Next Article in Journal
The Muscle-Conditioned Medium Containing Protocatechuic Acid Improves Insulin Resistance by Modulating Muscle Communication with Liver and Adipose Tissue
Previous Article in Journal
Recombinant SARS-CoV-2 Spike Protein Stimulates Secretion of Chymase, Tryptase, and IL-1β from Human Mast Cells, Augmented by IL-33
Previous Article in Special Issue
Transcriptomic Differentiation of Phenotypes in Chronic Rhinosinusitis and Its Implications for Understanding the Underlying Mechanisms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Mechanism of Action and Clinical Efficacy of Low-Dose Long-Term Macrolide Therapy in Chronic Rhinosinusitis

1
Department of Otorhinolaryngology-Head and Neck Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
2
Department of Otorhinolaryngology-Head and Neck Surgery, Ilsan Paik Hospital, Inje University College of Medicine, Goyang 10380, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(11), 9489; https://doi.org/10.3390/ijms24119489
Submission received: 16 March 2023 / Revised: 12 May 2023 / Accepted: 25 May 2023 / Published: 30 May 2023
(This article belongs to the Special Issue Chronic Rhinosinusitis: Aetiology, Immunology and Treatment)

Abstract

:
Various chronic inflammatory airway diseases can be treated with low-dose, long-term (LDLT) macrolide therapy. LDLT macrolides can be one of the therapeutic options for chronic rhinosinusitis (CRS) due to their immunomodulatory and anti-inflammatory actions. Currently, various immunomodulatory mechanisms of the LDLT macrolide treatment have been reported, as well as their antimicrobial properties. Several mechanisms have already been identified in CRS, including reduced cytokines such as interleukin (IL)-8, IL-6, IL-1β, tumor necrosis factor-α, transforming growth factor-β, inhibition of neutrophil recruitment, decreased mucus secretion, and increased mucociliary transport. Although some evidence of effectiveness for CRS has been published, the efficacy of this therapy has been inconsistent across clinical studies. LDLT macrolides are generally believed to act on the non-type 2 inflammatory endotype of CRS. However, the effectiveness of LDLT macrolide treatment in CRS is still controversial. Here, we reviewed the immunological mechanisms related to CRS in LDLT macrolide therapy and the treatment effects according to the clinical situation of CRS.

1. Introduction

Low-dose long-term (LDLT) macrolide therapy is a type of treatment in which the dosage is lower than that used to treat an acute bacterial infection and the duration is longer than that normally used. The regimen was first reported for the treatment of patients with diffuse panbronchiolitis with LDLT erythromycin in Japan in 1984 [1]. Since then, it has been widely used for chronic airway diseases such as chronic obstructive pulmonary disease (COPD), asthma, diffuse panbronchiolitis, bronchiectasis, cystic fibrosis, and idiopathic pulmonary fibrosis [2]. LDLT macrolide therapy has been found to enhance lung function and reduce the frequency and severity of exacerbations in people with these conditions [3]. It is thought that the immunomodulatory and anti-inflammatory potency of macrolides, through various mechanisms, can effectively control these diseases, as can their antimicrobial properties [4]. In addition, macrolide antibiotics have been reported to have therapeutic potential through immunomodulation in a variety of different diseases, such as rheumatoid arthritis, coronary artery disease, non-small cell lung cancer, periodontitis, and blepharitis [5,6].
Chronic rhinosinusitis (CRS) is one of the chronic inflammatory diseases of the upper respiratory tract. CRS has a similar pathophysiology to the above-mentioned lower airway inflammatory diseases, particularly asthma [7,8]. In an early study in 1970, macrolide therapy was able to reduce corticosteroid doses in patients with asthma [9]. A systematic review of the effects of long-term macrolide treatment on asthma found that the treatment reduced exacerbations and symptoms but did not significantly increase lung function [10]. In a multicenter randomized controlled trial (RCT) conducted in patients with severe asthma in Belgium, LDLT azithromycin reduced severe exacerbations and lower respiratory tract infections only in patients with non-eosinophilic severe asthma [11]. Therefore, the authors proposed a role for LDLT macrolides in severe asthmatic patients with corticosteroid insensitivity.
CRS is classified into CRS with nasal polyps (CRSwNP) and CRS without nasal polyps (CRSsNP) according to the phenotype and is divided into type 2 and non-type 2 according to the endotype [12]. As in asthma, corticosteroid therapy in CRS is effective for type 2 and CRSwNP patients, but some cases do not respond to it [13]. Eosinophilic CRSwNP was associated with higher type 2 cytokine expression, such as interleukin (IL)-5, IL-13, and eotaxin. On the other hand, non-type 2 CRS had more neutrophilic inflammation and IL-8 [14]. According to the results of studies about CRS endotypes and biomarkers, the medical treatment of CRS requires customization by patients, including corticosteroids, antibiotics, and biologicals [15]. Appropriate medical therapy for CRS includes a combination of treatments such as intranasal corticosteroid spray, short-term oral corticosteroids, and nasal saline irrigation. If medical treatment is ineffective, functional endoscopic sinus surgery (FESS) can be recommended [12]. Still, the role of antibiotics, including LDLT macrolides, in the treatment of CRS remains controversial.
In general, LDLT macrolide for the treatment of CRS is known to act on non-type 2 inflammation with low levels of eosinophils and immunoglobulin (Ig) E [16]. The treatment with LDLT macrolides in CRS was introduced by a Japanese group in 1991 [17]. Moriyama et al. reported that post-operative LDLT erythromycin showed better improvement of clinical symptoms and endoscopic findings compared to the non-treated group [18]. Since then, LDLT macrolides, including 14-membered lactone erythromycin, roxithromycin, clarithromycin, and 15-membered azithromycin [19], have been studied in various clinical trials and have been frequently prescribed to patients with CRS [20,21]. The European Position Paper on Rhinosinusitis and Nasal Polyps (EPOS) 2012 guidelines recommended LDLT macrolide treatment for CRSsNP patients with normal total IgE levels [22]. So far, this treatment has been known to be effective only in the non-type 2 endotype of CRS [23]. However, its use in CRS is not recommended in the EPOS 2020 guidelines [12]. The reason is that there are no large-scale RCTs on CRS and no specific studies on the clinical phenotype and endotype of CRS [24]. However, the prescription of macrolide-based treatment for CRS is emerging, and the frequency of prescription is high in actual clinical practice [25]. Macrolide antibiotics are the second most prescribed drug after penicillin/beta-lactams in the United States and are also preferred by doctors at university hospitals in South Korea [20,21].
In this review, we investigated the immunological mechanisms related to CRS of LDLT macrolide treatment, focusing on the therapeutic effect according to the clinical situation of CRS. In particular, the evidence of LDLT macrolide therapy was summarized based on the phenotype (CRSwNP and CRSsNP) and endotype of CRS (type 2 vs. non-type 2). Additionally, the duration of treatment, use in children, and side effects of LDLT macrolides were reviewed. Unless otherwise noted, all clinical studies were conducted on adult patients with CRS.

2. Mechanism of Action of LDLT Macrolide Therapy in CRS

Immunomodulation effects of LDLT macrolides in chronic airway disease are driven by multiple pathways, including cytokine and chemokine production, cellular recruitment, mucus secretion, barrier function, biofilm formation, and tissue fibrosis [2,4]. The effects of LDLT macrolide are mainly known to decrease type 1 cytokines and reduce neutrophil recruitment. However, macrolides have also been reported to reduce type 2 cytokines such as IL-4 and IL-5 in chronic airway disease [6]. Treatment with LDLT macrolides has also been found to affect mucociliary clearance and epithelial barrier function [26]. These effects may play a role in the pathophysiology of CRS (Figure 1).

