Next Article in Journal
Balancing Functional Tradeoffs between Protein Stability and ACE2 Binding in the SARS-CoV-2 Omicron BA.2, BA.2.75 and XBB Lineages: Dynamics-Based Network Models Reveal Epistatic Effects Modulating Compensatory Dynamic and Energetic Changes
Next Article in Special Issue
Assessing Pulmonary Epithelial Damage in Hantavirus Cardiopulmonary Syndrome: Challenging the Predominant Role of Vascular Endothelium through sRAGE as a Potential Biomarker
Previous Article in Journal
The Role of CTLA-4 in T Cell Exhaustion in Chronic Hepatitis B Virus Infection
Previous Article in Special Issue
The Flavonoid Cyanidin Shows Immunomodulatory and Broad-Spectrum Antiviral Properties, Including SARS-CoV-2
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Impact of Administering Intravenous Azithromycin within 7 Days of Hospitalization for Influenza Virus Pneumonia: A Propensity Score Analysis Using a Nationwide Administrative Database

1
Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
2
Department of Preventive Medicine and Community Health, University of Occupational and Environmental Health, Japan, Kitakyushu 807-0804, Japan
3
Department of Infectious Diseases, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
4
Department of Nursing, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
5
Department of Environmental Epidemiology, Institute of Industrial Ecological Science, University of Occupational and Environmental Health, Japan, Kitakyushu 807-0804, Japan
6
Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu 807-0804, Japan
7
Department of Health Policy and Informatics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan, Tokyo 113-8519, Japan
*
Author to whom correspondence should be addressed.
Viruses 2023, 15(5), 1142; https://doi.org/10.3390/v15051142
Submission received: 16 April 2023 / Revised: 8 May 2023 / Accepted: 9 May 2023 / Published: 10 May 2023
(This article belongs to the Special Issue Acute Lung Injury Induced by Viral Infection)

Abstract

:
The potential antimicrobial and anti-inflammatory effectiveness of azithromycin against severe influenza is yet unclear. We retrospectively investigated the effect of intravenous azithromycin administration within 7 days of hospitalization in patients with influenza virus pneumonia and respiratory failure. Using Japan’s national administrative database, we enrolled and classified 5066 patients with influenza virus pneumonia into severe, moderate, and mild groups based on their respiratory status within 7 days of hospitalization. The primary endpoints were total, 30-day, and 90-day mortality rates. The secondary endpoints were the duration of intensive-care unit management, invasive mechanical ventilation, and hospital stay. The inverse probability of the treatment weighting method with estimated propensity scores was used to minimize data collection bias. Use of intravenous azithromycin was proportional to the severity of respiratory failure (mild: 1.0%, moderate: 3.1%, severe: 14.8%). In the severe group, the 30-day mortality rate was significantly lower with azithromycin (26.49% vs. 36.65%, p = 0.038). In the moderate group, the mean duration of invasive mechanical ventilation after day 8 was shorter with azithromycin; there were no significant differences in other endpoints between the severe and moderate groups. These results suggest that intravenous azithromycin has favorable effects in patients with influenza virus pneumonia using mechanical ventilation or oxygen.

1. Introduction

Seasonal influenza viruses cause a seasonal epidemic, with around 290,000 to 640,000 respiratory deaths annually [1,2,3]. In contrast to those during the coronavirus disease (COVID-19) pandemic period, the number of patients with influenza has recently decreased. However, the number of patients with influenza increased in some areas, and we should be concerned about pandemic influenza and the possibility of a “twindemic” with COVID-19 [4,5,6]. Severe cases of influenza, similar to COVID-19, can cause excessive inflammation, resulting in acute respiratory distress syndrome (ARDS) and death [7]. The clinical efficacy of immunosuppressive therapy such as steroids, Janus kinase inhibitors, and anti-interleukin (IL)-6 antibody therapy has been shown in COVID-19 patients with respiratory failure, renewing interest in their efficacy in viral infection [8,9,10]. However, although antiviral agents have been established for influenza, treatment for excessive inflammation and severe respiratory failure has not been established [11,12]. Moreover, the effects of corticosteroids have been reported to worsen prognosis in influenza [13,14]. Thus, establishing other beneficial immunomodulatory treatments for patients with severe influenza is necessary.
Azithromycin, a macrolide antibiotic, has favorable effects on critical respiratory diseases owing to its antimicrobial and anti-inflammatory effects. A meta-analysis showed that beta-lactam plus macrolide outperformed beta-lactam alone, with an odds ratio (OR) of 0.80 for all-cause death, for community-acquired pneumonia (CAP) [15]. A secondary analysis of a multicenter randomized controlled trial (RCT) of patients with ARDS showed that macrolide use was associated with lower 180-day mortality (hazard ratio (HR), 0.46) [16]. Furthermore, an RCT including patients with influenza showed that inflammatory cytokine levels decreased more rapidly in the oseltamivir/azithromycin group than in the oseltamivir monotherapy group [17]. Therefore, azithromycin may have favorable effects in patients with severe influenza virus pneumonia, but its effectiveness has not yet been demonstrated.
As validation by a large prospective RCT is useful but time-consuming and costly, we retrospectively evaluated the effects of intravenous azithromycin administration in influenza virus pneumonia patients with respiratory failure, using the Diagnostic Procedure Combination (DPC) database, a nationwide Japanese administrative database. Although the DPC database is limited by being a retrospective study, it has advantages such as low financial and time costs, real-world data are available, and long-term drug use and evaluation can be easily performed. We used the inverse probability of treatment weighting (IPTW) method with estimated propensity scores to minimize the bias introduced by baseline characteristics in retrospectively collected data in the azithromycin and non-macrolide groups.

2. Materials and Methods

2.1. Study Design

The research methodologies used in this retrospective observational study were in accordance with the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) Statement [18].

2.2. Ethics

The requirement for informed consent was waived owing to the retrospective nature of the study and removal of any personally identifiable information from the extracted data. This study was approved by the Ethics Committee of Medical Research, University of Occupational and Environmental Health, Japan (approval number: R2-007), and conducted according to the guidelines of the Declaration of Helsinki.

2.3. Data Source

The DPC is a Japanese case-mix patient classification system launched in 2002 for payment management and the modernization of the healthcare system [19]. It covered approximately 80% of all acute care inpatient hospitalizations in Japan in 2020 [13,20,21]. The database contains the following patient details: age, sex, diagnosis, comorbidities at admission and during hospitalization (coded according to the International Classification of Diseases, Tenth Revision (ICD-10)), state of consciousness according to the Japan Coma Scale, medical procedures, medications, intensive care unit (ICU) admission, interventional procedures (including hemodialysis, mechanical ventilation, and administration of heart–lung medication), length of hospital stay, and discharge status (including in-hospital death). Although the DPC database does not include influenza virus types, information on influenza virus types circulating seasonally in Japan is available in the Japanese National Institute of Infectious Diseases’ database (https://www.niid.go.jp/niid/en/influenza-e.html (accessed on 7 May 2023)). According to the data presented in this database, the respective rates of A/H1, A/H3, and B viruses were 4%, 78%, and 18% during 2016/2017 (September 2016 to August 2017) and 23%, 32%, and 45% during 2017/2018 (September 2017 to August 2018). Thus, A/H1, A/H3, and B viruses should be the major virus types present in our study from April 2016 to March 2018.

2.4. Patient Selection and Definitions

We selected patients with influenza virus pneumonia (ICD-10 code J10.0 or J11.0) or influenza (ICD-10 code J10.1 or J10.8) with ARDS (J80). Data of patients admitted between April 2016 and March 2018 who met the criteria were extracted from the DPC database. To simply observe the effects of administering intravenous azithromycin within 7 days of hospitalization on standard therapy, patients who received macrolide antibiotics other than intravenous and oral erythromycin, clarithromycin, josamycin, roxithromycin and spiramycin, and oral azithromycin within 7 days of hospitalization were excluded from this analysis. Patients who did not use oxygen therapy were classified into the oxygen-free group (mild), those who used oxygen therapy or nasal high-flow (NHF) oxygen therapy were categorized into the oxygen group (moderate), and those who used non-invasive positive pressure ventilation (NPPV) or invasive mechanical ventilation (IMV) were classified into the ventilator group (severe) within 7 days of hospitalization. The patients were also divided into the azithromycin and non-macrolide groups, with patients treated with intravenous azithromycin within 7 days of hospitalization being included in the azithromycin group.

