Abstract
Pulmonary fibrosis (PF) is a progressive and fatal interstitial lung disease characterized by excessive extracellular matrix deposition and fibroblast activation. Current antifibrotic therapies, such as nintedanib and pirfenidone, slow disease progression but fail to halt fibrosis or significantly improve survival. Macrolides, a class of antibiotics with immunomodulatory and anti-inflammatory properties, have emerged as potential adjunctive therapies for PF. Preclinical studies demonstrate that macrolides attenuate fibrogenesis through multifaceted mechanisms: suppression of TGF-β/Smad and JNK/c-Jun signaling, inhibition of pro-fibrotic cytokine release, modulation of macrophage polarization toward antifibrotic M2 phenotypes, and induction of apoptosis in senescent cells. Clinically, macrolides have shown promise in reducing acute exacerbations in idiopathic pulmonary fibrosis (IPF), mitigating radiation pneumonitis, and attenuating post-infectious fibrotic changes. However, conflicting results from clinical trials and the absence of large-scale randomized studies highlight the need for further validation. This review evaluates the antifibrotic mechanisms and therapeutic potential of macrolides in PF, integrating preclinical and clinical evidence. We aim to inform future research directions by elucidating their role in modulating key pathways and addressing unresolved efficacy questions.
Keywords: antifibrotic drugs, macrolides, immunomodulation, pulmonary fibrosis
Introduction
Pulmonary fibrosis (PF) represents the advanced stage of several diffuse parenchymal lung diseases, 1 particularly interstitial lung disease (ILD), characterized by declining lung function, worsening quality of life, and shortened life expectancy.2,3 Idiopathic pulmonary fibrosis (IPF) is the most prevalent and characteristic form,4,5 with a median life expectancy of only 3–5 years without treatment.6,7 Thirteen to forty percent of patients with non-IPF ILDs also present a progressive fibrosing phenotype. 8 Though current antifibrotic agents such as nintedanib and pirfenidone have shown efficacy in slowing disease progression, they do not cure PF and have only minimally improved mortality rates. Furthermore, immunosuppression has been proven to provide no benefit and could potentially be harmful, particularly in patients with a usual interstitial pneumonia pattern. 9 Overall, the management of PF remains challenging, necessitating ongoing research into effective therapies and strategies to improve patient outcomes.
Though triggers, susceptibility factors, and initial inflammatory responses can differ among fibrosing ILDs,6,8,10 common mechanisms become prominent in the later phases. 11 Persistent micro-injuries to alveolar epithelial cells activate developmental signaling pathways, notably transforming growth factor (TGF)-β. This leads to cellular dysfunction, disrupted equilibrium in the alveolar epithelial-mesenchymal unit, progressive deposition of extracellular matrix (ECM) proteins, epithelial-to-mesenchymal transition (EMT), and subsequent distortion of lung structures 12 (Figure 1.). Recent studies have shown that lung dysbiosis, characterized by increased bacterial load and reduced microbial diversity, has been observed in IPF patients and is associated with disease progression and immune response, 13 potentially contributing to acute exacerbations, hospitalizations, and decreased survival. 14
Figure 1.
Regulatory mechanisms and functional characteristics of macrolides in pulmonary fibrosis. Macrolides inhibit EMT, fibroblast proliferation, and apoptosis resistance in both AECs and fibroblasts, ultimately suppressing the deposition of ECM proteins and preventing the formation of scar tissue and distortion of lung homeostasis. Moreover, macrolides promote the transition of activated macrophages from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype and mobilize other immune cells, including neutrophils and lymphocytes, to participate in the anti-fibrosis process.
AEC, alveolar epithelial cells; ECM, extracellular matrix; EMT, epithelial-to-mesenchymal transition; M1, classically activated or inflammatory macrophages; M2, alternatively activated or anti-inflammatory macrophages.
Macrolides, a group of antibiotics structured with the macrocyclic lactone ring structure, exert antimicrobial effects by binding to the 50S ribosomal subunit of the bacterial 70S ribosome to impede RNA-dependent protein synthesis. 15 While traditionally used against respiratory pathogens, 16 their immunomodulatory and anti-inflammatory properties have garnered increasing attention.17 –19 Preclinical and clinical studies suggest macrolides may mitigate fibrotic processes through mechanisms extending beyond antimicrobial effects, positioning them as potential adjunctive therapies for PF.20 –22 This review systematically evaluates macrolides’ antifibrotic mechanisms and therapeutic potential in PF, integrating preclinical and clinical evidence to clarify their role in key pathways and unresolved efficacy questions.
We conducted a comprehensive literature search in PubMed to identify relevant studies on macrolides in PF, including all available records through November 2024, while limiting the search to English-language publications. No lower date restriction was applied to ensure the inclusion of all historical evidence. To ensure comprehensive coverage, we supplemented the electronic search by manually screening reference lists from selected original research articles and review papers. The search strategy incorporated the following key terms: “macrolide” and its main members (“clarithromycin,” “erythromycin,” “roxithromycin,” and “azithromycin”), combined with disease-related terms including “pulmonary fibrosis,” “lung fibrosis,” “idiopathic pulmonary fibrosis,” “IPF,” “interstitial lung disease,” and “ILD.” To capture mechanistic and clinical aspects, we also included terms like “cytokine,” “neutrophil,” “pneumonia,” “pulmonary infection,” “acute lung injury,” “acute respiratory distress syndrome,” and “ARDS.” The screening process followed a three-stage approach: (1) initial review of titles and abstracts for relevance; (2) full-text evaluation when abstracts provided insufficient information; and (3) exclusion of clearly irrelevant studies. Finally, all included studies were systematically categorized into preclinical studies (in vitro/animal models) and clinical studies (human trials).
Macrolides and the non-antimicrobial effects
Macrolides, such as azithromycin (AZM), erythromycin (EM), roxithromycin (RXM), and clarithromycin (CAM), exhibit multifaceted immunomodulatory and anti-inflammatory properties that extend beyond their conventional antibacterial roles. 23 These pleiotropic effects are mediated through diverse molecular mechanisms, including: (1) Suppression of inflammatory signaling pathways,24,25 leading to downregulation of pro-inflammatory cytokines and chemokines while enhancing anti-inflammatory mediators like IL-10; (2) Modulation of innate and adaptive immunity via macrophage polarization,20,26,27 inhibition of neutrophil extracellular trap formation, and regulation of T-cell differentiation; (3) Mucoregulatory actions, including suppression of mucus hypersecretion by blocking EGFR/MAPK-dependent MUC5AC expression and reducing goblet cell hyperplasia28,29; (4) Biofilm disruption30 –32 through interference with bacterial quorum sensing and ECM components, enhancing antibiotic penetration; (5) Antiviral and epithelial repair-promoting effects, potentially via inhibition of viral replication and augmentation of autophagy-mediated epithelial barrier restoration.33 –35
Clinically, these properties have positioned macrolides as disease-modifying agents in chronic respiratory disorders.36,37 Low-dose, long-term macrolide therapy is now a cornerstone in diffuse panbronchiolitis and a guideline-recommended option for frequent exacerbators with COPD or non-cystic fibrosis bronchiectasis,38,39 demonstrating significant reductions in exacerbation frequency and sputum volume. Emerging evidence supports their utility in severe asthma, 40 particularly with neutrophilic phenotypes, post-viral airway hyperreactivity, 41 and cystic fibrosis. 42
Emerging evidence highlights macrolides’ potential to attenuate PF through mechanisms partially overlapping with chronic airway diseases, including TGF-β/Smad signaling suppression, inhibition of pro-fibrotic cytokines, and macrophage M2 polarization. However, it remains unclear whether their efficacy arises from broad immunomodulation or PF-specific pathways—and whether clinical evidence supports their therapeutic use.