2.1. Reducing Proinflammatory Cytokines

The major immune regulatory effect of macrolides is to reduce the production of proinflammatory cytokines in various inflammatory cells. Macrolides decrease the production of IL-6 and tumor necrosis factor (TNF)-α [27,28]. Azithromycin inhibits the inflammasome and reduces IL-1β secretion in monocytes and macrophages [29,30]. These inhibitory effects are regulated by the alteration of cellular signaling pathways, such as mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase 1/2 (ERK1/2), and nuclear factor (NF)-κB [4].
In CRSsNP patients, the nasal mucosa was cultured with clarithromycin, and the secretion of IL-5, IL-8, and granulocyte-macrophage colony-stimulating factor (GM-CSF) was decreased [31]. In addition, transforming growth factor (TGF)-β and NF-κB were decreased when nasal mucosal tissues were treated with clarithromycin [32]. However, the results were inconsistent in human samples treated with 250 mg of clarithromycin for three months.
After treatment with clarithromycin for eight weeks in CRSwNP patients, levels of IL-6, IL-8, and IL-1β in the nasal secretions were reduced [33]. Another study published by the same author showed decreased eosinophilic inflammatory markers, including regulated on activation, normal T cell expressed and secreted (RANTES), and eosinophilic cationic protein (ECP), after eight weeks of clarithromycin treatment [34]. In an in vitro study, erythromycin suppressed the production of eotaxin and RANTES in a lung fibroblast cell line (human fetal lung fibroblasts 1) [35]. Postoperative clarithromycin treatment significantly reduced ECP levels in nasal secretion at 12 and 24 weeks, but not in the control group [36]. However, conflicting results were found, with no difference in ECP level of nasal secretions between LDLT erythromycin and the placebo group [37].

2.2. Inhibition of Neutrophil Recruitment

IL-8, also known as C-X-C Motif Chemokine Ligand 8 (CXCL8), has been identified as a function of neutrophil recruitment. Erythromycin can inhibit the production of IL-8 by neutrophils and eosinophils [38,39]. Previously, Suzuki et al. reported that the administration of roxithromycin to patients with CRS reduced neutrophil counts and IL-8 levels in nasal secretion [40]. This effect was also confirmed by an RCT with 64 CRSsNP patients [41].
Reduced production of IL-8 and IL-1β can block the extravascular transmigration of neutrophils through inhibition of transcription factors such as NF-κB and activator protein-1 (AP-1) [42]. In healthy subjects and COPD patients, short-term administration of azithromycin reduced IL-8 and soluble vascular cell adhesion molecule (VCAM)-1 and modulated neutrophil function [43,44]. Furthermore, azithromycin suppressed the proliferation and cytokine production of CD4+ T cells, especially IL-17 secretion via the mammalian target of the rapamycin (mTOR) pathway [45].

2.3. Mucus Secretion and Mucociliary Clearance

Macrolides can reduce the expression of MUC5AC in airway epithelial cells [26]. Clarithromycin and erythromycin effectively inhibited the expression of MUC5AC in human nasal epithelial cells from CRSwNP patients [46]. Azithromycin also significantly reduced MUC5AC expression in human nasal epithelial cells [47]. In rats stimulated with intratracheal lipopolysaccharide (LPS), roxithromycin treatment significantly reduced Muc5ac expression and NF-κB nuclear translocation in the bronchial epithelium [48]. Azithromycin and clarithromycin showed the same effect in ovalbumin (OVA)-sensitized and LPS-instilled rats [46,47]. In human bronchial epithelial cells, clarithromycin inhibited the expression of MUC5AC and IL-13-induced goblet cell hyperplasia [49,50]. Similar to other inhibitory mechanisms, clarithromycin had an impact on NF-κB inactivation. In a CRS mouse model, the level of IL-10 was increased, and Muc5ac expression was inhibited by erythromycin treatment [51].
In patients with acute purulent rhinitis, clarithromycin treatment for two weeks reduced secretion volume and increased mucociliary transportability [52]. Clarithromycin had the same effect of significantly reducing mucus viscosity and nasal clearance time in CRS patients treated for four weeks [53,54]. In a three-month RCT in patients with CRS, saccharine transit time was significantly improved in the roxithromycin group compared to the placebo group [41]. Improvement in mucociliary clearance, as measured by saccharine transit time, persisted after 12 months of follow-up [55].

2.4. Epithelial Barrier Function

Asgrimsson et al. reported that azithromycin, but not erythromycin, induced expression of tight junction proteins, including claudin-1, claudin-4, occludin, and junctional adhesion molecule-A, and increased epithelial integrity in human bronchial epithelial cells [56]. During Pseudomonas aeruginosa infection in vitro, pretreatment with azithromycin prevented epithelial barrier dysfunction and enhanced recovery [57]. The potential protective effects of macrolides on human respiratory epithelium were investigated in vitro [58,59]. Macrolides, such as roxithromycin, clarithromycin, and azithromycin, reduced the production of reactive oxygen species generated by activated neutrophils [58]. These agents were able to attenuate the injurious effects of bioactive phospholipids and neutrophil-induced epithelial damage [59]. Lastly, roxithromycin treatment increased ciliary movement and mucociliary transport velocity in the rabbit trachea [60].

2.5. Inhibition of Biofilm Formation

Biofilms are a surrounding structure of microorganisms that can provide resistance to host immune responses and antimicrobial agents and are also important for CRS pathophysiology [61]. Bacterial biofilms induced by Staphylococcus aureus or Pseudomonas aeruginosa contribute to the severity and refractoriness of CRS [62]. Korkmaz et al. reported the biofilm eradication effect of eight weeks of clarithromycin treatment in a RCT in CRSwNP patients [63]. Compared to the mometasone furoate nasal spray group (1 of 11), there were more patients (6 of 12) with biofilm disappearance in the clarithromycin treatment group. Previous in vitro studies found that macrolides can inhibit the production of bacterial proteins and reduce biofilm formation by Pseudomonas aeruginosa [64,65]. Recently, antibiotic (ciprofloxacin and azithromycin)-eluting sinus stents have been experimentally demonstrated to inhibit Pseudomonas aeruginosa-induced biofilms [66]. The authors demonstrated that prolonged release of ciprofloxacin and azithromycin for 28 days reduced biofilm formation and eliminated existing biofilms.

2.6. Effects on Tissue Fibrosis

Several in vitro studies have shown that macrolides inhibit fibroblasts in nasal polyps. When nasal polyp-derived fibroblasts were treated with roxithromycin and then stimulated with lipopolysaccharide, fibroblast proliferation was inhibited [67]. This suppression phenomenon was actually observed in the fibroblasts of CRSwNP patients treated with roxithromycin for one month [68]. In addition, roxithromycin inhibited the production of nitric oxide [69], IL-6 and RANTES [69], matrix metalloproteinase (MMP)-2, and MMP-9 [70] in TNF-α-stimulated nasal polyp fibroblasts. Another in vitro study with nasal polyp fibroblasts demonstrated that erythromycin and roxithromycin treatment reduced TGF-β-induced α-smooth muscle actin (a myofibroblast marker), collagen production, nicotinamide adenine dinucleotide phosphate oxidase 4, and reactive oxygen species production [71]. Collectively, these findings indicate an inhibitory effect of macrolide treatment on fibroblast-induced nasal polyp formation and may explain the mechanism of polyp size reduction in patients with CRSwNP.

3. Comparison of Clinical Efficacy in CRS

Although there are few direct comparative studies of each clinical situation, RCT studies of macrolides are summarized. Most of the studies compared placebo with macrolides (Table 1), and some compared treatment with conventional CRS treatment, intranasal corticosteroid spray (Table 2). Herein, the results of clinical trials were comprehensively reviewed according to the presence or absence of nasal polyps, type of inflammation, total IgE level, and the presence or absence of allergy.

3.1. CRSwNP vs. CRSsNP

After long-term clarithromycin treatment (8 to 12 weeks) in 20 CRSwNP patients, 40% of patients had a reduction in nasal polyp size and a significant decrease in IL-8 levels in lavage fluid, while 60% remained unchanged [81]. Preoperative treatment with 500 mg of clarithromycin for eight weeks reduced polyp recurrence at 6 and 12 months postoperatively [77]. Computed tomography (CT) findings and SNOT-20 improved in CRSwNP patients treated with mometasone furoate monotherapy and LDLT clarithromycin combination therapy for eight weeks, but there was no statistically significant difference between the two groups [63]. In 52 CRSwNP patients treated with LDLT clarithromycin for 12 weeks, there were significant reductions in the Sinonasal Outcome Test (SNOT)-20 and Lund-Kennedy endoscopy score [82]. In addition, 54% (28 of 52) of those who improved on SNOT-20 had lower total IgE levels than others.
In CRSsNP patients, treatment with 150 mg roxithromycin for three months showed significant improvement in sinonasal symptoms (SNOT-20), nasal endoscopy findings, and mucociliary transit time [41]. When comparing the effects of mometasone furoate and LDLT clarithromycin at three months, there was no significant difference in visual analog scales of symptoms or endoscopic findings between the two groups [80]. Eight weeks of erythromycin treatment also showed clinical improvement in CRSsNP patients [83].
However, in a mixed cohort of CRSwNPs (52.0%) and CRSsNPs, except for severe polyposis, LDLT azithromycin did not differ between treatment and placebo groups [78]. Treatment with LDLT azithromycin for three months after FESS in CRS patients improved SNOT-22 compared to conventional treatment [76], whereas erythromycin treatment after FESS was ineffective in CRSwNP (55.2%) and CRSsNP [37]. There was no additional effect of clarithromycin for three months with budesonide aqua nasal spray in patients with CRS (56.8% with nasal polyps) [84]. Haruna et al. retrospectively analyzed patients who received LDLT macrolides treatment for 8–20 weeks; the clinical effect was good in CRSsNP patients, whereas the effect increased after polypectomy in CRSwNP patients [85]. In a biomarker study for the prediction of the macrolide treatment group in patients with CRS postoperatively, nasal tissue IgG4 level and overall symptom score were identified as predictive factors for refractoriness [86]. However, there was no difference in refractory rate between the LDLT clarithromycin treatment group (18 of 74) and the fluticasone propionate spray group (17 of 75).