2.5. Variables

The following variables were considered confounding factors: age (in years), sex, emergency admission, emergency transport, home healthcare, hospital volume (number of patients who met the inclusion criteria between April 2016 and March 2018), smoking, diagnosis of asthma, cancer, cardiovascular disease (diagnosis of acute myocardial infarction, cardiac valvular disease, cardiomyopathy, or pulmonary embolism), cerebrovascular disease (diagnosis of cerebral hemorrhage, subarachnoid hemorrhage, or cerebral infarction), chronic kidney disease, chronic respiratory failure, chronic obstructive pulmonary disease, diabetes mellitus, liver dysfunction (diagnosis of hepatitis, liver cirrhosis, or liver failure), neurological dysfunction (Japan Coma Scale score at admission ≥ 100 (indicating a response by closing eyes, no verbal response, and movement in response to pain) [22]), heart failure, hypertension, interstitial lung diseases, medication use (albumin, antithrombin III, heparin, immunoglobulin, insulin, transfusion of platelets or red blood cells, recombinant human soluble thrombomodulin (rhTM), sivelestat, vasopressors, oseltamivir, steroid, laninamivir, peramivir, carbapenem, cephalosporin (1st–4th generation), clindamycin, minocycline, new quinolone, penicillin antibiotics, or anti-methicillin-resistant Staphylococcus aureus (MRSA) drugs), emergency hemodialysis or maintenance hemodialysis, number of IMV wearing days, NPPV, NHF oxygen therapy, oxygen therapy, use of polymyxin B-immobilized fiber column, and ICU admission within 7 days of hospitalization. Intravenous administration of antibiotics was considered as a covariate; oral administration was not considered.

2.6. Outcomes

The primary endpoint was mortality (in-hospital mortality, 30-day mortality, and 90-day mortality). The secondary endpoints were the duration of ICU management, mechanical ventilation, and hospital stay throughout the hospitalization period.

2.7. Statistical Analyses

The propensity score was calculated using a logistic model with baseline variables that could affect azithromycin administration, including all the variables described above. The C-statistic (area under the operating characteristic curve) was used to evaluate the goodness of fit. Subsequently, we adjusted the covariates and compared the outcomes between the azithromycin and non-macrolide groups using the IPTW method [23]. Compared to other propensity score analyses, the IPTW method has the advantages of a larger effective sample size and lower impact from strong residual bias that can occur in sample selection by matching [24]. The covariates before adjustment were evaluated using the chi-squared (χ2) test for categorical variables and unpaired t-test for continuous variables. All analyses were conducted using STATA/IC 14.0 (StataCorp, College Station, TX, USA). Statistical significance was set at p < 0.05.

3. Results

Figure 1 depicts the flowchart of the patient selection. Data of 5359 patients with influenza virus pneumonia or influenza with ARDS were extracted from the DPC database as described in the Section 2. We excluded 293 patients treated with macrolides other than intravenous azithromycin, and 5066 patients were therefore included in this study. They were classified into three groups based on their respiratory status: 1720 mild patients (33.9%) without oxygen therapy, 2739 moderate patients (54.1%) with NHF oxygen therapy or oxygen therapy, and 607 severe patients (12.0%) with mechanical ventilation.

3.1. Mild Group

Of the 1720 mild patients, 17 (1.0%) were treated with intravenous azithromycin at 500 mg/day for >1 day, and 1703 (99.0%) were not treated with macrolides within 7 days of hospitalization. Cardiovascular disease, cerebrovascular disease, neurological dysfunction, maintenance hemodialysis, antithrombin III, rhTM, sivelestat, cephalosporin (1st, 2nd, and 4th generation), clindamycin, minocycline, anti-MRSA drugs, number of IMV weaning days, NPPV, NHF oxygen therapy, oxygen therapy, use of polymyxin B-immobilized fiber column, and ICU admission rate were excluded from the variables because there were no patients to include. Subsequently, we used the IPTW method with estimated propensity scores; the C-statistic of the propensity score was 0.823. However, we were unable to adjust for variables and outcomes between the azithromycin and non-macrolide groups using the IPTW method because of the small number of patients treated with intravenous azithromycin.

3.2. Moderate Group

Of the 2739 moderate patients, 85 (3.1%) were treated with intravenous azithromycin at 500 mg/day for >1 days, and 2654 (96.9%) were not treated with macrolides. The C-statistic for the propensity score was 0.804. Table 1 presents the patients’ baseline characteristics. Platelet transfusion, antithrombin III, sivelestat, immunoglobulin, polymyxin B-immobilized fiber column, NPPV, and IMV weaning days were excluded from the variables because there were no patients to include. The baseline variables including emergency admission (78.8% vs. 68.2%, p = 0.037), smoking (50.6% vs. 32.7%, p = 0.001), liver dysfunction (4.7% vs. 1.8%, p = 0.049), cancer (15.3% vs. 8.9%, p = 0.043), interstitial lung diseases (27.1% vs. 12.3%, p < 0.001), vasopressor use (11.8% vs. 4.5%, p = 0.002), emergency hemodialysis (2.4% vs. 0.4%, p = 0.010), albumin (4.7% vs. 1.0%, p = 0.002), heparin (14.1% vs. 5.3%, p = 0.001), insulin (25.9% vs. 12.3%, p < 0.001), rhTM (5.9% vs. 0.7%, p < 0.001), peramivir (71.8% vs. 59.0%, p = 0.018), carbapenem (20.0% vs. 8.9%, p < 0.001), cephalosporin (3rd generation) (44.7% vs. 31.7%, p = 0.012), anti-MRSA drugs (3.5% vs. 0.7%, p = 0.003), and steroid use (29.4% vs. 20.1%, p = 0.036) differed significantly between the groups before adjustment. After adjustment, the baseline characteristics were similar between the two groups, except for hospital volume (10.97 ± 1.3 vs. 14.55 ± 0.5, p = 0.010), laninamivir (0.47% vs. 2.55%, p < 0.001), and cephalosporins (2nd generation) (0.47% vs. 1.71%, p = 0.019).
Table 2 presents the primary and secondary endpoints in the moderate group assessed using the IPTW method with estimated propensity scores. There were no significant differences between the azithromycin and non-macrolide groups regarding total mortality (16.56% vs. 11.45%, p = 0.516), 30-day mortality (14.89% vs. 9.23%, p = 0.477), 90-day mortality (15.06% vs. 11.16%, p = 0.622), duration of ICU management (0.050 ± 0.03 days vs. 0.069 ± 0.01 days, p = 0.553), and hospital stay (17.55 ± 2.9 days vs. 17.95 ± 0.4 days, p = 0.891). However, the mean IMV duration after day 8 was shorter in the azithromycin group than in the non-macrolide group (0.028 ± 0.02 days vs. 0.154 ± 0.05 days, p = 0.026).