Antifibrotic mechanisms of macrolides
Macrolides have emerged as potential therapeutic agents for PF due to their multifaceted mechanisms of action. Both animal models and in vitro studies suggest a therapeutic role for macrolides in attenuating fibrotic processes.20,43 –45 Below, we categorize their effects based on distinct molecular and cellular pathways (Table 1).
Table 1.
Key antifibrotic and anti-inflammatory effects of macrolides in pulmonary fibrosis and acute lung injury: evidence from preclinical animal and in vitro studies.
| Macrolides members | Mechanism of action | Key findings | [Ref.] |
|---|---|---|---|
| AZM | Suppressed TGF-β1/Smad and JNK/c-Jun pathways | Reduced fibrotic markers (α-SMA, collagen I, LOX, LOXL-2), inflammation, and collagen deposition in mice; inhibited TGF-β1-driven fibrosis in lung fibroblasts | [45] |
| AZM | Shifted macrophage polarization to M2 phenotype | Reduced pro-inflammatory cytokines (IL-12, IL-6), increased IL-10, enhanced M2 markers, attenuated M1 markers | [27] |
| AZM | Promoted NOX4 degradation | Suppressed TGF-β-induced myofibroblast differentiation; reduced lung fibrosis | [74] |
| AZM | Impaired lysosomal function/autophagy | Reduced collagen secretion and pro-fibrotic gene expression in IPF fibroblasts | [73] |
| AZM | Restored Th1/Th2 balance | Reversed IL-10 dominance to IFN-γ dominance; reduced inflammation and fibrosis | [84] |
| AZM | — | Alleviated radiation-induced lung injury by reducing oxidative damage and cytokines | [66] |
| AZM | Modulated immunity | Reduced bleomycin-induced fibrosis and improved lung function | [20] |
| AZM | Upregulated Nrf2/HO-1 | Attenuated cisplatin-induced ALI by reducing oxidative stress and inflammation | [67] |
| AZM | Downregulated p53 | Attenuated BLM-induced ALI by reducing inflammation | [81] |
| AZM | Downregulated Mettl3 | Attenuated LPS-induced epithelial cell damage and promoted M2 polarization | [53] |
| Deglycosylated-AZM | Regulated TGF-β/Smad signaling | Inhibited myofibroblast activation and EMT; showed anti-inflammatory and antioxidant effects; improved lung function | [76] |
| EM | — | Improved survival, reduced inflammatory cell infiltration, suppressed NO overproduction in viral pneumonia | [64] |
| EM | — | Prevented LPS-induced microvascular leakage and neutrophil recruitment | [60] |
| EM | Suppressed TNF-α/PDGF | Ameliorated fibrosis by reducing collagen deposition | [43] |
| EM | — | Reduced bleomycin-induced acute lung injury when pretreated | [62] |
| EM; EM703 | Inhibited Smad3 mRNA, restored Smad7, reduced p-Smad2/3 | Decreased fibroblast proliferation and collagen production | [48] |
| EM703 | Regulated TGF-β/Smad signal | Reduced inflammation and inhibited fibroblast proliferation in bleomycin-induced mice | [49] |
| CAM | — | Reduced bacterial load and oxidative stress in K. pneumoniae infection | [70] |
| CAM | Inhibited NF-κB | Reduced ventilator-induced lung injury | [51] |
| CAM | — | Reduced radiation-induced pneumonitis and fibrosis markers | [69] |
| CAM; EM | — | Inhibited LPS-induced goblet cell secretion and neutrophil recruitment | [57] |
| CAM; RXM; Josamycin | — | Reduced bleomycin-induced acute lung injury (neutrophil infiltration, edema); decreased epithelial apoptosis | [58] |
| CAM; EM; RXM | Inhibited VCAM-1 | Reduced fibrosis markers and neutrophil/macrophage migration | [61] |
| RXM | Induced apoptosis via NOX4 pathway, inhibited SASP | Reduced TGF-β-induced fibroblast activation, lung injury, inflammation, and fibrosis | [44] |
| RXM; EM; Josamycin | Shifted macrophage polarization to M2 phenotype | Reduced IgG-ICx-induced lung injury by decreasing cytokine release (IL-1β, TNF-α) and NO production | [63] |
| Nafithromycin | — | Reduced inflammation in LPS-ALI (comparable to high-dose AZM) | [59] |
| Tylvalosin | Suppressed NF-κB/PLA2 | Reduced inflammation and oxidative stress | [52] |
| Telithromycin | Inhibited NF-κB | Reduced airway inflammation through dual-phase action | [68] |
Inhibition of TGF-β1/Smad signaling pathway
Macrolides, including AZM, EM, and its derivative EM703, have demonstrated significant potential in inhibiting the TGF-β/Smad signaling pathway, a central driver of fibrosis.46,47 Our team has clarified AZM attenuated bleomycin-induced PF in mice by suppressing TGF-β1/Smad signaling, leading to reduced expression of fibrotic markers such as α-smooth muscle actin (α-SMA), type I collagen, LOX, and LOXL-2, as well as decreased collagen deposition. 45 We also found that AZM inhibited TGF-β1-induced fibrotic responses in primary lung fibroblasts, further supporting its role in blocking TGF-β1-mediated fibrosis. 45 Similarly, EM and EM703 inhibited TGF-β-induced Smad3 mRNA expression, restored Smad7 levels, and reduced p-Smad2/3 protein activation, thereby suppressing fibroblast proliferation and collagen production in human lung fibroblasts, 48 while EM703 also attenuated inflammation and regulated TGF-β/Smad signaling in bleomycin-induced lung fibrosis in mice. 49
Inhibition of NF-κB signaling pathway
The NF-κB signaling pathway plays a vital role in regulating inflammation, immune responses, and fibrosis. 50 Macrolides have been shown to exert antifibrotic effects by modulating NF-κB activation and downstream inflammatory responses. The inhibition of NF-κB signaling by macrolides exerts multifaceted antifibrotic effects by suppressing the expression of pro-inflammatory cytokines 51 (e.g., IL-1β, TNF-α, IL-6, and IL-8), which are key drivers of inflammation and fibrosis, while also attenuating neutrophil recruitment through the downregulation of adhesion molecules 51 (e.g., E-selectin) and chemokines, thereby reducing tissue damage. In addition, NF-κB inhibition downregulates pro-fibrotic mediators such as TGF-β, which are central to fibroblast activation and ECM deposition, and promotes inflammation resolution by enhancing inflammatory cell apoptosis and reducing oxidative stress. 52 Furthermore, macrolides protect epithelial cells from apoptosis and ferroptosis, 53 preserving lung tissue integrity and function, collectively contributing to their therapeutic potential in PF.
Suppression of JNK/c-Jun signaling pathway
JNK1 promotes collagen deposition and pulmonary fibrosis by enhancing TGF-β1 signaling and pro-fibrotic gene expression in response to cellular stress and environmental insults.54,55 AZM has demonstrated inhibitory effects on this pathway. In the same study by our research team, 45 AZM attenuated bleomycin-induced PF in mice by inhibiting the JNK/c-Jun signaling pathway, leading to reduced inflammation and fibrosis.