3.2. Type 2 vs. Non-Type 2

Treatment with macrolides (clarithromycin or roxithromycin) for 2–3 months improved clinical symptoms in CRS patients, and the degree of clinical improvement was inversely correlated with eosinophil counts in the peripheral blood, the nasal smear, and the sinus mucosa [87]. However, the number of neutrophils, mast cells, and mononuclear cells did not correlate with symptomatic improvement, and the number of interferon-γ and IL-4-positive cells also did not correlate.
Zeng et al. compared the efficacy of fluticasone propionate nasal spray versus LDLT clarithromycin for postoperative treatment in CRS of different phenotypes. The study found that both medications were effective in reducing symptoms, but there were no significant differences between eosinophilic (>10% eosinophils/total infiltrating cells) and non-eosinophilic CRSwNP groups [79]. Asians, who are generally known to have more non-type 2 CRS, showed better treatment effects of LDLT macrolides than non-Asians in a meta-analysis [88].
A recent study showed that long-term treatment with clarithromycin was effective in CRSwNP patients without tissue eosinophilia (>10 eosinophils/high power field) [74]. When comparing oral steroids alone with oral steroids plus clarithromycin for 12 weeks in CRSwNP patients who underwent FESS, symptom scores and endoscopy scores improved significantly in the add-on treatment group [74]. In a case-control study of LDLT clarithromycin after surgery, responders (19 of 28, 67.9%) had lower blood eosinophil counts (0.16 ± 0.11 versus 0.39 ± 0.36 109/L) and tissue eosinophilia (>10 eosinophils/high power field, 17.6% versus 62.5%) compared to non-responders [89]. According to these studies, patients with type 2 inflammation of CRS have a lower response to LDLT macrolide therapy.
Aspirin or nonsteroidal anti-inflammatory drug (NSAID)-exacerbated respiratory disease (AERD/NERD) is characterized by asthma, CRSwNP, and aspirin or nonsteroidal anti-inflammatory drug intolerance [90]. In AERD patients with eosinophilic nasal polyps (>40% eosinophils), LDLT azithromycin treatment significantly reduced symptoms (visual analog scale and SNOT-22) and the need for surgery (74% versus 14%) compared to placebo [75]. In addition, another study showed that azithromycin significantly improved disease clearance in AERD patients compared to placebo [73]. In patients with refractory CRS who failed surgery and medical treatment, azithromycin treatment not only alleviated symptoms but also significantly reduced the amount of Staphylococcus aureus [72,73]. These recent studies have demonstrated that LDLT macrolide treatment is also effective in CRS patients with eosinophilic inflammation.

3.3. Normal vs. High Total IgE

Previous studies reported that only CRS patients with normal serum IgE levels (<200 μg/L [41] or ≤250 U/mL [87]) benefited from LDLT macrolide treatment. However, the relationship between total serum IgE levels and LDLT macrolide treatment effects is still controversial. According to the studies showing that LDLT macrolide treatment was effective, the total serum IgE in the patient group was 188.63 ± 57.25 IU/mL [77] and 165.0 ± 195.2 μL/L [37]. Maniakas et al. reported that total serum IgE was higher in the azithromycin success group compared to the azithromycin failure group [91]. In addition, atopy status did not affect the clinical effect of clarithromycin in CRSsNP patients [80].
Recently, of the 100 CRS patients who were administered LDLT roxithromycin, 29 were determined to be responders [92]. Among clinical parameters, including nasal secretion and serum IgE, IL-5, blood eosinophil/neutrophil, allergy, asthma, and nasal polyps, total IgE in nasal secretions was the only predictor of responder in multivariate models (odds ratio 4.76, 95% confidential interval 1.29–17.58). The authors suggest that local total IgE is a reliable biomarker instead of serum total IgE.

3.4. Allergic vs. Non-Allergic Patients

Yamada et al. evaluated the effectiveness of LDLT clarithromycin in patients with non-allergic CRSwNP and found a significant reduction in nasal polyp size and IL-8 secretion in 40% (8 of 20) of patients [81]. CRS patients with or without allergies have different responses to treatment with LDLT macrolides. In patients with confirmed CRSwNP allergy status by skin prick test, ECP levels in nasal secretions decreased in allergic patients, and IL-6 levels decreased only in allergic patients after eight weeks of clarithromycin treatment [34]. However, allergic status had no impact on the clinical efficacy of LDLT macrolides [85,93].

4. Other Considerations

There are several factors to consider while prescribing LDLT macrolides, including the type of medication, dose, duration, and timing of treatment. Patient characteristics such as age, underlying disease, and comorbidities should be considered in the treatment. In addition, we should be aware of the adverse effects of long-term treatment. Although there has been no well-designed study that directly compared the treatment effects between macrolides in CRS, there was a recent study comparing the effects of two drugs. Comparing the effects of clarithromycin and azithromycin treatment for four weeks, azithromycin was more effective for complete resolution of symptoms and CT scores [94]. In a systematic review and meta-analysis of clarithromycin in CRS compared with the intranasal corticosteroid spray, there was no significant difference in effectiveness [95]. However, combined treatment with clarithromycin and intranasal corticosteroid spray markedly improved clinical symptoms, endoscopic findings, and Lund-Mackay CT scores.

4.1. Duration of Treatment

LDLT macrolide treatment is known to be more effective the longer the treatment period. After treatment with 8 to 12 weeks of clarithromycin in CRS patients, symptoms and endoscopic findings improved in 71.1% of participants, and the clinical effect was correlated with the duration of treatment [96]. In a meta-analysis, the effects were more favorable in patients taking LDLT macrolides for 24 weeks in comparison to 8 and 12 weeks [97]. Treatment with clarithromycin for 24 weeks after FESS resulted in better CT scores compared to those for 12 weeks [36]. Treatment with clarithromycin showed clinical effects after four weeks and reached its maximum effect at 12 weeks in patients with CRSsNP [80]. Nakamura et al. compared the clinical efficacy of LDLT clarithromycin in patients with CRS postoperatively. In the 6-month treatment group, the rate of asymptomatic improvement was higher at 12 months after surgery than in the 3-month treatment group [98]. Taken together, the longer the treatment period, the better the clinical outcome of LDLT macrolides.

4.2. Pediatric CRS Patients

Some evidence for LDLT macrolide treatment has also been reported in pediatric patients with CRS [99]. A retrospective review of six patients (mean age: 7 ± 3.4 years) who were treated with either roxithromycin or clarithromycin found that macrolide add-on therapy improved nasal symptoms and reduced thick mucus secretions [100]. After administration of clarithromycin at a half dose (5–8 mg/kg) for eight weeks to 54 children with CRS, 63.0% were cured and 31.5% were improved [101].
Therapeutic effects of LDLT macrolides, such as improving lung function and reducing exacerbations, have been demonstrated in chronic inflammatory diseases of the lower respiratory tract, such as severe asthma and cystic fibrosis in children [99]. Unfortunately, no randomized, placebo-controlled clinical trials of LDLT treatment in children have been conducted.

4.3. Adverse Effects of LDLT Macrolides

During LDLT macrolide treatment for 8 to 12 weeks, there is no strong evidence of the development of drug-resistant bacteria strains [16]. However, LDLT azithromycin use over 12 to 24 months in pediatric patients with bronchiectasis resulted in an increased presence of macrolide-resistant organisms [102]. Macrolides also carry a risk of prolongation of the QT interval and consequent torsades de pointes arrhythmia [103,104]. On the other hand, the incidence of torsades de pointes with erythromycin was very rare (four cases out of 34,000 patients treated) [105]. In a national cohort that included 66,331 CRS patients, the risk of mortality and cardiovascular events was not significantly increased in patients who had been prescribed macrolides, particularly clarithromycin, compared to penicillin [106]. Clarithromycin treatment was known to increase the risk of stroke and myocardial infarction, but a nation-wide cohort study showed no association with overall mortality or long-term cardiovascular death [107,108]. Nonetheless, caution is required if the patient is at risk of a cardiac event prior to the initiation of LDLT macrolide treatment [16].