3.3. Severe Group

Of the 607 severe patients, 90 (14.8%) were treated with intravenous azithromycin at 500 mg/day for >1 d, and 517 (85.2%) were not treated with macrolides within 7 days of hospitalization. The C-statistic for the propensity score was 0.785. Table 3 presents the baseline characteristics of the patients before and after adjusting for confounders. Patients taking cephalosporins (2nd generation) were excluded from the study because there were no patients. The baseline variables including albumin (34.4% vs. 20.5%, p = 0.004), antithrombin III (12.2% vs. 5.0%, p = 0.008), heparin (62.2% vs. 46.6%, p = 0.006), immunoglobulin (14.4% vs. 6.2%, p = 0.006), insulin (57.8% vs. 37.7%, p < 0.001), red blood cell transfusion (22.2% vs. 11.6%, p = 0.006), rhTM (20.0% vs. 7.9%, p < 0.001), peramivir (86.7% vs. 74.1%, p = 0.010), carbapenem (41.1% vs. 27.7%, p = 0.010), anti-MRSA drug use (15.6% vs. 5.4%, p < 0.001), steroid use (62.2% vs. 39.5%, p < 0.001), number of IMV days (5.18 vs. 4.21, p < 0.001), and ICU admission rate (38.9% vs. 28.4%, p = 0.046) differed significantly between the groups before adjustment. However, after adjusting for confounders, the baseline patient characteristics were similar between the azithromycin and non-macrolide groups, except for maintenance hemodialysis (0.80% vs. 2.78%, p = 0.006).
Table 4 present the results of the primary and secondary endpoints in the severe group assessed using the IPTW method with estimated propensity scores. Patients treated with azithromycin had significantly lower 30-day mortality rates than those not treated with macrolides (26.49% vs. 36.65%, p = 0.038). There were no significant differences between the azithromycin and non-macrolide groups regarding total mortality (33.78% vs. 40.97%, p = 0.194), 90-day mortality (33.78% vs. 40.37%, p = 0.234), duration of ICU management (2.96 ± 0.4 days vs. 2.59 ± 0.2 days, p = 0.407), IMV (10.82 ± 1.2 days vs. 11.69 ± 0.8 days, p = 0.534), and hospital stay (28.32 ± 2.3 days vs. 27.33 ± 1.3 days, p = 0.699).

4. Discussion

In this study, we investigated the effects of administering intravenous azithromycin within 7 days of hospitalization using data from 5066 patients with influenza virus pneumonia registered in the nationwide administrative database of Japan. Azithromycin was used in 192 (3.79%) of the 5066 patients based on real-world data. The use of azithromycin was proportional to the severity of respiratory failure (mild group, 1.0%; moderate group, 3.1%; and severe group, 14.8%).
This study also demonstrated that intravenous azithromycin improved the duration of IMV in patients in the moderate group and 30-day mortality in patients in the severe group. There were no significant differences between the groups regarding total mortality and 90-day mortality in the severe group, but the azithromycin group had numerically lower results. It has been suggested that azithromycin may be effective in patients with more severe influenza pneumonia.
The efficacy of azithromycin for critical lung diseases such as CAP, ARDS, and acute exacerbation of interstitial pneumonia has been reported. In the United States, the most common risk factor for ARDS is severe sepsis with a suspected pulmonary source (46%) [25]. In a prospective cohort study of patients with severe CAP, the combination of beta-lactam and azithromycin was also associated with a significantly lower 30-day mortality (OR = 0.12, 95% confidence interval (CI): 0.007–0.57) compared with beta-lactam alone [26]. A meta-analysis of beta-lactam and macrolide combination therapy for CAP discovered that azithromycin-based combinations had higher clinical success rates than clarithromycin-based combinations (87.55% vs. 75.42%) [27]. A multicenter, multinational observational study of patients with severe CAP suggested that the combination of aspirin with CAM or azithromycin was associated with the 30-day survival rate (HR, 0.71, 95% CI: 0.58–0.88, p = 0.002) [28]. Influenza A viruses have also been reported to be a common cause of viral pneumonia and ARDS in adult patients, with ARDS observed in 39.4% of severe influenza cases [7]. In a report that assessed only patients with ARDS associated with viral pneumonia, 17% had ARDS associated with the influenza virus [29]. A secondary analysis of a multicenter RCT of patients with ARDS showed that macrolide use, including azithromycin, was associated with a lower 180-day mortality (HR, 0.46, 95% CI: 0.23–0.92, p = 0.028) and shorter time to successful weaning from mechanical ventilation (HR, 1.93, 95% CI: 1.18–3.17, p = 0.009) [16]. In a retrospective cohort evaluation of azithromycin’s effect on moderate or severe ARDS, azithromycin use was associated with a significant improvement in the 90-day survival rate (HR, 0.49, 95% CI: 0.27–0.87, p = 0.015) and shorter time to successful weaning from mechanical ventilation (HR, 1.74, 95% CI: 1.07–2.81, p = 0.026) [30]. Moreover, azithromycin significantly improved mortality in two studies on interstitial pneumonia. In a prospective, open-label study of acute exacerbations of chronic fibrosing interstitial pneumonia, the mortality rate was significantly lower in the azithromycin group than in the fluoroquinolone group (HR, 0.17, 95% CI: 0.05–0.61) [31]. In a retrospective study of acute exacerbations of idiopathic pulmonary fibrosis, the mortality rate was significantly lower in patients treated with azithromycin than in those treated with fluoroquinolones (26% vs. 70%, p < 0.001) [32]. These results suggest that the favorable effects of azithromycin on critical respiratory diseases may be due to its antimicrobial and anti-inflammatory effects.
The efficacy of macrolide antibiotics in patients with influenza has been reported in a few RCTs. Clarithromycin, a macrolide, in combination with naproxen and oseltamivir, has been associated with reduced 30-day mortality (p = 0.01), ICU admission rate (p = 0.009), and hospital stay (p < 0.0001) compared with oseltamivir alone [33]. The combination of clarithromycin and oseltamivir has also been shown to be more effective in improving clinical symptoms, such as rhinorrhea and fever, compared to oseltamivir alone [34,35]. In an RCT of patients with influenza, combination therapy with oseltamivir and azithromycin showed significant improvement in the levels of inflammatory cytokines, such as IL-6, CXCL8/IL-8, IL-17, CXCL9/MIG, sTNFR-1, and IL-18 [17]. Another RCT showed no improvement in inflammatory cytokines but faster improvement in clinical symptoms such as fever after day 4 in the oseltamivir and azithromycin groups (p = 0.048) [36]. Our results suggest that azithromycin may have favorable effects in patients with influenza and respiratory failure, consistent with these reports’ findings. However, another observational study showed no improvements in mortality with macrolides (clarithromycin, erythromycin, and azithromycin) in patients with influenza and severe respiratory failure [37]. Therefore, further studies, including RCTs, are warranted.
Macrolides have been demonstrated to possess some anti-inflammatory activity both in vitro and in vivo. Indeed, macrolides could modulate cytokine production as well as other immunological cellular properties (e.g., chemotaxis, degranulation, oxidative burst, and even apoptosis). Importantly, with regard to infectious diseases, the clinical relevance of macrolides may be beyond their antibacterial activity. They may improve the clinical course of viral respiratory infections (respiratory syncytial virus, influenza viruses, and coronavirus) at least through indirect mechanisms relying on variable anti-inflammatory activity [38]. Azithromycin, one of the macrolide antibiotics, is a low-cost, historically safe, and globally distributed antibiotic that is practical [39]. Azithromycin has immunomodulatory activity in the acute phase of inflammation suppression and late phase of chronic inflammation elimination [40]. The half-life is approximately 50 h, which is longer than that of other antibacterial agents, and such effective levels can be maintained for several days. Therefore, early administration can be expected to maintain antibacterial and anti-inflammatory effects during the acute phase. The effects of these immunomodulatory activities are due to the inhibition of extracellular signal-regulated kinase 1/2 phosphorylation and nuclear factor kappa B activation [40]. In vitro studies have demonstrated the ability of azithromycin to reduce the production of proinflammatory cytokines, such as IL-8, IL-6, and tumor necrosis factor-α; reduce oxidative stress; and regulate T-helper functions [41].
Azithromycin has also been shown to inhibit influenza and COVID-19 activity without cytotoxicity. It may be a candidate broad-spectrum antiviral drug [42]. Azithromycin has a significant anti-inflammatory effect on influenza infections [17], and there are reports showing antiviral effects that suppress the in vitro growth of many viruses [43]. However, the efficacy of azithromycin against COVID-19 has been negated in several RCTs such as the RECOVERY and ATOMIC2 trials. In the RECOVERY trial, which compared usual care alone to usual care plus azithromycin (500 mg oral/intravenous once daily), there were no significant differences in the 28-day all-cause mortality (rate ratio = 0.97, 95% CI: 0.87–1.07, p = 0.50), duration of hospital stay (median [interquartile range], 10 days [5 to >28 days] and 11 days [5 to >28 days], respectively), or proportion of patients discharged from the hospital alive within 28 days (rate ratio = 1.04, 95% CI: 0.98–1.10, p = 0.19) [44]. In the ATOMIC2 trial, which compared azithromycin (500 mg oral once daily for 14 days) plus standard care to standard care alone in 292 outpatients with mild to moderate illness, the 28-day mortality or hospitalization rates were 10% in the azithromycin group and 12% in the standard care group (adjusted OR = 0.91, 95% CI: 0.43–1.92, p = 0.80) [45]. However, many of these studies included patients with mild to moderate illness, and few studies evaluated only severe patients with respiratory failure or ARDS. As aforementioned, azithromycin has been reported to be effective against severe lung diseases such as CAP, ARDS, and acute exacerbation of interstitial pneumonia, and its effects in these populations with mild to moderate disease may have been underestimated.
In the moderate group, azithromycin was used in combination with 3rd generation cephalosporin antibiotics (44.7%), penicillin antibiotics (37.6%), and carbapenem antibiotics (20.0%). In the severe group, 3rd generation cephalosporin antibiotics (34.4%), penicillin antibiotics (34.4%), and carbapenem antibiotics (41.1%) were used in combination, and carbapenem antibiotics were more frequently used in the severe group than in the moderate group. This included changes of antibiotics within 7 days and the use of three or more antibiotics at the same time, although azithromycin was considered to be administered in combination with other antibiotics in most patients. Similarly, antibiotics were used in most patients in the non-macrolide group. The reason for this is considered to be the effect of secondary bacterial infections associated with influenza. Approximately 23% of patients with influenza develop secondary bacterial infections associated with influenza, and secondary pneumonia complications are associated with increased mortality [46]. When the pandemic of influenza A occurred in 2009, bacterial infections occurred in approximately 30% of the patients who died of influenza [47]. According to a summary of clinical reports, 73.5% of all patients with viral pneumonias, including influenza, received prophylactic antibiotics [48]. Thus, the use of antibiotics in severe cases of pneumonia with influenza seems to be considered in the clinical setting. The Japanese guidelines for pneumonia also recommend macrolide antibiotics in combination with conventional antibiotics for severe pneumonia [49]. Streptococcus pneumoniae, Haemophilus influenzae, and Staphylococcus aureus are the most common causes [50]. Particularly, Streptococcus pneumoniae has a lethal synergy with the influenza virus [51]. In a retrospective cohort study investigating the effect of azithromycin on pneumococcal pneumonia, treatment with azithromycin was associated with a lower rate of mortality compared with other treatment groups that did not receive macrolide antibiotics (OR, 0.26, 95% CI: 0.08–0.80, p = 0.018) [52]. Data from the United States show that 34% of pneumococcal strains are penicillin insensitive (15.7% intermediate, 18.5% resistant) and the rate of macrolide resistance is 26.9–28.6%, indicating that antibiotic resistance is a problem [53]. On the other hand, azithromycin has been reported to be effective against macrolide-resistant pneumococcal pneumonia. In pneumococcal pneumonia requiring ICU management, 60.8% of patients had macrolide resistance, but azithromycin still reduced mortality (OR, 0.27, 95% CI: 0.09–0.85, p = 0.024) [54]. Azithromycin was effective in six of seven patients with high-level resistance (minimum inhibitory concentration > 256 μg/mL) [55].
Collectively, our favorable results against severe influenza might be due to the anti-inflammatory and antiviral effects of azithromycin and its antimicrobial effects in complicated bacterial infections. These effects can also be expected for macrolide antibiotics other than azithromycin, which were excluded from this study. However, further studies are needed to clarify these points [56].
Azithromycin is a very valuable antibiotic that is on the World Health Organization’s list of essential medicines and is required for the treatment of trachoma, multidrug-resistant tuberculosis, non-tuberculous mycobacteria, bacterial pneumonia, sexually transmitted diseases, and other diseases. According to Japan’s National Action Plan on Antimicrobial Resistance (https://www.mhlw.go.jp/content/10900000/000885373.pdf (accessed on 7 May 2023)), the macrolide resistance rate of Streptococcus pneumoniae was about 80% in Japan. As the results of microbiological examinations are not included in the DPC database, we could not evaluate the effects of increases in drug-resistant bacteria due to the use of azithromycin. In this study using a Japanese nationwide administrative database, azithromycin was mainly used in critical cases. The use of azithromycin was proportional to the severity of respiratory failure (mild group, 1.0%; moderate group, 3.1%; and severe group, 14.8%), and the effects were shown in the 30-day mortality rate of the severe group and the duration of invasive mechanical ventilation after day 8 of the moderate group. Accordingly, it might be better if concomitant use of azithromycin were limited to critical cases, for avoiding increases in drug-resistant germs, though further studies are needed.
This study had limitations similar to those of previous studies that used the DPC database [13,20,21]. First, this was an observational and retrospective study without randomization. We used the IPTW method with the estimated propensity score to reduce the effect of this limitation, although there were still significant differences in confounders. Such confounders included maintenance hemodialysis in the severe group and hospital volume, laninamivir, and cephalosporins (2nd generation) in the moderate group. Regarding maintenance dialysis in the severe group, azithromycin may have been avoided for use in patients with maintenance dialysis because it requires more fluid infusion, usually 500 mL at a time. Second, we could not include clinical data, such as peripheral blood laboratory findings, physiological data including vital signs, radiological findings, and mechanical ventilation settings. Third, it was difficult to analyze data including the content of previous treatments, such as the rate of multiple hospitalizations and previous history of antibiotic use, because of the difficulty in linking DPC data. Fourth, the influence of complicated bacterial infections was unknown because the results of microbiological examinations were not included in the DPC database. Thus, it was difficult to speculate whether the favorable effects of azithromycin were due to anti-inflammatory or antimicrobial effects. However, many of the patients included in this study were administered antibiotics other than azithromycin, and it can be inferred that there was no difference in the antibacterial effect between the two groups. Therefore, they may be due to the anti-inflammatory effects of azithromycin. Fifth, we were unable to evaluate adverse events of azithromycin. Azithromycin and other macrolide antibiotics cause serious adverse events, such as fatal arrhythmia and myocardial infarction, but this study could not evaluate these effects [57]. Additionally, we could not know the effects of increases in drug-resistant bacteria, as described above.
Despite these limitations, this study has the advantage of using real-world data and including a large number of patients (more than 5000) with influenza virus pneumonia and acute respiratory failure. In addition, data from April 2016 to March 2018 were not affected by the COVID-19 pandemic.