Anti-inflammatory and antioxidant effects
The severity of acute lung injury (ALI) directly correlates with the extent of subsequent chronic inflammation and fibrosis, 56 suggesting that early intervention using the anti-inflammatory properties of macrolides may be valuable in modulating fibrotic mechanisms. Pretreatment with macrolides significantly decreased neutrophil infiltration,57 –60 inflammatory cell migration, 61 and lung edema 62 in ALI mice models. Macrolides, including EM, RXM, and josamycin, reduced inflammation by decreasing cytokines (IL-1β and TNF-α), iNOS expression, and NO production, as shown in influenza-virus-induced pneumonia and IgG immune complex-induced lung injury models.63,64 Similarly, radiation-induced lung injury (RILI) is a significant complication of thoracic radiotherapy, caused by the direct toxic effects of radiation on lung tissue, followed by an inflammatory cell infiltration that leads to interstitial pneumonia and fibrotic repair processes. 65 AZM attenuated the irradiation-induced increases in the mRNA expression of fibrotic markers in RILI mice by reducing inflammatory cytokine expression (IL-1β, IL-6, TNF-α, TGF-β1) and oxidative damage. 66 AZM attenuated cisplatin-induced ALI in rats by reducing inflammation via upregulation of Nrf2/HO-1 signaling and PPARγ, and also inhibiting necroptosis by enhancing cytoglobin and SIRT1-mediated oxidative stress responses. 67 Deglycosylated azithromycin (Deg-AZM) and telithromycin both exhibited anti-inflammatory by suppressing NF-κB activation, phospholipase A2 activity, decreasing inflammatory mediators, and enhancing inflammatory cell apoptosis. 68 These findings collectively highlight the shared anti-inflammatory mechanisms of macrolides across different models of lung injury.69,70
Modulation of autophagy and lysosomal function
Autophagy dysregulation is implicated in fibrosis.71,72 AZM has been shown to impair lysosomal function and autophagy in IPF fibroblasts, which not only reduces collagen secretion and pro-fibrotic gene expression 73 but also promotes NOX4 degradation through enhanced proteasome activity and autophagy inhibition, ultimately suppressing myofibroblast differentiation. 74
Inhibition of myofibroblast activation and EMT
EMT, a process where alveolar epithelial cells lose epithelial traits and acquire mesenchymal properties, 75 significantly contributes to fibrosis by expanding the myofibroblast pool. AZM has been shown to inhibit EMT and myofibroblast activation, key drivers of fibrosis. Specifically, AZM promotes NOX4 degradation, 74 suppressing TGF-β-induced myofibroblast differentiation and reducing lung fibrosis. In addition, Deg-AZM attenuates bleomycin-induced PF by further inhibiting EMT and myofibroblast activation. 76 These findings highlight AZM’s dual role in targeting both NOX4/TGF-β signaling and EMT, offering a multifaceted approach to mitigating fibrosis.
Induction of apoptosis in senescent cells
Cellular senescence, particularly in fibroblasts and epithelial cells, contributes to chronic inflammation and fibrosis.77,78 Senescent cells accumulate in fibrotic tissues and release pro-inflammatory cytokines, growth factors, and ECM components, collectively known as the senescence-associated secretory phenotype (SASP). 79 AZM and RXM were identified as novel senolytic drugs that preferentially target senescent human fibroblasts, inducing autophagy and metabolic changes, 80 EM did not demonstrate comparable senolytic effects. In addition, RXM targeted senescent cells by inducing apoptosis and inhibiting SASP factors through the NOX4 pathway, which reduced TGF-β-induced fibroblast activation, and diminished lung injury, inflammation, and fibrosis in bleomycin-induced mice. 44 AZM nanostructured lipid carrier (AZ-NLC) was shown to enhance senescent cell clearance in bleomycin-induced lung injury models through p53 downregulation and Bcl-2 expression modulation, further supporting the senotherapeutic potential of macrolides in PF. 81
Restoration of immune cell balance
Macrolides play a crucial role in regulating immune cell balance by targeting multiple immune cells.70,82,83 AZM promotes macrophage polarization toward the anti-inflammatory M2 phenotype by downregulating Mettl3 and inactivating the NF-κB pathway, leading to reduced pro-inflammatory cytokines (IL-12, IL-6) and increased anti-inflammatory IL-10, enhanced M2 markers (mannose receptor, CD23, arginase activity). 27 This polarization supports the tissue repair process and may help prevent pathological fibrosis. AZM can also reverse the Th2-like response (IL-10 dominance) to a Th1-like response (IFN-γ dominance) in bleomycin-induced PF in rats, 84 reducing inflammatory cell infiltration and fibrosis at the early stage of fibrosing alveolitis.
Inhibition of mucus production
Macrolides have been shown to exert beneficial effects in chronic airway diseases through their ability to reduce mucus viscosity and enhance mucociliary clearance,85,86 contributing to improved pulmonary function and decreased infection risk in affected patients. Experimental studies using guinea pig tracheal models have demonstrated that CAM and EM can effectively suppress LPS-induced goblet cell secretion and attenuate neutrophil recruitment, 57 suggesting the potential anti-inflammatory effect. While these findings are promising, further targeted trials are needed to assess their additional effects in patients with PF. In recent years, rapid advancements have been made in developing drug delivery systems for PF. 87 For instance, inhalable nanoparticles loaded with antifibrotic drugs enable rapid and site-specific delivery to the pulmonary airways, offering potential early relief for patients. 88 However, the airway mucus layer poses a natural barrier to inhaled drug delivery. In this context, macrolides may play an adjunctive role in antifibrotic therapy by indirectly suppressing mucus production.
Modulation of lung microbiota
In chronic pulmonary diseases such as PF, reduced microbial diversity and increased bacterial load are closely linked to disrupted pulmonary homeostasis, which is associated with poor clinical outcomes and disease progression.13,89,90 Biofilms, structured bacterial clusters encased in a self-produced matrix 91 , can persist in the lungs, promote chronic inflammation, and complicate infections, thereby contributing to disease progression. 92 Notably, chronic airway inflammation in PF patients is often accompanied by increased harboring of potentially pathogenic pathogens, such as Haemophilus, Neisseria, and Streptococcus species than healthy controls. 93 Patients with AE-IPF showed significant respiratory microbiome alterations as well, including increased Campylobacter and Stenotrophomonas species compared to stable controls. 94 This dysbiosis may further contribute to airway damage and fibrosis progression through biofilm-mediated persistent stimulation.93,95,96 Macrolides exert multifaceted effects on bacterial biofilms by disrupting quorum sensing,32,97 suppressing virulence factors,98,99 and degrading established biofilm matrices. 31 They simultaneously inhibit bacterial-epithelial adhesion 30 while enhancing mucosal clearance and epithelial repair. These combined mechanisms ameliorate biofilm-mediated inflammation and bacterial persistence, potentially mitigating disease progression in chronic respiratory conditions. Based on current evidence, macrolides may represent a potential adjunctive therapy combined with antifibrotic medications to provide new therapeutic possibilities. However, this hypothesis requires further validation through additional preclinical studies.
Potential prokinetic effects of macrolides
Evidence suggests that gastroesophageal reflux disease (GERD) and esophageal dysmotility may contribute to the development and progression of PF.100,101 As potent motilin receptor agonists, macrolides enhance gastrointestinal motility, accelerate gastric emptying, and reduce GERD frequency. 102 This prokinetic effect may synergize with their direct anti-inflammatory properties, potentially alleviating reflux-mediated lung injury and improving PF-related symptoms such as chronic cough. 103 However, the clinical efficacy of targeting GERD in PF remains contentious. A pooled analysis of randomized controlled trials demonstrated that proton pump inhibitors slowed forced vital capacity decline in IPF patients, although they failed to improve survival or reduce hospitalization rates 104 and may even increase the risk of severe respiratory infections in advanced IPF. 105 Further prospective studies are warranted to delineate the relative contributions of macrolides’ dual mechanisms—prokinetic effects and direct antifibrotic actions—in modulating PF progression.