5. Conclusions

Because CRS is a highly heterogeneous disease entity, the clinical efficacy of LDLT macrolide therapy is variable. Several RCTs have demonstrated that LDLT macrolides can improve symptoms and quality of life in patients with CRS, particularly those with CRSsNP, normal total IgE levels, and corticosteroid resistance [109]. The immunomodulatory and anti-inflammatory properties of macrolides contribute not only to the reduction of neutrophilic inflammation but also to the decrease of eosinophilic inflammation, mucus clearance, and mucosal stabilization. In addition, it can increase the effectiveness of treatment by removing bacterial biofilm and preventing or reducing polyp formation by inhibiting tissue fibrosis. These various mechanisms may have an impact on CRS treatment for numerous clinical conditions. The efficacy of LDLT macrolide therapy may be influenced by the endotype and phenotype of CRS.
Further research is needed to fully understand the mechanisms underlying the therapeutic effect of macrolides in CRS and to identify the most appropriate patients for this treatment approach, including non-antibiotic macrolides [2,110]. Currently, non-antibiotic macrolides such as EM900, an erythromycin derivative, are being developed and researched and may be spotlighted as an important treatment modality for CRS in the future [111]. Nonetheless, current evidence suggests that low-dose, long-term macrolide therapy is a promising option for the management of CRS. LDLT macrolide treatment may be the main treatment for certain subtypes of CRS and may be used as an additional treatment with corticosteroids for other types of CRS.

