5.22)
2.30
(1.07–4.90)
2.18
(0.84–5.64)
2.66
(1.49–4.76)
CI: confidence interval; CCI: Charlson Comorbidity Index.
Table 5. Association between nature of the antibiotics and COVID-19 outcomes after three doses of BNT162b2/CoronaVac.
Table 5. Association between nature of the antibiotics and COVID-19 outcomes after three doses of BNT162b2/CoronaVac.
EventsPerson-DaysNo. of PersonsIncidence Rate
(per 100,000 Person-Days)
Adjusted IRR
(95% CI)
COVID-19 infection
Anti-aerobic vs. anti-anaerobic
Never users 21,11066,365,213171,16931.80883-
Anti-aerobic352710,273,82926,36334.329951.11 (1.07–1.15)
Anti-anaerobic16,87945,822,622119,73236.835521.16 (1.14–1.19)
Both36579,095,13625,07440.20831.21 (1.16–1.25)
Narrow- vs. broad-spectrum
Never users 21,11066,365,213171,16931.80883-
Narrow-spectrum15884,630,58311,61334.293741.14 (1.09–1.20)
Broad-spectrum20,27654,929,426144,53436.912821.16 (1.14–1.18)
Both21995,631,57815,02239.047671.19 (1.14–1.24)
Intravenous vs. oral
Never users21,11066,365,213171,16931.80883-
Oral22,44761,588,197159,48736.446921.16 (1.14–1.18)
Intravenous302704,960235942.839311.15 (1.02–1.29)
Both13142,898,430932345.334891.24 (1.17–1.31)
COVID-19-related hospitalization
Anti-aerobic vs. anti-anaerobic
Never users 155070,426,545171,1692.200875-
Anti-aerobic36810,926,34526,3633.3680061.65 (1.47–1.85)
Anti-anaerobic193248,955,027119,7323.9464791.71 (1.60–1.83)
Both5899,739,84825,0746.0473221.98 (1.80–2.18)
Narrow- vs. broad-spectrum
Never users 155070,426,545171,1692.200875-
Narrow-spectrum1064,933,94911,6132.1483811.32 (1.08–1.61)
Broad-spectrum251658,654,25214,45344.2895441.77 (1.67–1.89)
Both2676,033,01915,0224.4256451.77 (1.55–2.02)
Intravenous vs. oral
Never users155070,426,545171,1692.200875-
Oral237365,784,698159,4873.6072221.66 (1.56–1.77)
Intravenous86749,421235911.475531.90 (1.53–2.36)
Both4303,087,101932313.928932.49 (2.23–2.78)
Severe COVID-19 (ICU admission/ventilatory support/death)
Anti-aerobic vs. anti-anaerobic
Never users 7870,655,136171,1690.110395-
Anti-aerobic1710,982,96526,3630.1547851.49 (0.88–2.53)
Anti-anaerobic10049,255,135119,7320.2030251.70 (1.26–2.29)
Both219,829,12725,0740.2136511.28 (0.79–2.08)
Narrow- vs. broad-spectrum
Never users 7870,655,136171,1690.110395-
Narrow-spectrum34,950,89711,6130.0605950.79 (0.25–2.51)
Broad-spectrum12559,041,017144,5340.2117171.67 (1.26–2.22)
Both106,075,31315,0220.1646011.28 (0.66–2.47)
Intravenous vs. oral
Never users7870,655,136171,1690.110395-
Oral11266,147,965159,4870.1693171.53 (1.14–2.04)
Intravenous5763,06423590.6552531.92 (0.77–4.76)
Both213,156,19893230.6653572.04 (1.25–3.34)
COVID-19-related death
Anti-aerobic vs. anti-anaerobic
Never users 2170,665,654171,1690.029717-
Anti-aerobic710,984,95026,3630.0637242.24 (0.95–5.27)
Anti-anaerobic5149,263,702119,7320.1035242.89 (1.73–4.83)
Both89,831,81525,0740.0813681.59 (0.70–3.61)
Narrow- vs. broad-spectrum
Never users 2170,665,654171,1690.029717-
Narrow-spectrum24,951,04611,6130.0403962.19 (0.51–9.39)
Broad-spectrum5959,053,118144,5340.099912.60 (1.57–4.30)
Both56,076,30315,0220.0822872.31 (0.87–6.15)
Intravenous vs. oral
Never users2170,665,654171,1690.029717-
Oral5366,158,741159,4870.080112.51 (1.51–4.17)
Intravenous3763,47123590.3929423.23 (0.95–10.93)
Both103,158,25593230.3166312.72 (1.26–5.86)
IRR: incidence rate ratio; CI: confidence interval.
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

Cheung, K.S.; Yan, V.K.C.; Lam, L.K.; Ye, X.; Hung, I.F.N.; Chan, E.W.; Leung, W.K. Antibiotic Use Prior to COVID-19 Vaccine Is Associated with Higher Risk of COVID-19 and Adverse Outcomes: A Propensity-Scored Matched Territory-Wide Cohort. Vaccines 2023, 11, 1341. https://doi.org/10.3390/vaccines11081341

AMA Style

Cheung KS, Yan VKC, Lam LK, Ye X, Hung IFN, Chan EW, Leung WK. Antibiotic Use Prior to COVID-19 Vaccine Is Associated with Higher Risk of COVID-19 and Adverse Outcomes: A Propensity-Scored Matched Territory-Wide Cohort. Vaccines. 2023; 11(8):1341. https://doi.org/10.3390/vaccines11081341

Chicago/Turabian Style

Cheung, Ka Shing, Vincent K. C. Yan, Lok Ka Lam, Xuxiao Ye, Ivan F. N. Hung, Esther W. Chan, and Wai K. Leung. 2023. "Antibiotic Use Prior to COVID-19 Vaccine Is Associated with Higher Risk of COVID-19 and Adverse Outcomes: A Propensity-Scored Matched Territory-Wide Cohort" Vaccines 11, no. 8: 1341. https://doi.org/10.3390/vaccines11081341

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