Clinical evidence on macrolides in PF
Macrolides exert antifibrotic effects through multiple mechanisms, including inhibition of TGF-β1/Smad and JNK/c-Jun signaling, anti-inflammatory and antioxidant effects, modulation of autophagy and lysosomal function, induction of apoptosis in senescent cells, and restoration of immune balance, making macrolides a potential candidate for the adjuvant treatment of PF. However, further clinical studies are needed to validate these findings and translate them into therapeutic applications. This section summarizes key clinical studies, highlighting their findings and implications for PF management (Table 2).
Table 2.
Macrolides in pulmonary fibrosis: Clinical evidence summary.
| [Ref.] | Study type | Intervention | Main findings | Limitation |
|---|---|---|---|---|
| [113] | RCT | AZM 500 mg 3×/week ×12 week | No improvement in cough (LCQ/VAS) in IPF; more GI side effects | Small sample (n = 25), high dropout (20%), mostly male, unmeasured confounders |
| [103] | Open-label trial | AZM 250 mg qd ×3 months | 49.6% cough reduction in sarcoidosis | No control (n = 21), selection bias |
| [118] | Retrospective cohort study | CAM 200 mg qd ×3 months | ↓Grade⩾2 RP post-IMRT (3% vs 18%) | Retrospective (n = 89), selection bias |
| [117] | Retrospective cohort study | CAM 200 mg qd ×3 months | ↓Grade⩾2 RP (9.6% vs 16%) and Grade⩾3 (0.73% vs 3.8%) post-SBRT | Retrospective, no randomization, different follow-up time |
| [22] | Retrospective cohort study | AZM 250 mg 3×/week ⩾12 months | ↓All-cause hospitalizations by 72%, no lung function decline, well tolerated (93% adherence). | Retrospective, selection bias, unclassified admission causes |
| [108] | Retrospective cohort study | AZM 500mg qd ×5 days + steroids | ↓60-day mortality in AE-IPF (26% vs 70%) compared with fluoroquinolone therapy | Retrospective (n = 85), selection bias, temporal bias (10 years) |
| [107] | Prospective (historical controls) | AZM 500 mg qd ×5 days + steroids (partial) | ↓60-day mortality in AE-CFIP (20% vs 69.6%) compared with fluoroquinolone therapy | Open-label (n = 20), retrospective, historical controls |
| [109] | Retrospective cohort study | Macrolides (unclear regimen) + steroids | Improved survival in rapid IPF progression (OR = 0.37) | Retrospective, unclear regimens, unmeasured confounders |
| [106] | Retrospective cohort study | EM 400 mg qd CAM 200–400 mg qd (long-term) |
↓AE incidence (13.8% vs 34.8%), ↑36 months survival | Retrospective (n = 52), selection bias, unclear regimens |
| [115] | Case report | EM 400 mg qd ×4 year | Improved HRCT/FVC in HLA-B54+ interstitial pneumonia patient | N = 1, no controls |
| [119] | Case series | AZM 500 mg q3w ⩾3 months + pirfenidone + steroids | Improved symptoms/radiology in post-H1N1 fibrosis | N = 3, no controls |
| [116] | Case report | CAM 500 mg qd→250 mg qd maintenance | Stabilized airway-centered fibrosis after steroid failure | N = 1, no controls |
| [114] | Case report | CAM 500 mg bid→maintenance | Improved desquamative interstitial pneumonia symptoms/CT | N = 1, no controls |
RP, radiation pneumonitis.
Effects on acute exacerbations in PF
Long-term macrolide therapy has been associated with a reduction in acute exacerbations (AEs) in PF. A retrospective cohort study found that patients receiving EM or CAM experienced significantly fewer AEs and improved 36-month survival rates. 106 These benefits are likely attributed to the anti-inflammatory properties of macrolides in long-term use, which may suppress the inflammatory cascade driving disease progression.
Two studies investigated the short-term use of AZM during acute exacerbations of IPF and chronic fibrosing interstitial pneumonia (AE-CFIP).107,108 In IPF patients, short-term AZM combined with corticosteroids significantly reduced 60-day mortality. 108 Similarly, in AE-CFIP patients, AZM reduced 60-day mortality and improved median survival time. 107 A retrospective study of IPF patients with rapidly progressing respiratory failure found that macrolide use was associated with improved survival, 109 suggesting a potential benefit in AEs when combined with high-dose corticosteroids. However, current guidelines emphasize that corticosteroids lack definitive benefit in IPF exacerbations and may even contribute to adverse outcomes. 110 Notably, while macrolides possess immunomodulatory effects, their acute therapeutic effects in these studies are likely mediated primarily through antibacterial or antiviral activity against pathogens, rather than immune modulation.35,111 The potential role of immunomodulation in AEs remains unverified and requires further high-quality evidence. 112
Potential improvement in prognosis and symptoms in PF
In addition to reducing AEs, long-term prophylactic macrolide therapy has been associated with improved survival in IPF patients. 106 Another retrospective observational study demonstrated that prophylactic AZM significantly reduced all-cause non-elective hospitalizations in IPF patients and decreased rescue antibiotic use. 22 These benefits were achieved without impacting the rate of decline in lung function, suggesting that macrolides may exert their effects through mechanisms beyond direct antifibrotic activity, such as reducing infection-related complications that contribute to disease progression. However, a randomized controlled trial 113 found that low-dose AZM did not improve cough-related quality of life, cough severity, or objective cough frequency in IPF patients, with more frequent gastrointestinal side effects, suggesting that efficacy may vary depending on dosing regimens and patient populations.
Long-term macrolide therapy has shown benefits in chronic pulmonary diseases beyond IPF. A non-controlled open-label trial found that long-term AZM therapy significantly reduced cough frequency by about 49.6% and improved quality of life in pulmonary sarcoidosis patients. 103 However, the noncontrolled design and small sample size limit causal inference, highlighting the need for robust trials to confirm the findings.
Several case reports114 –116 described improvements in symptoms and radiological findings in patients with specific subtypes of interstitial pneumonia, such as airway-centered interstitial fibrosis 116 and desquamative interstitial pneumonia. 114 These cases suggest that macrolides may slow disease progression in certain PF subtypes, such as ILD patients with human leukocyte antigen (HLA)-B54 positivity. 115
Prophylactic use in RP
Macrolides have been explored for their prophylactic role in reducing radiation pneumonitis (RP), a common complication of thoracic radiotherapy. 65 Two retrospective cohort studies reported that prophylactic administration of CAM significantly reduced the incidence of Grade ⩾2 RP and Grade ⩾3 RP.117,118 These effects are likely mediated by the suppression of radiation-induced inflammation, highlighting the potential of macrolides in mitigating treatment-related complications. However, the optimal CAM regimen of dose/duration and protective durability against RP remains unclear.
Role in post-infectious PF
Macrolides have also been investigated for their potential role in mitigating fibrotic changes following severe respiratory infections. A case series reported that AZM combined with pirfenidone and prednisolone improved symptoms, effort tolerance, and radiological findings in three patients with PF following H1N1 ARDS, 119 suggesting its adjunctive value to antifibrotic therapy potentially through the anti-inflammatory and immunomodulatory effects.