Author Contributions

Conceptualization, G.R. and H.Y.K.; data curation, E.L., S.I.P., M.P. and S.D.H.; writing—original draft preparation, G.R. and E.L.; writing—review and editing, M.P., S.D.H., Y.G.J. and H.Y.K.; funding acquisition, H.Y.K. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (Ministry of Trade, Industry, and Energy) (20016285).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kudoh, S.; Azuma, A.; Yamamoto, M.; Izumi, T.; Ando, M. Improvement of survival in patients with diffuse panbronchiolitis treated with low-dose erythromycin. Am. J. Respir. Crit. Care Med. 1998, 157, 1829–1832. [Google Scholar] [PubMed]
  2. Kricker, J.A.; Page, C.P.; Gardarsson, F.R.; Baldursson, O.; Gudjonsson, T.; Parnham, M.J. Nonantimicrobial Actions of Macrolides: Overview and Perspectives for Future Development. Pharmacol. Rev. 2021, 73, 233–262. [Google Scholar] [PubMed]
  3. Smith, D.; Du Rand, I.A.; Addy, C.; Collyns, T.; Hart, S.; Mitchelmore, P.; Rahman, N.; Saggu, R. British Thoracic Society guideline for the use of long-term macrolides in adults with respiratory disease. BMJ Open Respir. Res. 2020, 7, e000489. [Google Scholar] [PubMed]
  4. 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]
  5. Culić, O.; Eraković, V.; Parnham, M.J. Anti-inflammatory effects of macrolide antibiotics. Eur. J. Pharmacol. 2001, 429, 209–229. [Google Scholar]
  6. Zimmermann, P.; Ziesenitz, V.C.; Curtis, N.; Ritz, N. The Immunomodulatory Effects of Macrolides-A Systematic Review of the Underlying Mechanisms. Front. Immunol. 2018, 9, 302. [Google Scholar] [CrossRef]
  7. Grossman, J. One airway, one disease. Chest 1997, 111, 11s–16s. [Google Scholar]
  8. Samitas, K.; Carter, A.; Kariyawasam, H.H.; Xanthou, G. Upper and lower airway remodelling mechanisms in asthma, allergic rhinitis and chronic rhinosinusitis: The one airway concept revisited. Allergy 2018, 73, 993–1002. [Google Scholar]
  9. Itkin, I.H.; Menzel, M.L. The use of macrolide antibiotic substances in the treatment of asthma. J. Allergy 1970, 45, 146–162. [Google Scholar]
  10. Undela, K.; Goldsmith, L.; Kew, K.M.; Ferrara, G. Macrolides versus placebo for chronic asthma. Cochrane Database Syst. Rev. 2021, 11, Cd002997. [Google Scholar]
  11. Brusselle, G.G.; Vanderstichele, C.; Jordens, P.; Deman, R.; Slabbynck, H.; Ringoet, V.; Verleden, G.; Demedts, I.K.; Verhamme, K.; Delporte, A.; et al. Azithromycin for prevention of exacerbations in severe asthma (AZISAST): A multicentre randomised double-blind placebo-controlled trial. Thorax 2013, 68, 322–329. [Google Scholar]
  12. Fokkens, W.J.; Lund, V.J.; Hopkins, C.; Hellings, P.W.; Kern, R.; Reitsma, S.; Toppila-Salmi, S.; Bernal-Sprekelsen, M.; Mullol, J.; Alobid, I.; et al. European Position Paper on Rhinosinusitis and Nasal Polyps 2020. Rhinology 2020, 58, 1–464. [Google Scholar]
  13. Gurrola, J., 2nd; Borish, L. Chronic rhinosinusitis: Endotypes, biomarkers, and treatment response. J. Allergy Clin. Immunol. 2017, 140, 1499–1508. [Google Scholar]
  14. Tomassen, P.; Vandeplas, G.; Van Zele, T.; Cardell, L.O.; Arebro, J.; Olze, H.; Förster-Ruhrmann, U.; Kowalski, M.L.; Olszewska-Ziąber, A.; Holtappels, G.; et al. Inflammatory endotypes of chronic rhinosinusitis based on cluster analysis of biomarkers. J. Allergy Clin. Immunol. 2016, 137, 1449–1456.e4. [Google Scholar]
  15. Xu, Z.; Huang, Y.; Delemarre, T.; Cavaliere, C.; Zhang, N.; Bachert, C. Advances in chronic rhinosinusitis in 2020 and 2021. J. Allergy Clin. Immunol. 2022, 149, 854–866. [Google Scholar]
  16. Lees, K.A.; Orlandi, R.R.; Oakley, G.; Alt, J.A. The Role of Macrolides and Doxycycline in Chronic Rhinosinusitis. Immunol. Allergy Clin. N. Am. 2020, 40, 303–315. [Google Scholar]
  17. Kikukchi, S.; Suzaki, H.; Aoki, A.; Ito, O.; Nomura, Y. Clinical effect of long-term low-dose erythromycin therapy for chronic sinusitis. Pract Otlo (Koyoto) 1991, 84, 41–47. [Google Scholar] [CrossRef]
  18. Moriyama, H.; Yanagi, K.; Ohtori, N.; Fukami, M. Evaluation of endoscopic sinus surgery for chronic sinusitis: Post-operative erythromycin therapy. Rhinology 1995, 33, 166–170. [Google Scholar]
  19. Janas, A.; Przybylski, P. 14- and 15-membered lactone macrolides and their analogues and hybrids: Structure, molecular mechanism of action and biological activity. Eur. J. Med. Chem. 2019, 182, 111662. [Google Scholar]
  20. Ryu, G.; Baek, B.-J. A Survey on the Management of Chronic Rhinosinusitis in the Korean Rhinologic Society. J. Rhinol. 2021, 28, 44–49. [Google Scholar] [CrossRef]
  21. Smith, S.S.; Kim, R.; Douglas, R. Is there a role for antibiotics in the treatment of chronic rhinosinusitis? J. Allergy Clin. Immunol. 2022, 149, 1504–1512. [Google Scholar] [CrossRef] [PubMed]
  22. Fokkens, W.J.; Lund, V.J.; Mullol, J.; Bachert, C.; Alobid, I.; Baroody, F.; Cohen, N.; Cervin, A.; Douglas, R.; Gevaert, P.; et al. European Position Paper on Rhinosinusitis and Nasal Polyps 2012. Rhinol. Suppl. 2012, 23, 1–298. [Google Scholar]
  23. Oakley, G.M.; Harvey, R.J.; Lund, V.J. The Role of Macrolides in Chronic Rhinosinusitis (CRSsNP and CRSwNP). Curr. Allergy Asthma Rep. 2017, 17, 30. [Google Scholar]
  24. Lasso, A.; Masoudian, P.; Quinn, J.G.; Cowan, J.; Labajian, V.; Bonaparte, J.P.; Kilty, S. Long-term low-dose macrolides for chronic rhinosinusitis in adults—A systematic review of the literature. Clin. Otolaryngol. 2017, 42, 637–650. [Google Scholar] [PubMed]
  25. Hopkins, C.; Williamson, E.; Morris, S.; Clarke, C.S.; Thomas, M.; Evans, H.; Little, P.; Lund, V.J.; Blackshaw, H.; Schilder, A.; et al. Antibiotic usage in chronic rhinosinusitis: Analysis of national primary care electronic health records. Rhinology 2019, 57, 420–429. [Google Scholar]
  26. Cervin, A.; Wallwork, B. Anti-inflammatory effects of macrolide antibiotics in the treatment of chronic rhinosinusitis. Otolaryngol. Clin. N. Am. 2005, 38, 1339–1350. [Google Scholar]
  27. Yokota, S.; Okabayashi, T.; Hirakawa, S.; Tsutsumi, H.; Himi, T.; Fujii, N. Clarithromycin suppresses human respiratory syncytial virus infection-induced Streptococcus pneumoniae adhesion and cytokine production in a pulmonary epithelial cell line. Mediators Inflamm. 2012, 2012, 528568. [Google Scholar]
  28. Lin, S.J.; Kuo, M.L.; Hsiao, H.S.; Lee, P.T. Azithromycin modulates immune response of human monocyte-derived dendritic cells and CD4(+) T cells. Int. Immunopharmacol. 2016, 40, 318–326. [Google Scholar]
  29. Gualdoni, G.A.; Lingscheid, T.; Schmetterer, K.G.; Hennig, A.; Steinberger, P.; Zlabinger, G.J. Azithromycin inhibits IL-1 secretion and non-canonical inflammasome activation. Sci. Rep. 2015, 5, 12016. [Google Scholar]
  30. Lendermon, E.A.; Coon, T.A.; Bednash, J.S.; Weathington, N.M.; McDyer, J.F.; Mallampalli, R.K. Azithromycin decreases NALP3 mRNA stability in monocytes to limit inflammasome-dependent inflammation. Respir. Res. 2017, 18, 131. [Google Scholar] [CrossRef]
  31. Wallwork, B.; Coman, W.; Feron, F.; Mackay-Sim, A.; Cervin, A. Clarithromycin and prednisolone inhibit cytokine production in chronic rhinosinusitis. Laryngoscope 2002, 112, 1827–1830. [Google Scholar] [CrossRef]
  32. Wallwork, B.; Coman, W.; Mackay-Sim, A.; Cervin, A. Effect of clarithromycin on nuclear factor-kappa B and transforming growth factor-beta in chronic rhinosinusitis. Laryngoscope 2004, 114, 286–290. [Google Scholar] [CrossRef]
  33. Peric, A.; Vojvodic, D.; Baletic, N.; Peric, A.; Miljanovic, O. Influence of allergy on the immunomodulatory and clinical effects of long-term low-dose macrolide treatment of nasal polyposis. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc Czech Repub. 2010, 154, 327–333. [Google Scholar] [CrossRef]