5. Conclusions

Using nationwide administrative database data, we discovered the favorable effects of intravenous azithromycin administration in patients with influenza virus pneumonia on mechanical ventilation or oxygen, based on propensity score analysis. The significance of this study is that it uses real-world data from a large number of patients with influenza virus pneumonia and acute respiratory failure, which is difficult to achieve in large prospective studies. Nevertheless, this study had some limitations owing to its retrospective and observational design, and well-designed RCTs are required to evaluate the efficacy of azithromycin further.

Author Contributions

Conceptualization, T.T. (Takatomo Tokito), T.K., K.M., N.S. and Y.F.; methodology, T.T. (Takatomo Tokito), T.K., K.M., N.S. and Y.F.; formal analysis, T.T. (Takatomo Tokito), T.K. and K.M.; investigation, T.T. (Takatomo Tokito) and T.K.; data curation, T.T. (Takatomo Tokito), T.K., K.M., K.T. and K.F.; writing—original draft preparation, T.T. (Takatomo Tokito) and T.K.; writing—review and editing, T.T. (Takatomo Tokito), T.K., K.M., K.T., D.O., H.Y., S.T., H.I., N.S., T.T. (Takahiro Takazono), Y.O., Y.I., Y.F., K.Y., S.M., K.F. and H.M.; project administration, S.M. and H.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Ministry of Health, Labour, and Welfare, Japan (grant number JP20AA2005).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee of Medical Research, University of Occupational and Environmental Health, Japan (approval number: R2-007).