Prospects of macrolides in PF treatment
While preclinical and early clinical studies highlight the promising therapeutic potential of macrolides in PF, further research is essential to establish them as a standard treatment option. Key areas for future investigation include large-scale randomized controlled trials, optimization of dosing and duration, timing of administration, and deeper mechanistic insights into their antifibrotic and anti-inflammatory effects. In addition, long-term safety and efficacy evaluations, as well as real-world studies, are crucial to confirm their clinical benefits and pave the way for personalized and effective treatment strategies in PF.
Protective effects on alveolar epithelial cells during early injury: Understanding whether macrolides can prevent excessive damage to AECs early in the injury process could offer a preventive strategy for PF progression.
Fibroblast, endothelial cell, and EMT mechanisms: Further research into how macrolides affect these key cells and processes will deepen our understanding of their antifibrotic potential.
Combination therapies: Exploring combinations of macrolides with other antifibrotic agents, as well as the molecular mechanisms behind their synergy, could lead to more effective treatment protocols, while also fully considering potential adverse effects and drug-drug interactions to ensure safety and efficacy in individualized treatment plans.
Macrolide modifications: Engineering macrolides to enhance their antifibrotic effects, such as increasing their anti-inflammatory or immunosuppressive properties, is a potential direction for future research.
Personalized Medicine: Identifying which patients with PF are most likely to benefit from macrolide therapy based on their EMT profiles and TGF-β activity could optimize treatment strategies. Personalized medicine approaches may include genetic testing or biomarkers to predict responses to macrolides.
Rigorous clinical trials: Randomized controlled trials and real-world studies will be necessary to determine the long-term benefits of macrolides in PF and solidify their place in clinical practice.
Conclusion
Macrolides show considerable potential in treating PF, owing to their anti-inflammatory and antimicrobial properties, as well as the ability to modulate key cellular processes such as fibroblast activation, EMT, and cellular senescence. Their combination with other antifibrotic therapies may offer a multifaceted approach to treating this complex and progressive disease. Ongoing research is needed to confirm their efficacy in clinical settings and to explore potential improvements through combination therapies and drug modifications.
Acknowledgments
None.
Footnotes
ORCID iDs: Qingqing Jia
https://orcid.org/0009-0004-8880-4309
Xiang Tong
https://orcid.org/0009-0007-2412-3901
Contributor Information
Qingqing Jia, Department of Pulmonary Medicine and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China.
Qian Wang, Department of Pulmonary Medicine and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China.
Qilong Zhou, Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China.
Hong Fan, Department of Pulmonary Medicine and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, China.
Xiang Tong, Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Guoxuexiang 37, Chengdu, Sichuan 610041, China.
Declarations
Ethics approval and consent to participate: Not applicable.
Consent for publication: Not applicable.
Author contributions: Qingqing Jia: Conceptualization; Writing – original draft.
Qian Wang: Writing – review & editing.
Qilong Zhou: Writing – review & editing.
Hong Fan: Supervision.
Xiang Tong: Conceptualization; Writing – original draft.
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This research was funded by the National Natural Science Foundation of China, grant number 82304593.
Competing interests: The authors declare that there is no conflict of interest.
Availability of data and materials: Not applicable.
References
- 1. Wijsenbeek M, Cottin V. Spectrum of fibrotic lung diseases. N Engl J Med 2020; 383: 958–968. [DOI] [PubMed] [Google Scholar]
- 2. Cottin V, Hirani NA, Hotchkin DL, et al. Presentation, diagnosis and clinical course of the spectrum of progressive-fibrosing interstitial lung diseases. Eur Respir Rev 2018; 27: 180076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Liu GY, Budinger GRS, Dematte JE. Advances in the management of idiopathic pulmonary fibrosis and progressive pulmonary fibrosis. BMJ 2022; 377: e066354. [DOI] [PubMed] [Google Scholar]
- 4. Cottin V, Wollin L, Fischer A, et al. Fibrosing interstitial lung diseases: knowns and unknowns. Eur Respir Rev 2019; 28: 180100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. George PM, Spagnolo P, Kreuter M, et al. Progressive fibrosing interstitial lung disease: clinical uncertainties, consensus recommendations, and research priorities. Lancet Respir Med 2020; 8: 925–934. [DOI] [PubMed] [Google Scholar]
- 6. Lederer DJ, Martinez FJ. Idiopathic pulmonary fibrosis. N Engl J Med 2018; 378: 1811–1823. [DOI] [PubMed] [Google Scholar]
- 7. Montesi SB, Fisher JH, Martinez FJ, et al. Update in interstitial lung disease 2019. Am J Respir Crit Care Med 2020; 202: 500–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Wijsenbeek M, Kreuter M, Olson A, et al. Progressive fibrosing interstitial lung diseases: current practice in diagnosis and management. Curr Med Res Opin 2019; 35: 2015–2024. [DOI] [PubMed] [Google Scholar]
- 9. Idiopathic Pulmonary Fibrosis Clinical Research Network; Raghu G, Anstrom KJ, et al. Prednisone, azathioprine, and N-acetylcysteine for pulmonary fibrosis. N Engl J Med 2012; 366: 1968–1977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Wijsenbeek M, Suzuki A, Maher TM. Interstitial lung diseases. Lancet 2022; 400: 769–786. [DOI] [PubMed] [Google Scholar]
- 11. Distler JHW, Györfi A-H, Ramanujam M, et al. Shared and distinct mechanisms of fibrosis. Nat Rev Rheumatol 2019; 15: 705–730. [DOI] [PubMed] [Google Scholar]
- 12. Fernandez IE, Eickelberg O. New cellular and molecular mechanisms of lung injury and fibrosis in idiopathic pulmonary fibrosis. Lancet 2012; 380: 680–688. [DOI] [PubMed] [Google Scholar]
- 13. Martinez FJ, Yow E, Flaherty KR, et al. Effect of antimicrobial therapy on respiratory hospitalization or death in adults with idiopathic pulmonary fibrosis: the CleanUP-IPF randomized clinical trial. JAMA 2021; 325: 1841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Morris A, Gibson K, Collman RG. The lung microbiome in idiopathic pulmonary fibrosis. What does it mean and what should we do about it? Am J Respir Crit Care Med 2014; 190: 850–852. [DOI] [PubMed] [Google Scholar]
- 15. Bahal N, Nahata MC. The new macrolide antibiotics: azithromycin, clarithromycin, dirithromycin, and roxithromycin. Ann Pharmacother 1992; 26: 46–55. [DOI] [PubMed] [Google Scholar]
- 16. Asano K, Tryka E, Cho JS, et al. Macrolide therapy in chronic inflammatory diseases. Mediators Inflamm 2012; 2012: 692352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Cameron EJ, McSharry C, Chaudhuri R, et al. Long-term macrolide treatment of chronic inflammatory airway diseases: risks, benefits and future developments. Clin Exp Allergy J Br Soc Allergy Clin Immunol 2012; 42: 1302–1312. [DOI] [PubMed] [Google Scholar]
- 18. Jaffé A, Bush A. Anti-inflammatory effects of macrolides in lung disease. Pediatr Pulmonol 2001; 31: 464–473. [DOI] [PubMed] [Google Scholar]