  34. Perić, A.; Vojvodić, D.; Matković-Jožin, S. Effect of long-term, low-dose clarithromycin on T helper 2 cytokines, eosinophilic cationic protein and the ‘regulated on activation, normal T cell expressed and secreted’ chemokine in the nasal secretions of patients with nasal polyposis. J. Laryngol. Otol. 2012, 126, 495–502. [Google Scholar] [CrossRef]
  35. Sato, E.; Nelson, D.K.; Koyama, S.; Hoyt, J.C.; Robbins, R.A. Erythromycin modulates eosinophil chemotactic cytokine production by human lung fibroblasts in vitro. Antimicrob. Agents Chemother. 2001, 45, 401–406. [Google Scholar] [CrossRef]
  36. Varvyanskaya, A.; Lopatin, A. Efficacy of long-term low-dose macrolide therapy in preventing early recurrence of nasal polyps after endoscopic sinus surgery. Int. Forum Allergy Rhinol. 2014, 4, 533–541. [Google Scholar] [CrossRef]
  37. Haxel, B.R.; Clemens, M.; Karaiskaki, N.; Dippold, U.; Kettern, L.; Mann, W.J. Controlled trial for long-term low-dose erythromycin after sinus surgery for chronic rhinosinusitis. Laryngoscope 2015, 125, 1048–1055. [Google Scholar] [CrossRef]
  38. Oishi, K.; Sonoda, F.; Kobayashi, S.; Iwagaki, A.; Nagatake, T.; Matsushima, K.; Matsumoto, K. Role of interleukin-8 (IL-8) and an inhibitory effect of erythromycin on IL-8 release in the airways of patients with chronic airway diseases. Infect. Immun. 1994, 62, 4145–4152. [Google Scholar] [CrossRef]
  39. Kohyama, T.; Takizawa, H.; Kawasaki, S.; Akiyama, N.; Sato, M.; Ito, K. Fourteen-member macrolides inhibit interleukin-8 release by human eosinophils from atopic donors. Antimicrob. Agents Chemother. 1999, 43, 907–911. [Google Scholar] [CrossRef]
  40. Suzuki, H.; Shimomura, A.; Ikeda, K.; Oshima, T.; Takasaka, T. Effects of long-term low-dose macrolide administration on neutrophil recruitment and IL-8 in the nasal discharge of chronic sinusitis patients. Tohoku J. Exp. Med. 1997, 182, 115–124. [Google Scholar] [CrossRef]
  41. Wallwork, B.; Coman, W.; Mackay-Sim, A.; Greiff, L.; Cervin, A. A double-blind, randomized, placebo-controlled trial of macrolide in the treatment of chronic rhinosinusitis. Laryngoscope 2006, 116, 189–193. [Google Scholar] [CrossRef] [PubMed]
  42. Suzuki, H.; Ikeda, K. Mode of action of long-term low-dose macrolide therapy for chronic sinusitis in the light of neutrophil recruitment. Curr. Drug Targets Inflamm. Allergy 2002, 1, 117–126. [Google Scholar] [CrossRef] [PubMed]
  43. Culić, O.; Eraković, V.; Cepelak, I.; Barisić, K.; Brajsa, K.; Ferencić, Z.; Galović, R.; Glojnarić, I.; Manojlović, Z.; Munić, V.; et al. Azithromycin modulates neutrophil function and circulating inflammatory mediators in healthy human subjects. Eur. J. Pharmacol. 2002, 450, 277–289. [Google Scholar] [CrossRef] [PubMed]
  44. Parnham, M.J.; Culić, O.; Eraković, V.; Munić, V.; Popović-Grle, S.; Barisić, K.; Bosnar, M.; Brajsa, K.; Cepelak, I.; Cuzić, S.; et al. Modulation of neutrophil and inflammation markers in chronic obstructive pulmonary disease by short-term azithromycin treatment. Eur. J. Pharmacol. 2005, 517, 132–143. [Google Scholar] [CrossRef]
  45. Ratzinger, F.; Haslacher, H.; Poeppl, W.; Hoermann, G.; Kovarik, J.J.; Jutz, S.; Steinberger, P.; Burgmann, H.; Pickl, W.F.; Schmetterer, K.G. Azithromycin suppresses CD4(+) T-cell activation by direct modulation of mTOR activity. Sci. Rep. 2014, 4, 7438. [Google Scholar] [CrossRef]
  46. Shimizu, T.; Shimizu, S.; Hattori, R.; Gabazza, E.C.; Majima, Y. In vivo and in vitro effects of macrolide antibiotics on mucus secretion in airway epithelial cells. Am. J. Respir. Crit. Care Med. 2003, 168, 581–587. [Google Scholar] [CrossRef]
  47. Shimizu, T.; Shimizu, S. Azithromycin inhibits mucus hypersecretion from airway epithelial cells. Mediators Inflamm. 2012, 2012, 265714. [Google Scholar] [CrossRef]
  48. Ou, X.M.; Feng, Y.L.; Wen, F.Q.; Wang, K.; Yang, J.; Deng, Z.P.; Liu, D.S.; Li, Y.P. Macrolides attenuate mucus hypersecretion in rat airways through inactivation of NF-kappaB. Respirology 2008, 13, 63–72. [Google Scholar] [CrossRef]
  49. Hara, K.; Kondo, M.; Tsuji, M.; Takeyama, K.; Tamaoki, J. Clarithromycin suppresses IL-13-induced goblet cell metaplasia via the TMEM16A-dependent pathway in guinea pig airway epithelial cells. Respir. Investig. 2019, 57, 79–88. [Google Scholar] [CrossRef]
  50. Tanabe, T.; Kanoh, S.; Tsushima, K.; Yamazaki, Y.; Kubo, K.; Rubin, B.K. Clarithromycin inhibits interleukin-13-induced goblet cell hyperplasia in human airway cells. Am. J. Respir. Cell Mol. Biol. 2011, 45, 1075–1083. [Google Scholar] [CrossRef]
  51. Yen, T.T.; Jiang, R.S.; Chang, C.Y.; Wu, C.Y.; Liang, K.L. Erythromycin reduces nasal inflammation by inhibiting immunoglobulin production, attenuating mucus secretion, and modulating cytokine expression. Sci. Rep. 2021, 11, 21737. [Google Scholar] [CrossRef]
  52. Rubin, B.K.; Druce, H.; Ramirez, O.E.; Palmer, R. Effect of clarithromycin on nasal mucus properties in healthy subjects and in patients with purulent rhinitis. Am. J. Respir. Crit. Care Med. 1997, 155, 2018–2023. [Google Scholar] [CrossRef]
  53. Nishi, K.; Mizuguchi, M.; Tachibana, H.; Ooka, T.; Amemiya, T.; Myou, S.; Fujimura, M.; Matsuda, T. Effect of clarithromycin on symptoms and mucociliary transport in patients with sino-bronchial syndrome. Nihon Kyobu Shikkan Gakkai Zasshi 1995, 33, 1392–1400. [Google Scholar]
  54. Rhee, C.S.; Majima, Y.; Arima, S.; Jung, H.W.; Jinn, T.H.; Min, Y.G.; Sakakura, Y. Effects of clarithromycin on rheological properties of nasal mucus in patients with chronic sinusitis. Ann. Otol. Rhinol. Laryngol. 2000, 109, 484–487. [Google Scholar]
  55. Cervin, A.; Kalm, O.; Sandkull, P.; Lindberg, S. One-year low-dose erythromycin treatment of persistent chronic sinusitis after sinus surgery: Clinical outcome and effects on mucociliary parameters and nasal nitric oxide. Otolaryngol. Head Neck Surg. 2002, 126, 481–489. [Google Scholar] [CrossRef]
  56. Asgrimsson, V.; Gudjonsson, T.; Gudmundsson, G.H.; Baldursson, O. Novel effects of azithromycin on tight junction proteins in human airway epithelia. Antimicrob. Agents Chemother. 2006, 50, 1805–1812. [Google Scholar] [CrossRef]
  57. Halldorsson, S.; Gudjonsson, T.; Gottfredsson, M.; Singh, P.K.; Gudmundsson, G.H.; Baldursson, O. Azithromycin maintains airway epithelial integrity during Pseudomonas aeruginosa infection. Am. J. Respir. Cell Mol. Biol. 2010, 42, 62–68. [Google Scholar] [CrossRef]
  58. Anderson, R.; Theron, A.J.; Feldman, C. Membrane-stabilizing, anti-inflammatory interactions of macrolides with human neutrophils. Inflammation 1996, 20, 693–705. [Google Scholar] [CrossRef]
  59. Feldman, C.; Anderson, R.; Theron, A.J.; Ramafi, G.; Cole, P.J.; Wilson, R. Roxithromycin, clarithromycin, and azithromycin attenuate the injurious effects of bioactive phospholipids on human respiratory epithelium in vitro. Inflammation 1997, 21, 655–665. [Google Scholar] [CrossRef]
  60. Nakano, T.; Ohashi, Y.; Tanaka, A.; Kakinoki, Y.; Washio, Y.; Nakai, Y. Roxythromycin reinforces epithelial defence function in rabbit trachea. Acta Otolaryngol. Suppl. 1998, 538, 233–238. [Google Scholar]
  61. Suh, J.D.; Cohen, N.A.; Palmer, J.N. Biofilms in chronic rhinosinusitis. Curr. Opin. Otolaryngol. Head Neck Surg. 2010, 18, 27–31. [Google Scholar] [CrossRef] [PubMed]
  62. Maina, I.W.; Patel, N.N.; Cohen, N.A. Understanding the Role of Biofilms and Superantigens in Chronic Rhinosinusitis. Curr. Otorhinolaryngol. Rep. 2018, 6, 253–262. [Google Scholar] [CrossRef] [PubMed]
  63. Korkmaz, H.; Ocal, B.; Tatar, E.C.; Tatar, I.; Ozdek, A.; Saylam, G.; Celik, H.H. Biofilms in chronic rhinosinusitis with polyps: Is eradication possible? Eur. Arch. Otorhinolaryngol. 2014, 271, 2695–2702. [Google Scholar] [CrossRef] [PubMed]
  64. Ichimiya, T.; Takeoka, K.; Hiramatsu, K.; Hirai, K.; Yamasaki, T.; Nasu, M. The influence of azithromycin on the biofilm formation of Pseudomonas aeruginosa in vitro. Chemotherapy 1996, 42, 186–191. [Google Scholar] [CrossRef] [PubMed]