Informed Consent Statement

Patient consent was waived due to the retrospective design of the study and removal of any personally identifiable information from the extracted data.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nicholson, K.G.; Wood, J.M.; Zambon, M. Influenza. Lancet 2003, 362, 1733–1745. [Google Scholar] [CrossRef] [PubMed]
  2. Shrestha, S.S.; Swerdlow, D.L.; Borse, R.H.; Prabhu, V.S.; Finelli, L.; Atkins, C.Y.; Owusu-Edusei, K.; Bell, B.; Mead, P.S.; Biggerstaff, M.; et al. Estimating the Burden of 2009 Pandemic Influenza A (H1N1) in the United States (April 2009–April 2010). Clin. Infect. Dis. 2011, 52 (Suppl. S1), S75–S82. [Google Scholar] [CrossRef] [PubMed]
  3. Iuliano, A.D.; Roguski, K.M.; Chang, H.H.; Muscatello, D.J.; Palekar, R.; Tempia, S.; Cohen, C.; Gran, J.M.; Schanzer, D.; Cowling, B.J.; et al. Estimates of Global Seasonal Influenza-associated Respiratory Mortality: A Modelling Study. Lancet 2018, 391, 1285–1300. [Google Scholar] [CrossRef] [PubMed]
  4. Krauland, M.G.; Galloway, D.D.; Raviotta, J.M.; Zimmerman, R.K.; Roberts, M.S. Impact of Low Rates of Influenza on Next-season Influenza Infections. Am. J. Prev. Med. 2022, 62, 503–510. [Google Scholar] [CrossRef]
  5. Lee, K.; Jalal, H.; Raviotta, J.M.; Krauland, M.G.; Zimmerman, R.K.; Burke, D.S.; Roberts, M.S. Estimating the Impact of Low Influenza Activity in 2020 on Population Immunity and Future Influenza Seasons in the United States. Open Forum Infect. Dis. 2021, 9, ofab607. [Google Scholar] [CrossRef] [PubMed]
  6. Del Rio, C.; Omer, S.B.; Malani, P.N. Winter of Omicron-The Evolving COVID-19 Pandemic. JAMA 2022, 327, 319–320. [Google Scholar] [CrossRef]
  7. Hong, W.C.; Sun, S.F.; Hsu, C.W.; Lee, D.L.; Lee, C.H. Clinical Characteristics and Predictors of Mortality in Critically Ill Adult Patients with Influenza Infection. Int. J. Environ. Res. Public Health 2021, 18, 3682. [Google Scholar] [CrossRef]
  8. Horby, P.; Lim, W.S.; Emberson, J.R.; Mafham, M.; Bell, J.L.; Linsell, L.; Staplin, N.; Brightling, C.; Ustianowski, A.; Elmahi, E.; et al. Dexamethasone in Hospitalized Patients with COVID-19. N. Engl. J. Med. 2021, 384, 693–704. [Google Scholar] [CrossRef] [PubMed]
  9. Kalil, A.C.; Patterson, T.F.; Mehta, A.K.; Tomashek, K.M.; Wolfe, C.R.; Ghazaryan, V.; Marconi, V.C.; Ruiz-Palacios, G.M.; Hsieh, L.; Kline, S.; et al. Baricitinib plus Remdesivir for Hospitalized Adults with COVID-19. N. Engl. J. Med. 2021, 384, 795–807. [Google Scholar] [CrossRef]
  10. Shankar-Hari, M.; Vale, C.L.; Godolphin, P.J.; Fisher, D.; Higgins, J.P.T.; Spiga, F.; Savovic, J.; Tierney, J.; Baron, G.; Benbenishty, J.S.; et al. Association Between Administration of IL-6 Antagonists and Mortality Among Patients Hospitalized for COVID-19: A Meta-Analysis. JAMA 2021, 326, 499–518. [Google Scholar] [CrossRef]
  11. de Jong, M.D.; Ison, M.G.; Monto, A.S.; Metev, H.; Clark, C.; O’Neil, B.; Elder, J.; McCullough, A.; Collis, P.; Sheridan, W.P. Evaluation of Intravenous Peramivir for Treatment of Influenza in Hospitalized Patients. Clin. Infect. Dis. 2014, 59, e172–e185. [Google Scholar] [CrossRef] [PubMed]
  12. Kohno, S.; Kida, H.; Mizuguchi, M.; Hirotsu, N.; Ishida, T.; Kadota, J.; Shimada, J. Intravenous Peramivir for Treatment of Influenza A and B Virus Infection in High-risk Patients. Antimicrob. Agents Chemother. 2011, 55, 2803–2812. [Google Scholar] [CrossRef] [PubMed]
  13. Okuno, D.; Kido, T.; Muramatsu, K.; Tokutsu, K.; Moriyama, S.; Miyamura, T.; Hara, A.; Ishimoto, H.; Yamaguchi, H.; Miyazaki, T.; et al. Impact of Corticosteroid Administration Within 7 Days of the Hospitalization for Influenza Pneumonia with Respiratory Failure: A Propensity Score Analysis Using a Nationwide Administrative Database. J. Clin. Med. 2021, 10, 494. [Google Scholar] [CrossRef] [PubMed]
  14. Ni, Y.N.; Chen, G.; Sun, J.; Liang, B.M.; Liang, Z.A. The Effect of Corticosteroids on Mortality of Patients with Influenza Pneumonia: A Systematic Review and Meta-analysis. Crit. Care 2019, 23, 99. [Google Scholar] [CrossRef] [PubMed]
  15. Horita, N.; Otsuka, T.; Haranaga, S.; Namkoong, H.; Miki, M.; Miyashita, N.; Higa, F.; Takahashi, H.; Yoshida, M.; Kohno, S.; et al. Beta-lactam Plus Macrolides or Beta-lactam Alone for Community-acquired Pneumonia: A Systematic Review and Meta-analysis. Respirology 2016, 21, 1193–1200. [Google Scholar] [CrossRef]
  16. Walkey, A.J.; Wiener, R.S. Macrolide Antibiotics and Survival in Patients with Acute Lung Injury. Chest 2012, 141, 1153–1159. [Google Scholar] [CrossRef]
  17. Lee, N.; Wong, C.K.; Chan, M.C.W.; Yeung, E.S.L.; Tam, W.W.S.; Tsang, O.T.Y.; Choi, K.W.; Chan, P.K.S.; Kwok, A.; Lui, G.C.Y.; et al. Anti-inflammatory Effects of Adjunctive Macrolide Treatment in Adults Hospitalized with Influenza: A Randomized Controlled Trial. Antivir. Res. 2017, 144, 48–56. [Google Scholar] [CrossRef]
  18. von Elm, E.; Altman, D.G.; Egger, M.; Pocock, S.J.; Gøtzsche, P.C.; Vandenbroucke, J.P. The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) Statement: Guidelines for Reporting Observational Studies. Int. J. Surg. 2014, 12, 1495–1499. [Google Scholar] [CrossRef]
  19. Hayashida, K.; Murakami, G.; Matsuda, S.; Fushimi, K. History and Profile of Diagnosis Procedure Combination (DPC): Development of a Real Data Collection System for Acute Inpatient Care in Japan. J. Epidemiol. 2021, 31, 1–11. [Google Scholar] [CrossRef]
  20. Kido, T.; Muramatsu, K.; Asakawa, T.; Otsubo, H.; Ogoshi, T.; Oda, K.; Kubo, T.; Fujino, Y.; Matsuda, S.; Mayumi, T.; et al. The Relationship Between High-dose Corticosteroid Treatment and Mortality in Acute Respiratory Distress Syndrome: A Retrospective and Observational Study Using a Nationwide Administrative Database in Japan. BMC Pulm. Med. 2018, 18, 28. [Google Scholar] [CrossRef]
  21. Kido, T.; Muramatsu, K.; Yatera, K.; Asakawa, T.; Otsubo, H.; Kubo, T.; Fujino, Y.; Matsuda, S.; Mayumi, T.; Mukae, H. Efficacy of Early Sivelestat Administration on Acute Lung Injury and Acute Respiratory Distress Syndrome. Respirology 2017, 22, 708–713. [Google Scholar] [CrossRef]
  22. Iwagami, M.; Yasunaga, H.; Doi, K.; Horiguchi, H.; Fushimi, K.; Matsubara, T.; Yahagi, N.; Noiri, E. Postoperative Polymyxin B Hemoperfusion and Mortality in Patients with Abdominal Septic Shock: A Propensity-matched Analysis. Crit. Care Med. 2014, 42, 1187–1193. [Google Scholar] [CrossRef] [PubMed]
  23. Austin, P.C. The Performance of Different Propensity Score Methods for Estimating Marginal Hazard Ratios. Stat Med. 2013, 32, 2837–2849. [Google Scholar] [CrossRef] [PubMed]
  24. Chesnaye, N.C.; Stel, V.S.; Tripepi, G.; Dekker, F.W.; Fu, E.L.; Zoccali, C.; Jager, K.J. An Introduction to Inverse Probability of Treatment Weighting in Observational Research. Clin. Kidney J. 2021, 15, 14–20. [Google Scholar] [CrossRef]
  25. Rubenfeld, G.D.; Caldwell, E.; Peabody, E.; Weaver, J.; Martin, D.P.; Neff, M.; Neff, M.; Stern, E.J.; Hudson, L.D. Incidence and Outcomes of Acute Lung Injury. N. Engl. J. Med. 2005, 353, 1685–1693. [Google Scholar] [CrossRef] [PubMed]
  26. Ito, A.; Ishida, T.; Tachibana, H.; Tokumasu, H.; Yamazaki, A.; Washio, Y. Azithromycin Combination Therapy for Community-acquired Pneumonia: Propensity Score Analysis. Sci. Rep. 2019, 9, 18406. [Google Scholar] [CrossRef] [PubMed]
  27. Al-Salloum, J.; Gillani, S.W.; Mahmood, R.K.; Gulam, S.M. Comparative Efficacy of Azithromycin Versus Clarithromycin in Combination with Beta-lactams to Treat Community-acquired Pneumonia in Hospitalized Patients: A Systematic Review. J. Int. Med. Res. 2021, 49, 3000605211049943. [Google Scholar] [CrossRef]
  28. Falcone, M.; Russo, A.; Shindo, Y.; Farcomeni, A.; Pieralli, F.; Cangemi, R.; Liu, J.; Xia, J.; Okumura, J.; Sano, M.; et al. A Hypothesis-Generating Study of the Combination of Aspirin plus Macrolides in Patients with Severe Community-Acquired Pneumonia. Antimicrob. Agents Chemother. 2019, 63, e01556-18. [Google Scholar] [CrossRef]
  29. Wu, Z.; Zhang, R.; Liu, D.; Liu, X.; Zhang, J.; Zhang, Z.; Chen, S.; He, W.; Li, Y.; Xu, Y.; et al. Acute Respiratory Distress Syndrome Caused by Human Adenovirus in Adults: A Prospective Observational Study in Guangdong, China. Front. Med. 2021, 8, 791163. [Google Scholar] [CrossRef]