- 19. Mrowietz U. Macrolide immunosuppressants. Eur J Dermatol EJD 1999; 9: 346–351. [PubMed] [Google Scholar]
- 20. Wuyts WA, Willems S, Vos R, et al. Azithromycin reduces pulmonary fibrosis in a bleomycin mouse model. Exp Lung Res 2010; 36: 602–614. [DOI] [PubMed] [Google Scholar]
- 21. Gijs P-J, Daccord C, Bernasconi E, et al. Azithromycin alters spatial and temporal dynamics of airway microbiota in idiopathic pulmonary fibrosis. ERJ Open Res 2023; 9: 00720–02022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Macaluso C, Maritano Furcada J, Alzaher O, et al. The potential impact of azithromycin in idiopathic pulmonary fibrosis. Eur Respir J 2019; 53: 1800628. [DOI] [PubMed] [Google Scholar]
- 23. Kanoh S, Rubin BK. Mechanisms of action and clinical application of macrolides as immunomodulatory medications. Clin Microbiol Rev 2010; 23: 590–615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Niessen NM, Gibson PG, Baines KJ, et al. Sputum TNF markers are increased in neutrophilic and severe asthma and are reduced by azithromycin treatment. Allergy 2021; 76: 2090–2101. [DOI] [PubMed] [Google Scholar]
- 25. Zimmermann P, Ziesenitz VC, Curtis N, et al. The immunomodulatory effects of macrolides-a systematic review of the underlying mechanisms. Front Immunol 2018; 9: 302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Arampatzioglou A, Papazoglou D, Konstantinidis T, et al. Clarithromycin enhances the antibacterial activity and wound healing capacity in type 2 diabetes mellitus by increasing LL-37 load on neutrophil extracellular traps. Front Immunol 2018; 9: 2064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Murphy BS, Sundareshan V, Cory TJ, et al. Azithromycin alters macrophage phenotype. J Antimicrob Chemother 2008; 61: 554–560. [DOI] [PubMed] [Google Scholar]
- 28. Imamura Y, Yanagihara K, Mizuta Y, et al. Azithromycin inhibits MUC5AC production induced by the Pseudomonas aeruginosa autoinducer N-(3-Oxododecanoyl) homoserine lactone in NCI-H292 cells. Antimicrob Agents Chemother 2004; 48: 3457–3461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Shao MXG, Nadel JA. Neutrophil elastase induces MUC5AC mucin production in human airway epithelial cells via a cascade involving protein kinase C, reactive oxygen species, and TNF-alpha-converting enzyme. J Immunol Baltim Md 1950 2005; 175: 4009–4016. [DOI] [PubMed] [Google Scholar]
- 30. Piatti G. Bacterial adhesion to respiratory mucosa and its modulation by antibiotics at sub-inhibitory concentrations. Pharmacol Res 1994; 30: 289–299. [DOI] [PubMed] [Google Scholar]
- 31. Starner TD, Shrout JD, Parsek MR, et al. Subinhibitory concentrations of azithromycin decrease nontypeable Haemophilus influenzae biofilm formation and diminish established biofilms. Antimicrob Agents Chemother 2008; 52: 137–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Tateda K, Comte R, Pechere J-C, et al. Azithromycin inhibits quorum sensing in Pseudomonas aeruginosa. Antimicrob Agents Chemother 2001; 45: 1930–1933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Slater M, Torr E, Harrison T, et al. The differential effects of azithromycin on the airway epithelium in vitro and in vivo. Physiol Rep 2016; 4: e12960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Ling K-M, Garratt LW, Banerjee B, et al. Azithromycin partially mitigates dysregulated repair of lung allograft small airway epithelium. Transplantation 2020; 104: 1166–1176. [DOI] [PubMed] [Google Scholar]
- 35. Oliver ME, Hinks TSC. Azithromycin in viral infections. Rev Med Virol 2021; 31: e2163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. López-Boado YS, Rubin BK. Macrolides as immunomodulatory medications for the therapy of chronic lung diseases. Curr Opin Pharmacol 2008; 8: 286–291. [DOI] [PubMed] [Google Scholar]
- 37. Kricker JA, Page CP, Gardarsson FR, et al. Nonantimicrobial actions of macrolides: overview and perspectives for future development. Pharmacol Rev 2021; 73: 233–262. [DOI] [PubMed] [Google Scholar]
- 38. Lee E, Sol IS, Kim JD, et al. Long-term macrolide treatment for non-cystic fibrosis bronchiectasis in children: a meta-analysis. Sci Rep 2021; 11: 24287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Yao G-Y, Ma Y-L, Zhang M-Q, et al. Macrolide therapy decreases chronic obstructive pulmonary disease exacerbation: a meta-analysis. Respir Int Rev Thorac Dis 2013; 86: 254–260. [DOI] [PubMed] [Google Scholar]
- 40. Wong EHC, Porter JD, Edwards MR, et al. The role of macrolides in asthma: current evidence and future directions. Lancet Respir Med 2014; 2: 657–670. [DOI] [PubMed] [Google Scholar]
- 41. Khezri MR, Zolbanin NM, Ghasemnejad-berenji M, et al. Azithromycin: immunomodulatory and antiviral properties for SARS-CoV-2 infection. Eur J Pharmacol 2021; 905: 174191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Southern KW, Solis-Moya A, Kurz D, et al. Macrolide antibiotics (including azithromycin) for cystic fibrosis. Cochrane Database Syst Rev 2024; 2: CD002203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Chen B, Jiang L, Zhao W, et al. Ameliorating effect of erythromycin on bleomycin-induced pulmonary fibrosis: role of alveolar macrophage activation and cytokine release. Respirol Carlton Vic 1997; 2: 151–155. [DOI] [PubMed] [Google Scholar]
- 44. Zhang X, Dong Y, Li W-C, et al. Roxithromycin attenuates bleomycin-induced pulmonary fibrosis by targeting senescent cells. Acta Pharmacol Sin 2021; 42: 2058–2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Tong X, Zhang S, Wang D, et al. Azithromycin attenuates bleomycin-induced pulmonary fibrosis partly by inhibiting the expression of LOX and LOXL-2. Front Pharmacol 2021; 12: 709819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Enzel D, Kriventsov M, Sataieva T, et al. Cellular and molecular genetic mechanisms of lung fibrosis development and the role of vitamin D: a review. Int J Mol Sci 2024; 25: 8946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Hu H-H, Chen D-Q, Wang Y-N, et al. New insights into TGF-β/smad signaling in tissue fibrosis. Chem Biol Interact 2018; 292: 76–83. [DOI] [PubMed] [Google Scholar]
- 48. Yu C, Azuma A, Li Y, et al. EM703, a new derivative of erythromycin, inhibits transforming growth factor-beta signaling in human lung fibroblasts. Exp Lung Res 2008; 34: 343–354. [DOI] [PubMed] [Google Scholar]
- 49. Li YJ, Azuma A, Usuki J, et al. EM703 improves bleomycin-induced pulmonary fibrosis in mice by the inhibition of TGF-beta signaling in lung fibroblasts. Respir Res 2006; 7: 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Sauer A, Peukert K, Putensen C, et al. Antibiotics as immunomodulators: a potential pharmacologic approach for ARDS treatment. Eur Respir Rev 2021; 30: 210093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Amado-Rodríguez L, González-López A, López-Alonso I, et al. Anti-inflammatory effects of clarithromycin in ventilator-induced lung injury. Respir Res 2013; 14: 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Zhao Z, Tang X, Zhao X, et al. Tylvalosin exhibits anti-inflammatory property and attenuates acute lung injury in different models possibly through suppression of NF-κB activation. Biochem Pharmacol 2014; 90: 73–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Xu S, Xing J, Zheng L, et al. Azithromycin regulates Mettl3-mediated NF-κB pathway to enhance M2 polarization of RAW264.7 macrophages and attenuate LPS-triggered cytotoxicity of MLE-12 alveolar cells. Int Immunopharmacol 2024; 137: 112426. [DOI] [PubMed] [Google Scholar]