  65. Sofer, D.; Gilboa-Garber, N.; Belz, A.; Garber, N.C. ‘Subinhibitory’ erythromycin represses production of Pseudomonas aeruginosa lectins, autoinducer and virulence factors. Chemotherapy 1999, 45, 335–341. [Google Scholar] [CrossRef]
  66. Lim, D.J.; Skinner, D.; McLemore, J.; Rivers, N.; Elder, J.B.; Allen, M.; Koch, C.; West, J.; Zhang, S.; Thompson, H.M.; et al. In-vitro evaluation of a ciprofloxacin and azithromycin sinus stent for Pseudomonas aeruginosa biofilms. Int. Forum Allergy Rhinol. 2020, 10, 121–127. [Google Scholar] [CrossRef]
  67. Nonaka, M.; Pawankar, R.; Saji, F.; Yagi, T. Effect of roxithromycin on IL-8 synthesis and proliferation of nasal polyp fibroblasts. Acta Otolaryngol. Suppl. 1998, 539, 71–75. [Google Scholar]
  68. Nonaka, M.; Pawankar, R.; Tomiyama, S.; Yagi, T. A macrolide antibiotic, roxithromycin, inhibits the growth of nasal polyp fibroblasts. Am. J. Rhinol. 1999, 13, 267–272. [Google Scholar] [CrossRef]
  69. Asano, K.; Kamakazu, K.; Hisamitsu, T.; Suzaki, H. Suppressive activity of macrolide antibiotics on nitric oxide production from nasal polyp fibroblasts in vitro. Acta Otolaryngol. 2003, 123, 1064–1069. [Google Scholar] [CrossRef]
  70. Kanai, K.; Asano, K.; Hisamitsu, T.; Suzaki, H. Suppression of matrix metalloproteinase production from nasal fibroblasts by macrolide antibiotics in vitro. Eur. Respir. J. 2004, 23, 671–678. [Google Scholar] [CrossRef]
  71. Park, H.H.; Park, I.H.; Cho, J.S.; Lee, Y.M.; Lee, H.M. The effect of macrolides on myofibroblast differentiation and collagen production in nasal polyp-derived fibroblasts. Am. J. Rhinol. Allergy 2010, 24, 348–353. [Google Scholar] [CrossRef]
  72. Renteria, A.E.; Maniakas, A.; Mfuna, L.E.; Asmar, M.H.; Gonzalez, E.; Desrosiers, M. Low-dose and long-term azithromycin significantly decreases Staphylococcus aureus in the microbiome of refractory CRS patients. Int. Forum Allergy Rhinol. 2021, 11, 93–105. [Google Scholar] [CrossRef]
  73. Maniakas, A.; Asmar, M.H.; Renteria, A.E.; Nayan, S.; Alromaih, S.; Endam, L.M.; Sampalis, J.S.; Desrosiers, M. Azithromycin in high-risk, refractory chronic rhinosinusitus after endoscopic sinus surgery and corticosteroid irrigations: A double-blind, randomized, placebo-controlled trial. Int. Forum Allergy Rhinol. 2021, 11, 747–754. [Google Scholar] [CrossRef]
  74. Lin, C.F.; Wang, M.C.; Merton, A.T.; Ho, N.H.; Wu, P.S.; Hsu, A.T.; Wang, Y.P. Add-on effect of clarithromycin to oral steroids as post- operative therapy for chronic rhinosinusitis with nasal polyps: A randomised controlled trial. Rhinology 2020, 58, 550–558. [Google Scholar] [CrossRef]
  75. de Oliveira, I.S.; Guimaraes, A.F.; Arantes Pêgas, G.F.; Machado, C.J.; Cassali, G.D.; Crosara, P.; Barbosa Nunes, F.; Gonçalves Becker, H.M.; Santos Guimarães, R.E.; Tormin Borges Crosara, P.F. Azithromycin for chronic eosinophilic rhinosinusitis with nasal polyp: A placebo-controlled trial. Rhinology 2020, 58, 610–617. [Google Scholar] [CrossRef]
  76. Amali, A.; Saedi, B.; Rahavi-Ezabadi, S.; Ghazavi, H.; Hassanpoor, N. Long-term postoperative azithromycin in patients with chronic rhinosinusitis: A randomized clinical trial. Am. J. Rhinol. Allergy 2015, 29, 421–424. [Google Scholar] [CrossRef]
  77. Perić, A.; Baletić, N.; Milojević, M.; Sotirović, J.; Živić, L.; Perić, A.V.; Vojvodić, D. Effects of Preoperative Clarithromycin Administration in Patients with Nasal Polyposis. West Indian Med. J. 2014, 63, 721–727. [Google Scholar]
  78. Videler, W.J.; Badia, L.; Harvey, R.J.; Gane, S.; Georgalas, C.; van der Meulen, F.W.; Menger, D.J.; Lehtonen, M.T.; Toppila-Salmi, S.K.; Vento, S.I.; et al. Lack of efficacy of long-term, low-dose azithromycin in chronic rhinosinusitis: A randomized controlled trial. Allergy 2011, 66, 1457–1468. [Google Scholar] [CrossRef]
  79. Zeng, M.; Wang, H.; Liao, B.; Wang, H.; Long, X.B.; Ma, J.; Liu, J.X.; Cao, P.P.; Ning, Q.; Liu, Z. Comparison of efficacy of fluticasone propionate versus clarithromycin for postoperative treatment of different phenotypic chronic rhinosinusitis: A randomized controlled trial. Rhinology 2019, 57, 101–109. [Google Scholar] [CrossRef]
  80. Zeng, M.; Long, X.B.; Cui, Y.H.; Liu, Z. Comparison of efficacy of mometasone furoate versus clarithromycin in the treatment of chronic rhinosinusitis without nasal polyps in Chinese adults. Am. J. Rhinol. Allergy 2011, 25, e203–e207. [Google Scholar] [CrossRef]
  81. Yamada, T.; Fujieda, S.; Mori, S.; Yamamoto, H.; Saito, H. Macrolide treatment decreased the size of nasal polyps and IL-8 levels in nasal lavage. Am. J. Rhinol. 2000, 14, 143–148. [Google Scholar] [CrossRef] [PubMed]
  82. Bezerra, T.F.P.; Pezato, R.; de Barros, P.M.; Coutinho, L.L.; Costa, L.F.; Pinna, F.; Voegels, R. Prospective evaluation of clarithromycin in recurrent chronic rhinosinusitis with nasal polyps. Braz. J. Otorhinolaryngol. 2021, 87, 298–304. [Google Scholar] [CrossRef] [PubMed]
  83. Jiang, R.S.; Wu, S.H.; Tsai, C.C.; Li, Y.H.; Liang, K.L. Efficacy of Chinese herbal medicine compared with a macrolide in the treatment of chronic rhinosinusitis without nasal polyps. Am. J. Rhinol. Allergy 2012, 26, 293–297. [Google Scholar] [CrossRef] [PubMed]
  84. Deng, J.; Chen, F.; Lai, Y.; Luo, Q.; Xu, R.; Ou, C.; Fu, Q.; Shi, J. Lack of additional effects of long-term, low-dose clarithromycin combined treatment compared with topical steroids alone for chronic rhinosinusitis in China: A randomized, controlled trial. Int. Forum Allergy Rhinol. 2018, 8, 8–14. [Google Scholar] [CrossRef]
  85. Haruna, S.; Shimada, C.; Ozawa, M.; Fukami, S.; Moriyama, H. A study of poor responders for long-term, low-dose macrolide administration for chronic sinusitis. Rhinology 2009, 47, 66–71. [Google Scholar]
  86. Zeng, M.; Wang, H.; Liao, B.; Wang, H.; Long, X.B.; Ma, J.; Liu, J.X.; Liu, Z. Clinical and Biological Markers Predict the Efficacy of Glucocorticoid- and Macrolide-Based Postoperative Therapy in Patients With Chronic Rhinosinusitis. Am. J. Rhinol. Allergy 2021, 35, 596–606. [Google Scholar] [CrossRef]
  87. Suzuki, H.; Ikeda, K.; Honma, R.; Gotoh, S.; Oshima, T.; Furukawa, M.; Takasaka, T. Prognostic factors of chronic rhinosinusitis under long-term low-dose macrolide therapy. ORL J. Otorhinolaryngol. Relat. Spec. 2000, 62, 121–127. [Google Scholar] [CrossRef]
  88. Shen, S.; Lou, H.; Wang, C.; Zhang, L. Macrolide antibiotics in the treatment of chronic rhinosinusitis: Evidence from a meta-analysis. J. Thorac. Dis. 2018, 10, 5913–5923. [Google Scholar] [CrossRef]
  89. Oakley, G.M.; Christensen, J.M.; Sacks, R.; Earls, P.; Harvey, R.J. Characteristics of macrolide responders in persistent post-surgical rhinosinusitis. Rhinology 2018, 56, 111–117. [Google Scholar] [CrossRef]
  90. Stevens, W.W.; Staudacher, A.G.; Hulse, K.E.; Carter, R.G.; Winter, D.R.; Abdala-Valencia, H.; Kato, A.; Suh, L.; Norton, J.E.; Huang, J.H.; et al. Activation of the 15-lipoxygenase pathway in aspirin-exacerbated respiratory disease. J. Allergy Clin. Immunol. 2021, 147, 600–612. [Google Scholar] [CrossRef]
  91. Maniakas, A.; Desrosiers, M. Azithromycin add-on therapy in high-risk postendoscopic sinus surgery patients failing corticosteroid irrigations: A clinical practice audit. Am. J. Rhinol. Allergy 2014, 28, 151–155. [Google Scholar] [CrossRef]
  92. Seresirikachorn, K.; Kerr, S.J.; Aeumjaturapat, S.; Chusakul, S.; Kanjanaumporn, J.; Wongpiyabovorn, J.; Snidvongs, K. Predictive factors for identifying macrolide responder in treating chronic rhinosinusitis. Rhinology 2021, 59, 284–291. [Google Scholar] [CrossRef]
  93. Ichimura, K.; Shimazaki, Y.; Ishibashi, T.; Higo, R. Effect of new macrolide roxithromycin upon nasal polyps associated with chronic sinusitis. Auris Nasus Larynx 1996, 23, 48–56. [Google Scholar] [CrossRef]