  30. Kawamura, K.; Ichikado, K.; Takaki, M.; Eguchi, Y.; Anan, K.; Suga, M. Adjunctive therapy with azithromycin for moderate and severe acute respiratory distress syndrome: A retrospective, propensity score-matching analysis of prospectively collected data at a single center. Int. J. Antimicrob. Agents 2018, 51, 918–924. [Google Scholar] [CrossRef]
  31. Kawamura, K.; Ichikado, K.; Suga, M.; Yoshioka, M. Efficacy of Azithromycin for Treatment of Acute Exacerbation of Chronic Fibrosing Interstitial Pneumonia: A Prospective, Open-label Study with Historical Controls. Respiration 2014, 87, 478–484. [Google Scholar] [CrossRef] [PubMed]
  32. Kawamura, K.; Ichikado, K.; Yasuda, Y.; Anan, K.; Suga, M. Azithromycin for Idiopathic Acute Exacerbation of Idiopathic Pulmonary Fibrosis: A Retrospective Single-center Study. BMC Pulm. Med. 2017, 17, 94. [Google Scholar] [CrossRef]
  33. Hung, I.F.N.; To, K.K.W.; Chan, J.F.W.; Cheng, V.C.C.; Liu, K.S.H.; Tam, A.; Chan, T.C.; Zhang, A.J.; Li, P.; Wong, T.L.; et al. Efficacy of Clarithromycin-naproxen-oseltamivir Combination in the Treatment of Patients Hospitalized for Influenza A(H3N2) Infection: An Open-label Randomized, Controlled, Phase Iib/III trial. Chest 2017, 151, 1069–1080. [Google Scholar] [CrossRef] [PubMed]
  34. Higashi, F.; Kubo, H.; Yasuda, H.; Nukiwa, T.; Yamaya, M. Additional Treatment with Clarithromycin Reduces Fever Duration in Patients with Influenza. Respir. Investig. 2014, 52, 302–309. [Google Scholar] [CrossRef]
  35. Yatera, K.; Umeki, K.; Yamasaki, K.; Noguchi, S.; Nishida, C.; Ishimoto, H.; Sakamoto, N.; Ishii, H.; Kadota, J.I.; Mukae, H. The Additive Effect of Clarithromycin on Influenza A Infection in the Elderly Patients and Patients with Comorbid Diseases. Respir. Investig. 2017, 55, 380–383. [Google Scholar] [CrossRef]
  36. Kakeya, H.; Seki, M.; Izumikawa, K.; Kosai, K.; Morinaga, Y.; Kurihara, S.; Nakamura, S.; Imamura, Y.; Miyazaki, T.; Tsukamoto, M.; et al. Efficacy of Combination Therapy with Oseltamivir Phosphate and Azithromycin for Influenza: A Multicenter, Open-label, Randomized Study. PLoS ONE 2014, 9, e91293. [Google Scholar] [CrossRef] [PubMed]
  37. Martín-Loeches, I.; Bermejo-Martin, J.F.; Vallés, J.; Granada, R.; Vidaur, L.; Vergara-Serrano, J.C.; Martín, M.; Figueira, J.C.; Sirvent, J.M.; Blanquer, J.; et al. Macrolide-based Regimens in Absence of Bacterial Co-infection in Critically Ill H1N1 Patients with Primary Viral Pneumonia. Intensive Care Med. 2013, 39, 693–702. [Google Scholar] [CrossRef]
  38. Poddighe, D.; Aljofan, M. Clinical Evidences on the Antiviral Properties of Macrolide Antibiotics in the COVID-19 Era and Beyond. Antivir. Chem. Chemother. 2020, 28, 2040206620961712. [Google Scholar] [CrossRef]
  39. Firth, A.; Prathapan, P. Azithromycin: The First Broad-spectrum Therapeutic. Eur. J. Med. Chem. 2020, 207, 112739. [Google Scholar] [CrossRef]
  40. Kanoh, S.; Rubin, B.K. Mechanisms of Action and Clinical Application of Macrolides as Immunomodulatory Medications. Clin. Microbiol. Rev. 2010, 23, 590–615. [Google Scholar] [CrossRef]
  41. Pani, A.; Lauriola, M.; Romandini, A.; Scaglione, F. Macrolides and Viral Infections: Focus on Azithromycin in COVID-19 Pathology. Int. J. Antimicrob. Agents 2020, 56, 106053. [Google Scholar] [CrossRef] [PubMed]
  42. Du, X.; Zuo, X.; Meng, F.; Han, C.; Ouyang, W.; Han, Y.; Ouyang, W.; Han, Y.; Gu, Y.; Zhao, X.; et al. Direct Inhibitory Effect on Viral Entry of Influenza A and SARS-CoV-2 Viruses by Azithromycin. Cell Prolif. 2021, 54, e12953. [Google Scholar] [CrossRef] [PubMed]
  43. Oliver, M.E.; Hinks, T.S.C. Azithromycin in Viral Infections. Rev. Med. Virol. 2021, 31, e2163. [Google Scholar] [CrossRef] [PubMed]
  44. Abaleke, E.; Abbas, M.; Abbasi, S.; Abbott, A.; Abdelaziz, A.; Abdelbadiee, S.; Abdelfattah, M.; Abdul, B.; Rasheed, A.A.; Abdul-Kadir, R.; et al. Azithromycin in Patients Admitted to Hospital with COVID-19 (RECOVERY): A Randomised, Controlled, Open-label, Platform Trial. Lancet 2021, 397, 605–612. [Google Scholar] [CrossRef] [PubMed]
  45. Hinks, T.S.C.; Cureton, L.; Knight, R.; Wang, A.; Cane, J.L.; Barber, V.S.; Black, J.; Dutton, S.J.; Melhorn, J.; Jabeen, M.; et al. Azithromycin Versus Standard Care in Patients with Mild-to-Moderate COVID-19 (ATOMIC2): An Open-label, Randomised Trial. Lancet Respir. Med. 2021, 9, 1130–1140. [Google Scholar] [CrossRef]
  46. Melamed, K.H.; Williams, J.; Wang, X.; Hu, S.; Nguyen, C.; Cui, J.; Deng, J.C. Development of Secondary Bacterial Pneumonia in Adults Presenting with Influenza Versus Noninfluenza Viral Respiratory Infection. Ther. Adv. Respir. Dis. 2020, 14, 1753466620963026. [Google Scholar] [CrossRef]
  47. Shieh, W.J.; Blau, D.M.; Denison, A.M.; Deleon-Carnes, M.; Adem, P.; Bhatnagar, J.; Sumner, J.; Liu, L.; Patel, M.; Batten, B.; et al. 2009 Pandemic Influenza A (H1N1): Pathology and Pathogenesis of 100 Fatal Cases in the United States. Am. J. Pathol. 2010, 177, 166–175. [Google Scholar] [CrossRef]
  48. Manohar, P.; Loh, B.; Nachimuthu, R.; Hua, X.; Welburn, S.C.; Leptihn, S. Secondary Bacterial Infections in Patients with Viral Pneumonia. Front. Med. 2020, 7, 420. [Google Scholar] [CrossRef]
  49. The Japanese Respiratory Society. The JRS Guidelines for the Management of Pneumonia in Adults 2017; The Japanese Respiratory Society: Tokyo, Japan, 2017; Volume 1, pp. 17–19. (In Japanese) [Google Scholar]
  50. Morris, D.E.; Cleary, D.W.; Clarke, S.C. Secondary Bacterial Infections Associated with Influenza Pandemics. Front. Microbiol. 2017, 8, 1041. [Google Scholar] [CrossRef]
  51. McCullers, J.A. Insights into the Interaction Between Influenza Virus and Pneumococcus. Clin. Microbiol. Rev. 2006, 19, 571–582. [Google Scholar] [CrossRef]
  52. Shorr, A.F.; Zilberberg, M.D.; Kan, J.; Hoffman, J.; Micek, S.T.; Kollef, M.H. Azithromycin and Survival in Streptococcus pneumoniae Pneumonia: A Retrospective Study. BMJ Open 2013, 3, e002898. [Google Scholar] [CrossRef] [PubMed]
  53. Blondeau, J.M.; Shebelski, S.D.; Hesje, C.K. Killing of Streptococcus pneumoniae by Azithromycin, Clarithromycin, Erythromycin, Telithromycin and Gemifloxacin Using Drug Minimum Inhibitory Concentrations and Mutant Prevention Concentrations. Int. J. Antimicrob. Agents 2015, 45, 594–599. [Google Scholar] [CrossRef] [PubMed]
  54. Shorr, A.F.; Simmons, J.; Hampton, N.; Micek, S.T.; Kollef, M.H. Pneumococal Community-acquired Pneumonia in the Intensive Care Unit: Azithromycin Remains Protective Despite Macrolide Resistance. Respir. Med. 2021, 177, 106307. [Google Scholar] [CrossRef] [PubMed]
  55. Yanagihara, K.; Izumikawa, K.; Higa, F.; Tateyama, M.; Tokimatsu, I.; Hiramatsu, K.; Fujita, J.; Kadota, J.; Kohno, S. Efficacy of Azithromycin in the Treatment of Community-acquired Pneumonia, Including Patients with Macrolide-resistant Streptococcus pneumoniae Infection. Intern. Med. 2009, 48, 527–535. [Google Scholar] [CrossRef]
  56. Parnham, M.J.; Erakovic Haber, V.; Giamarellos-Bourboulis, E.J.; Perletti, G.; Verleden, G.M.; Vos, R. Azithromycin: Mechanisms of Action and Their Relevance for Clinical Applications. Pharmacol. Ther. 2014, 143, 225–245. [Google Scholar] [CrossRef]
  57. Mortensen, E.M.; Halm, E.A.; Pugh, M.J.; Copeland, L.A.; Metersky, M.; Fine, M.J.; Johnson, C.S.; Alvarez, C.A.; Frei, C.R.; Good, C.; et al. Association of Azithromycin with Mortality and Cardiovascular Events Among Older Patients Hospitalized with Pneumonia. JAMA 2014, 311, 2199–2208. [Google Scholar] [CrossRef]
Figure 1. Flow chart of patient selection. Patients were classified according to treatment or no treatment within 7 days of hospitalization.
Figure 1. Flow chart of patient selection. Patients were classified according to treatment or no treatment within 7 days of hospitalization.
Viruses 15 01142 g001
Table 1. Baseline characteristics of the patients in the moderate group with or without intravenous azithromycin administration before and after adjusting for confounders using the propensity score weighting method.