- 54. van der Velden JLJ, Ye Y, Nolin JD, et al. JNK inhibition reduces lung remodeling and pulmonary fibrotic systemic markers. Clin Transl Med 2016; 5: 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Alcorn JF, van der Velden J, Brown AL, et al. c-jun N-terminal kinase 1 is required for the development of pulmonary fibrosis. Am J Respir Cell Mol Biol 2009; 40: 422–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Shen AS, Haslett C, Feldsien DC, et al. The intensity of chronic lung inflammation and fibrosis after bleomycin is directly related to the severity of acute injury. Am Rev Respir Dis 1988; 137: 564–571. [DOI] [PubMed] [Google Scholar]
- 57. Tamaoki J, Takeyama K, Yamawaki I, et al. Lipopolysaccharide-induced goblet cell hypersecretion in the guinea pig trachea: Inhibition by macrolides. Am J Physiol 1997; 272: L15-19. [DOI] [PubMed] [Google Scholar]
- 58. Kawashima M, yatsunami J, Fukuno Y, et al. Inhibitory effects of 14-membered ring macrolide antibiotics on bleomycin-induced acute lung injury. Lung 2002; 180: 73–89. [DOI] [PubMed] [Google Scholar]
- 59. Trivedi J, Shaikh J, Chavan N, et al. Pretreatment of nafithromycin attenuates inflammatory response in murine lipopolysaccharide induced acute lung injury. Cytokine 2020; 129: 155049. [DOI] [PubMed] [Google Scholar]
- 60. Tamaoki J, Tagaya E, Yamawaki I, et al. Effect of erythromycin on endotoxin-induced microvascular leakage in the rat trachea and lungs. Am J Respir Crit Care Med 1995; 151: 1582–1588. [DOI] [PubMed] [Google Scholar]
- 61. Li Y, Azuma A, Takahashi S, et al. Fourteen-membered ring macrolides inhibit vascular cell adhesion molecule 1 messenger RNA induction and leukocyte migration: Role in preventing lung injury and fibrosis in bleomycin-challenged mice. Chest 2002; 122: 2137–2145. [DOI] [PubMed] [Google Scholar]
- 62. Azuma A, Furuta T, Enomoto T, et al. Preventive effect of erythromycin on experimental bleomycin-induced acute lung injury in rats. Thorax 1998; 53: 186–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Tamaoki J, Kondo M, Kohri K, et al. Macrolide antibiotics protect against immune complex-induced lung injury in rats: role of nitric oxide from alveolar macrophages. J Immunol Baltim Md 1950 1999; 163: 2909–2915. [PubMed] [Google Scholar]
- 64. Sato K, Suga M, Akaike T, et al. Therapeutic effect of erythromycin on influenza virus-induced lung injury in mice. Am J Respir Crit Care Med 1998; 157: 853–857. [DOI] [PubMed] [Google Scholar]
- 65. Tsoutsou PG, Koukourakis MI. Radiation pneumonitis and fibrosis: mechanisms underlying its pathogenesis and implications for future research. Int J Radiat Oncol Biol Phys 2006; 66: 1281–1293. [DOI] [PubMed] [Google Scholar]
- 66. Tang F, Li R, Xue J, et al. Azithromycin attenuates acute radiation-induced lung injury in mice. Oncol Lett 2017; 14: 5211–5220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Hassanein EHM, Sayed GA, Alzoghaibi AM, et al. Azithromycin mitigates cisplatin-induced lung oxidative stress, inflammation and necroptosis by upregulating SIRT1, PPARγ, and Nrf2/HO-1 signaling. Pharm Basel Switz 2022; 16: 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Leiva M, Ruiz-Bravo A, Jimenez-Valera M. Effects of telithromycin in in vitro and in vivo models of lipopolysaccharide-induced airway inflammation. Chest 2008; 134: 20–29. [DOI] [PubMed] [Google Scholar]
- 69. Lee SJ, Yi C, Heo RW, et al. Clarithromycin attenuates radiation-induced lung injury in mice. PloS One 2015; 10: e0131671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Kumar V, Harjai K, Chhibber S. Effect of clarithromycin on lung inflammation and alveolar macrophage function in klebsiella penumoniae B5055-induced acute lung infection in BALB/c mice. J Chemother Florence Italy 2008; 20: 609–614. [DOI] [PubMed] [Google Scholar]
- 71. Liao S-X, Sun P-P, Gu Y-H, et al. Autophagy and pulmonary disease. Ther Adv Respir Dis 2019; 13: 1753466619890538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Ornatowski W, Lu Q, Yegambaram M, et al. Complex interplay between autophagy and oxidative stress in the development of pulmonary disease. Redox Biol 2020; 36: 101679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Krempaska K, Barnowski S, Gavini J, et al. αAzithromycin has enhanced effects on lung fibroblasts from idiopathic pulmonary fibrosis (IPF) patients compared to controls. Respir Res 2020; 21: 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Tsubouchi K, Araya J, Minagawa S, et al. Azithromycin attenuates myofibroblast differentiation and lung fibrosis development through proteasomal degradation of NOX4. Autophagy 2017; 13: 1420–1434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Mottais A, Riberi L, Falco A, et al. Epithelial-mesenchymal transition mechanisms in chronic airway diseases: a common process to target? Int J Mol Sci 2023; 24: 12412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Ruan H, Gao S, Li S, et al. Deglycosylated azithromycin attenuates bleomycin-induced pulmonary fibrosis via the TGF-β1 signaling pathway. Mol Basel Switz 2021; 26: 2820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Hohmann MS, Habiel DM, Coelho AL, et al. Quercetin enhances ligand-induced apoptosis in senescent idiopathic pulmonary fibrosis fibroblasts and reduces lung fibrosis in vivo. Am J Respir Cell Mol Biol 2019; 60: 28–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Sellarés J, Rojas M. Quercetin in idiopathic pulmonary fibrosis: another brick in the senolytic wall. Am J Respir Cell Mol Biol 2019; 60: 3–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Birch J, Gil J. Senescence and the SASP: Many therapeutic avenues. Genes Dev 2020; 34: 1565–1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Ozsvari B, Nuttall JR, Sotgia F, et al. Azithromycin and roxithromycin define a new family of ‘senolytic’ drugs that target senescent human fibroblasts. Aging 2018; 10: 3294–3307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Alrashedi MG, Ali AS, Ahmed OA, et al. Local delivery of azithromycin nanoformulation attenuated acute lung injury in mice. Mol Basel Switz 2022; 27: 8293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Wang L, Wang D, Zhang T, et al. The role of immunometabolism in macrophage polarization and its impact on acute lung injury/acute respiratory distress syndrome. Front Immunol 2023; 14: 1117548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Altenburg J, de Graaff CS, van der Werf TS, et al. Immunomodulatory effects of macrolide antibiotics – part 1: biological mechanisms. Respir Int Rev Thorac Dis 2011; 81: 67–74. [DOI] [PubMed] [Google Scholar]
- 84. Ma J, He B, Li N, et al. Intervention by azithromycin on bleomycin-induced lung injury in rats and its mechanisms. Zhonghua Jie He He Hu Xi Za Zhi Zhonghua Jiehe He Huxi Zazhi Chin J Tuberc Respir Dis; 25: 392–395. [PubMed] [Google Scholar]
- 85. Shimizu T, Shimizu S, Hattori R, et al. 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. [DOI] [PubMed] [Google Scholar]
- 86. Tanabe T, Kanoh S, Tsushima K, et al. Clarithromycin inhibits interleukin-13-induced goblet cell hyperplasia in human airway cells. Am J Respir Cell Mol Biol 2011; 45: 1075–1083. [DOI] [PubMed] [Google Scholar]