  94. Askari, M.; Khezri, M.; Zoghi, G. Introducing a Novel Combination Therapy with Macrolides for the Treatment of Chronic Rhinosinusitis: A Randomized Controlled Trial. Iran. J. Med. Sci. 2022, 47, 533–540. [Google Scholar]
  95. Huang, Z.; Zhou, B. Clarithromycin for the treatment of adult chronic rhinosinusitis: A systematic review and meta-analysis. Int. Forum Allergy Rhinol. 2019, 9, 545–555. [Google Scholar] [CrossRef]
  96. Hashiba, M.; Baba, S. Efficacy of long-term administration of clarithromycin in the treatment of intractable chronic sinusitis. Acta Otolaryngol. Suppl. 1996, 525, 73–78. [Google Scholar]
  97. Seresirikachorn, K.; Suwanparin, N.; Srisunthornphanich, C.; Chitsuthipakorn, W.; Kanjanawasee, D.; Snidvongs, K. Factors of success of low-dose macrolides in chronic sinusitis: Systematic review and meta-analysis. Laryngoscope 2019, 129, 1510–1519. [Google Scholar] [CrossRef]
  98. Nakamura, Y.; Suzuki, M.; Yokota, M.; Ozaki, S.; Ohno, N.; Hamajima, Y.; Nakayama, M.; Hashiba, M.; Murakami, S. Optimal duration of macrolide treatment for chronic sinusitis after endoscopic sinus surgery. Auris Nasus Larynx 2013, 40, 366–372. [Google Scholar] [CrossRef]
  99. Sun, J.; Li, Y. Long-term, low-dose macrolide antibiotic treatment in pediatric chronic airway diseases. Pediatr. Res. 2022, 91, 1036–1042. [Google Scholar] [CrossRef]
  100. Seresirikachorn, K.; Chetthanon, T.; Suwansirisuk, T.; Aeumjaturapat, S.; Chusakul, S.; Kanjanaumporn, J.; Snidvongs, K. Low-dose macrolides for treating pediatric rhinosinusitis: A retrospective study and literature review. SAGE Open Med. 2020, 8, 2050312120933642. [Google Scholar] [CrossRef]
  101. Iino, Y.; Sasaki, Y.; Miyazawa, T.; Kodera, K. Nasopharyngeal flora and drug susceptibility in children with macrolide therapy. Laryngoscope 2003, 113, 1780–1785. [Google Scholar] [CrossRef] [PubMed]
  102. Valery, P.C.; Morris, P.S.; Byrnes, C.A.; Grimwood, K.; Torzillo, P.J.; Bauert, P.A.; Masters, I.B.; Diaz, A.; McCallum, G.B.; Mobberley, C.; et al. Long-term azithromycin for Indigenous children with non-cystic-fibrosis bronchiectasis or chronic suppurative lung disease (Bronchiectasis Intervention Study): A multicentre, double-blind, randomised controlled trial. Lancet Respir. Med. 2013, 1, 610–620. [Google Scholar] [CrossRef] [PubMed]
  103. Ray, W.A.; Murray, K.T.; Hall, K.; Arbogast, P.G.; Stein, C.M. Azithromycin and the risk of cardiovascular death. N. Engl. J. Med. 2012, 366, 1881–1890. [Google Scholar] [CrossRef] [PubMed]
  104. Svanström, H.; Pasternak, B.; Hviid, A. Use of clarithromycin and roxithromycin and risk of cardiac death: Cohort study. BMJ 2014, 349, g4930. [Google Scholar] [CrossRef]
  105. Azuma, A.; Kudoh, S. Securing the safety and efficacy of macrolide therapy for chronic small airway diseases. Intern. Med. 2005, 44, 167–168. [Google Scholar] [CrossRef]
  106. Williamson, E.; Denaxas, S.; Morris, S.; Clarke, C.S.; Thomas, M.; Evans, H.; Direk, K.; Gonzalez-Izquierdo, A.; Little, P.; Lund, V.; et al. Risk of mortality and cardiovascular events following macrolide prescription in chronic rhinosinusitis patients: A cohort study using linked primary care electronic health records. Rhinology 2019, 57, 252–260. [Google Scholar] [CrossRef]
  107. Mosholder, A.D.; Lee, J.Y.; Zhou, E.H.; Kang, E.M.; Ghosh, M.; Izem, R.; Major, J.M.; Graham, D.J. Long-Term Risk of Acute Myocardial Infarction, Stroke, and Death With Outpatient Use of Clarithromycin: A Retrospective Cohort Study. Am. J. Epidemiol. 2018, 187, 786–792. [Google Scholar] [CrossRef]
  108. Inghammar, M.; Nibell, O.; Pasternak, B.; Melbye, M.; Svanström, H.; Hviid, A. Long-Term Risk of Cardiovascular Death With Use of Clarithromycin and Roxithromycin: A Nationwide Cohort Study. Am. J. Epidemiol. 2018, 187, 777–785. [Google Scholar] [CrossRef]
  109. Cavada, M.N.; Grayson, J.W.; Sacks, R. What is the evidence for macrolide therapy in chronic rhinosinusitis? Curr. Opin Otolaryngol. Head Neck Surg. 2020, 28, 6–10. [Google Scholar] [CrossRef]
  110. Reijnders, T.D.Y.; Saris, A.; Schultz, M.J.; van der Poll, T. Immunomodulation by macrolides: Therapeutic potential for critical care. Lancet Respir. Med. 2020, 8, 619–630. [Google Scholar] [CrossRef]
  111. Tojima, I.; Shimizu, S.; Ogawa, T.; Kouzaki, H.; Omura, S.; Sunazuka, T.; Shimizu, T. Anti-inflammatory effects of a novel non-antibiotic macrolide, EM900, on mucus secretion of airway epithelium. Auris Nasus Larynx 2015, 42, 332–336. [Google Scholar] [CrossRef]
Figure 1. Mechanism of action and clinical implications of low-dose, long-term macrolide therapy in chronic rhinosinusitis.
Figure 1. Mechanism of action and clinical implications of low-dose, long-term macrolide therapy in chronic rhinosinusitis.
Ijms 24 09489 g001
Table 1. Randomized placebo-controlled trials using low-dose long-term macrolides in chronic rhinosinusitis.
Table 1. Randomized placebo-controlled trials using low-dose long-term macrolides in chronic rhinosinusitis.
ReferencePatientsNumberNasal PolypMacrolideDuration
Renteria, 2021 [72]Refractory CRS post-FESS4891.7%Azithromycin 250 mg 3 times per week4 months
Maniakas, 2021 [73]Refractory CRS post-FESS12890.0%Azithromycin 250 mg 3 times per week4 months
Lin Chien-Fu, 2020 [74]CRSwNP
post-FESS
126100%Clarithromycin 500 mg3 months
de Oliveira, 2020 [75]AERD59100%Azithromycin 500 mg 3 times per week3 months
Haxel, 2015 [37]CRS post-FESS6755.2%Erythromycin 250 mg3 months
Amali, 2015 [76]CRS post-FESS6642.4%Azithromycin 250 mg3 months
Perić, 2014 [77]CRSwNP80100%Clarithromycin 500 mg2 months
Videler, 2011 [78]CRS6052%Azithromycin 500 mg per week3 months
Wallwork, 2006 [41]CRSsNP640%Roxithromycin 150 mg3 months
CRS, chronic rhinosinusitis; CRSsNP, chronic rhinosinusitis without nasal polyps; CRSwNP, chronic rhinosinusitis with nasal polyps; FESS, functional endoscopic sinus surgery.
Table 2. Randomized controlled trials using low-dose long-term macrolides with comparison or add-on of intranasal corticosteroid sprays in chronic rhinosinusitis.
Table 2. Randomized controlled trials using low-dose long-term macrolides with comparison or add-on of intranasal corticosteroid sprays in chronic rhinosinusitis.
ReferencePatientsNumberNasal PolypMacrolideDurationComparison
Zeng, 2019 [79]CRS post-FESS20570.0%Clarithromycin 250 mg3 monthsFluticasone propionate
Deng, 2018 [76]CRS7456.8%Clarithromycin 250 mg3 monthsBudesonide aqua nasal spray (add-on)
Varvyanskaya, 2014 [36]CRSwNP post-FESS66100%Clarithromycin 250 mg3 months or 6 monthsMometasone furoate (add-on)
Korkmaz, 2014 [63]CRSwNP85100%Clarithromycin 250 mg8 weeksMometasone furoate (add-on)
Zeng, 2011 [80]CRSsNP430%Clarithromycin 250 mg3 monthsMometasone furoate
CRS, chronic rhinosinusitis; CRSsNP, chronic rhinosinusitis without nasal polyps; CRSwNP, chronic rhinosinusitis with nasal polyps; FESS, functional endoscopic sinus surgery.
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

Ryu, G.; Lee, E.; Park, S.I.; Park, M.; Hong, S.D.; Jung, Y.G.; Kim, H.Y. The Mechanism of Action and Clinical Efficacy of Low-Dose Long-Term Macrolide Therapy in Chronic Rhinosinusitis. Int. J. Mol. Sci. 2023, 24, 9489. https://doi.org/10.3390/ijms24119489

AMA Style

Ryu G, Lee E, Park SI, Park M, Hong SD, Jung YG, Kim HY. The Mechanism of Action and Clinical Efficacy of Low-Dose Long-Term Macrolide Therapy in Chronic Rhinosinusitis. International Journal of Molecular Sciences. 2023; 24(11):9489. https://doi.org/10.3390/ijms24119489

Chicago/Turabian Style

Ryu, Gwanghui, Eunkyu Lee, Song I Park, Minhae Park, Sang Duk Hong, Yong Gi Jung, and Hyo Yeol Kim. 2023. "The Mechanism of Action and Clinical Efficacy of Low-Dose Long-Term Macrolide Therapy in Chronic Rhinosinusitis" International Journal of Molecular Sciences 24, no. 11: 9489. https://doi.org/10.3390/ijms24119489

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