Table 1. Baseline characteristics of the patients in the moderate group with or without intravenous azithromycin administration before and after adjusting for confounders using the propensity score weighting method.
Before AdjustmentAfter Adjustment
VariablesIntravenous Azithromycin
(n = 85)
Without Macrolide
(n = 2654)
p-ValueIntravenous Azithromycin
(n = 85)
Without Macrolide
(n = 2654)
p-Value
Sex (female)37.645.50.15133.3945.290.061
Age (years)72.38 ± 2.170.84 ± 0.50.08767.92 ± 3.270.87 ± 0.50.353
Hospital volume per year11.79 ± 0.914.65 ± 0.50.09210.97 ± 1.314.55 ± 0.50.010
Emergency admission78.868.20.03773.7668.540.469
Emergency transport65.955.30.05460.8555.600.488
Home healthcare4.710.20.0986.1710.000.206
Smoking50.632.70.00129.1733.200.464
Diabetes mellitus18.817.00.65213.5917.000.441
Chronic kidney disease12.98.20.1215.918.340.350
Liver dysfunction4.71.80.0494.781.910.353
Cerebrovascular disease5.910.70.15518.5310.550.350
Heart failure12.915.40.53410.7215.330.218
Cardiovascular disease4.74.20.8273.894.230.911
Hypertension24.725.40.89220.2625.370.307
Cancer15.38.90.0435.929.100.161
COPD5.97.40.5936.977.350.914
Asthma12.913.70.84611.5213.650.495
Interstitial lung disease27.112.3<0.00116.2812.770.424
Chronic respiratory failure4.71.80.0601.721.920.770
Neurological dysfunction2.42.70.8569.782.680.361
Vasopressor11.84.50.00213.824.840.267
Maintenance hemodialysis1.22.30.4940.722.260.319
Emergency hemodialysis2.40.40.0100.310.480.270
Platelet transfusion0.00.0-0.00.0-
Red blood cell transfusion2.41.60.6012.961.680.682
Antithrombin III0.00.0-0.00.0-
rhTM5.90.7<0.0012.150.940.391
Heparin14.15.30.0018.275.660.537
Insulin25.912.3<0.00112.9812.740.951
Albumin4.71.00.0021.001.170.751
Sivelestat0.00.0-0.00.0-
Immunoglobulin0.00.0-0.00.0-
Oseltamivir9.410.40.7698.2510.350.579
Laninamivir1.22.60.4130.472.55<0.001
Peramivir71.859.00.01863.2259.360.635
Penicillin antibiotics37.637.10.92039.1637.150.808
Cephalosporin (1st generation, iv)1.21.00.8570.720.990.858
Cephalosporin (2nd generation, iv)1.21.70.6970.471.710.019
Cephalosporin (3rd generation, iv)44.731.70.01227.2932.060.415
Cephalosporin (4th generation, iv)3.51.40.0961.041.400.573
Carbapenem20.08.9<0.00111.679.310.639
Clindamycin1.20.50.3390.510.480.919
New quinolone5.95.70.92814.905.680.121
Anti-MRSA drug3.50.70.0031.510.770.712
Minocycline4.71.90.0652.801.970.437
Steroid use29.420.10.03616.2520.400.376
PMX0.00.0-0.00.0-
ICU admission rate2.41.00.2360.761.060.583
Data are presented as the % or the mean ± standard error, unless otherwise stated. The groups are adjusted using the inverse probability of treatment weighting method [24]. Abbreviations: COPD, chronic obstructive pulmonary disorder; IMV, invasive mechanical ventilation; NPPV, non-invasive positive pressure ventilation; oral, oral administration; PMX, polymyxin B-immobilized fiber column; rhTM, recombinant human soluble thrombomodulin; MRSA, methicillin-resistant Staphylococcus aureus; ICU, intensive care unit; iv, intravenously.
Table 2. Primary and secondary endpoint estimations using the propensity score weighting method in patients in the moderate group.
Table 2. Primary and secondary endpoint estimations using the propensity score weighting method in patients in the moderate group.
OutcomesIntravenous Azithromycin
(n = 85)
Without Macrolide
(n = 2654)
p-Value
30-day mortality (%)14.899.230.477
90-day mortality (%)15.0611.160.622
In-hospital mortality (%)16.5611.450.516
ICU management, mean days (±SE)0.050 ± 0.030.069 ± 0.010.553
IMV, mean days (±SE)0.028 ± 0.020.154 ± 0.050.026
Hospital stay, mean days (±SE)17.55 ± 2.917.95 ± 0.40.891
Data are presented as the mean ± standard error. The groups are adjusted using the inverse probability of treatment weighting method. Abbreviations: IMV, invasive mechanical ventilation; ICU, intensive care unit; SE, standard error.
Table 3. Baseline characteristics of the patients in the severe group with intravenous administration of azithromycin or without macrolide before and after adjustment for confounders using the propensity score weighting method.
Table 3. Baseline characteristics of the patients in the severe group with intravenous administration of azithromycin or without macrolide before and after adjustment for confounders using the propensity score weighting method.
Before AdjustmentAfter Adjustment
VariablesIntravenous Azithromycin
(n = 90)
Without Macrolide
(n = 517)
p-ValueIntravenous Azithromycin
(n = 90)
Without Macrolide
(n = 517)
p-Value
Sex (female) 34.4438.490.46543.1438.240.424
Age (years)67.99 ± 1.768.03 ± 1.00.07569.91 ± 1.768.05 ± 0.90.282
Hospital volume per year9.19 ± 0.811.85 ± 0.80.43310.11 ± 1.111.40 ± 0.70.298
Emergency admission90.089.170.81492.0789.490.295
Emergency transport78.975.00.43480.8175.920.101
Home healthcare4.49.50.11910.128.710.775
Smoking51.147.80.55950.2148.830.727
Diabetes mellitus24.419.50.28523.3220.350.539
Chronic kidney disease17.815.70.61413.2315.870.440
Liver dysfunction5.62.10.0613.272.560.673
Cerebrovascular disease8.98.70.95411.028.760.556
Heart failure15.623.60.09126.5422.500.541
Cardiovascular disease6.76.40.91951.4963.060.519
Hypertension18.921.90.52627.3321.290.368
Cancer7.85.20.3318.685.790.295
COPD2.27.00.0874.356.250.336
Asthma4.46.60.4413.906.170.130
Interstitial lung disease20.015.90.32912.9016.190.241
Chronic respiratory failure1.13.30.2611.712.960.419
Neurological dysfunction14.422.40.08717.7021.450.335
Vasopressor63.356.30.21266.7557.890.158
Maintenance hemodialysis1.13.10.2930.802.780.006
Emergency hemodialysis1.13.10.2931.572.790.443
Platelet transfusion11.17.40.2228.267.820.835
Red blood cell transfusion22.211.60.00615.5713.600.530
Antithrombin III12.25.00.0087.926.130.491
rhTM20.07.9<0.00110.569.410.667
Heparin62.246.60.00653.5348.760.434
Insulin57.837.7<0.00147.6440.850.220
Albumin34.420.50.00426.1122.920.579
Sivelestat5.62.90.19355.8534.580.539
Immunoglobulin14.46.20.0069.857.450.428
Oseltamivir6.76.20.8634.686.120.283
Laninamivir1.11.20.9680.471.100.078
Peramivir86.774.10.01082.2676.050.153
Penicillin antibiotics34.434.40.99837.2734.460.589
Cephalosporins (1st generation, iv)2.22.10.9542.152.030.924
Cephalosporins (2nd generation, iv)0.00.0-0.00.0-
Cephalosporins (3rd generation, iv)34.433.30.82733.3533.320.995
Cephalosporins (4th generation, iv)3.32.30.0232.222.420.830
Carbapenem41.127.70.01035.9630.120.337
Clindamycin1.11.20.9680.941.120.723
New quinolone12.219.30.10715.2718.280.550
Anti-MRSA drug15.65.4<0.0019.276.980.346
Minocycline3.32.10.4824.452.360.489
Steroid use62.239.5<0.00145.4342.780.599
PMX1.10.60.5660.450.620.471
Oxygen therapy41.145.50.44449.9644.850.397
NPPV5.64.30.5813.774.250.750
IMV wearing days5.184.21<0.0014.614.350.183
ICU admission rate38.928.40.04638.0330.260.119
Data are presented as the % or the mean ± standard error, unless otherwise stated. The groups are adjusted using the inverse probability of treatment weighting method. Abbreviations: COPD, chronic obstructive pulmonary disorder; IMV, invasive mechanical ventilation; NPPV, non-invasive positive pressure ventilation; oral, oral administration; PMX, polymyxin B-immobilized fiber column; rhTM, recombinant human soluble thrombomodulin; MRSA, methicillin-resistant Staphylococcus aureus; ICU, intensive care unit.
Table 4. Primary and secondary endpoint estimations using the propensity score weighting method in patients in the severe group.
Table 4. Primary and secondary endpoint estimations using the propensity score weighting method in patients in the severe group.
OutcomesIntravenous Azithromycin
(n = 90)
Without Macrolide
(n = 517)
p-Value
30-day mortality (%)26.49 36.650.038
90-day mortality (%)33.7840.370.234
In-hospital mortality (%)33.7840.970.194
ICU management, mean days (±SE)2.96 ± 0.42.59 ± 0.20.407
IMV, mean days (±SE)10.82 ± 1.211.69 ± 0.80.534
Hospital stay, mean days (±SE)28.32 ± 2.327.33 ± 1.30.699
Data are presented as the mean ± standard error. The groups are adjusted using the inverse probability of treatment weighting method. Abbreviations: IMV, invasive mechanical ventilation; ICU, intensive care unit; SE, standard error.
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