- 87. Diwan R, Bhatt HN, Beaven E, et al. Emerging delivery approaches for targeted pulmonary fibrosis treatment. Adv Drug Deliv Rev 2024; 204: 115147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Ferguson LT, Ma X, Myerson JW, et al. Mechanisms by which liposomes improve inhaled drug delivery for alveolar diseases. Adv Nanobiomed Res 2023; 3: 2200106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Faverio P, Bini F, Vaghi A, et al. Long-term macrolides in diffuse interstitial lung diseases. Eur Respir Rev 2017; 26: 170082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Molyneaux PL, Willis-Owen SAG, Cox MJ, et al. Host-microbial interactions in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2017; 195: 1640–1650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Costerton JW, Stewart PS, Greenberg EP. Bacterial biofilms: a common cause of persistent infections. Science 1999; 284: 1318–1322. [DOI] [PubMed] [Google Scholar]
- 92. Aparna MS, Yadav S. Biofilms: microbes and disease. Braz J Infect Dis 2008; 12: 526–530. [DOI] [PubMed] [Google Scholar]
- 93. Molyneaux PL, Cox MJ, Willis-Owen SAG, et al. The role of bacteria in the pathogenesis and progression of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2014; 190: 906–913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Molyneaux PL, Cox MJ, Wells AU, et al. Changes in the respiratory microbiome during acute exacerbations of idiopathic pulmonary fibrosis. Respir Res 2017; 18: 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. O’Dwyer DN, Ashley SL, Gurczynski SJ, et al. Lung microbiota contribute to pulmonary inflammation and disease progression in pulmonary fibrosis. Am J Respir Crit Care Med 2019; 199: 1127–1138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Warheit-Niemi HI, Edwards SJ, SenGupta S, et al. Fibrotic lung disease inhibits immune responses to staphylococcal pneumonia via impaired neutrophil and macrophage function. JCI Insight 2022; 7: e152690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Hoffmann N, Lee B, Hentzer M, et al. Azithromycin blocks quorum sensing and alginate polymer formation and increases the sensitivity to serum and stationary-growth-phase killing of Pseudomonas aeruginosa and attenuates chronic P. aeruginosa lung infection in Cftr−/− mice. Antimicrob Agents Chemother 2007; 51: 3677–3687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Sandri A, Ortombina A, Boschi F, et al. Inhibition of Pseudomonas aeruginosa secreted virulence factors reduces lung inflammation in CF mice. Virulence 2018; 9: 1008–1018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Alhajlan M, Alhariri M, Omri A. Efficacy and safety of liposomal clarithromycin and its effect on Pseudomonas aeruginosa virulence factors. Antimicrob Agents Chemother 2013; 57: 2694–2704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Lee JS, Ryu JH, Elicker BM, et al. Gastroesophageal reflux therapy is associated with longer survival in patients with idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2011; 184: 1390–1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Raghu G, Freudenberger TD, Yang S, et al. High prevalence of abnormal acid gastro-oesophageal reflux in idiopathic pulmonary fibrosis. Eur Respir J 2006; 27: 136–142. [DOI] [PubMed] [Google Scholar]
- 102. Broad J, Sanger GJ. The antibiotic azithromycin is a motilin receptor agonist in human stomach: comparison with erythromycin. Br J Pharmacol 2013; 168: 1859–1867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Fraser SD, Thackray-Nocera S, Shepherd M, et al. Azithromycin for sarcoidosis cough: an open-label exploratory clinical trial. ERJ Open Res 2020; 6: 00534–02020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Lee JS, Collard HR, Anstrom KJ, et al. Anti-acid treatment and disease progression in idiopathic pulmonary fibrosis: an analysis of data from three randomised controlled trials. Lancet Respir Med 2013; 1: 369–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Kreuter M, Wuyts W, Renzoni E, et al. Antacid therapy and disease outcomes in idiopathic pulmonary fibrosis: a pooled analysis. Lancet Respir Med 2016; 4: 381–389. [DOI] [PubMed] [Google Scholar]
- 106. Kuse N, Abe S, Hayashi H, et al. Long-term efficacy of macrolide treatment in idiopathic pulmonary fibrosis: a retrospective analysis. Sarcoidosis Vasc Diffuse Lung Dis 2016; 33: 242–246. [PubMed] [Google Scholar]
- 107. Kawamura K, Ichikado K, Suga M, et al. Efficacy of azithromycin for treatment of acute exacerbation of chronic fibrosing interstitial pneumonia: a prospective, open-label study with historical controls. Respir Int Rev Thorac Dis 2014; 87: 478–484. [DOI] [PubMed] [Google Scholar]
- 108. Kawamura K, Ichikado K, Yasuda Y, et al. Azithromycin for idiopathic acute exacerbation of idiopathic pulmonary fibrosis: a retrospective single-center study. BMC Pulm Med 2017; 17: 94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Oda K, Yatera K, Fujino Y, et al. Efficacy of concurrent treatments in idiopathic pulmonary fibrosis patients with a rapid progression of respiratory failure: an analysis of a national administrative database in Japan. BMC Pulm Med 2016; 16: 91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Raghu G, Remy-Jardin M, Richeldi L, et al. Idiopathic pulmonary fibrosis (an update) and progressive pulmonary fibrosis in adults: an official ATS/ERS/JRS/ALAT clinical practice guideline. Am J Respir Crit Care Med 2022; 205: e18–e47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Hirai K, Homma T, Yamaguchi F, et al. Acute exacerbation of idiopathic pulmonary fibrosis induced by pertussis: the first case report. BMC Pulm Med 2019; 19: 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Kolb M, Bondue B, Pesci A, et al. Acute exacerbations of progressive-fibrosing interstitial lung diseases. Eur Respir Rev 2018; 27: 180071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Guler SA, Clarenbach C, Brutsche M, et al. Azithromycin for the treatment of chronic cough in idiopathic pulmonary fibrosis: a randomized controlled crossover trial. Ann Am Thorac Soc 2021; 18: 2018–2026. [DOI] [PubMed] [Google Scholar]
- 114. Knyazhitskiy A, Masson RG, Corkey R, et al. Beneficial response to macrolide antibiotic in a patient with desquamative interstitial pneumonia refractory to corticosteroid therapy. Chest 2008; 134: 185–187. [DOI] [PubMed] [Google Scholar]
- 115. Ishimoto H, Sakamoto N, Hara A, et al. Interstitial pneumonia with HLA-B54 antigen that responded well to erythromycin. Intern Med 2022; 61: 3559–3562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Jouneau S, Kerjouan M, Caulet-Maugendre S, et al. Clarithromycin stops lung function decline in airway-centered interstitial fibrosis. Respiration 2013; 85: 156–159. [DOI] [PubMed] [Google Scholar]
- 117. Takeda A, Tsurugai Y, Sanuki N, et al. Clarithromycin mitigates radiation pneumonitis in patients with lung cancer treated with stereotactic body radiotherapy. J Thorac Dis 2018; 10: 247–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Ohguri T, Morisaki T, Itamura H, et al. Prophylactic effect of clarithromycin on radiation pneumonitis in IMRT for lung cancer. Anticancer Res 2024; 44: 2989–2995. [DOI] [PubMed] [Google Scholar]
- 119. Saha A, Vaidya PJ, Chavhan VB, et al. Combined pirfenidone, azithromycin and prednisolone in post- H1N1 ARDS pulmonary fibrosis. [DOI] [PMC free article] [PubMed] [Google Scholar]