Tokito, T.; Kido, T.; Muramatsu, K.; Tokutsu, K.; Okuno, D.; Yura, H.; Takemoto, S.; Ishimoto, H.; Takazono, T.; Sakamoto, N.; et al. Impact of Administering Intravenous Azithromycin within 7 Days of Hospitalization for Influenza Virus Pneumonia: A Propensity Score Analysis Using a Nationwide Administrative Database. Viruses 2023, 15, 1142. https://doi.org/10.3390/v15051142

AMA Style

Tokito T, Kido T, Muramatsu K, Tokutsu K, Okuno D, Yura H, Takemoto S, Ishimoto H, Takazono T, Sakamoto N, et al. Impact of Administering Intravenous Azithromycin within 7 Days of Hospitalization for Influenza Virus Pneumonia: A Propensity Score Analysis Using a Nationwide Administrative Database. Viruses. 2023; 15(5):1142. https://doi.org/10.3390/v15051142

Chicago/Turabian Style

Tokito, Takatomo, Takashi Kido, Keiji Muramatsu, Kei Tokutsu, Daisuke Okuno, Hirokazu Yura, Shinnosuke Takemoto, Hiroshi Ishimoto, Takahiro Takazono, Noriho Sakamoto, and et al. 2023. "Impact of Administering Intravenous Azithromycin within 7 Days of Hospitalization for Influenza Virus Pneumonia: A Propensity Score Analysis Using a Nationwide Administrative Database" Viruses 15, no. 5: 1142. https://doi.org/10.3390/v15051142

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