Publication Cover
Canadian Journal of Respiratory, Critical Care, and Sleep Medicine
Revue canadienne des soins respiratoires et critiques et de la médecine du sommeil
Volume 7, 2023 - Issue 4
967
Views
0
CrossRef citations to date
0
Altmetric
Clinical Respiratory Review

Bronchiectasis: From targets to therapies

Pages 206-212 | Received 22 Mar 2023, Accepted 07 Jul 2023, Published online: 27 Jul 2023

Abstract

Bronchiectasis is a chronic obstructive airway disease characterized by abnormal bronchial dilatation and the accumulation of muco-purulent endoluminal secretions. Progressive lung function deterioration, frequent bouts of respiratory infection and daily productive cough negatively impact the lives of affected individuals. Multiple causal factors can initiate bronchiectasis; however, defects in host defenses represent a cardinal feature. The first line of airway defense against inhaled pathogens is mucociliary clearance. Systemic host defense defects, particularly those affecting opsonophagocytosis and killing of bacteria are equally essential to prevent bronchiectasis. Substantial evidence suggests that unopposed neutrophil protease activity in the airway lumen leads to the loss of bronchial elasticity and excessive accumulation of mucus. The focus of this review is on the advances in our understanding of pathogenic mechanisms that are bringing new targeted therapies for individuals suffering from the clinical manifestations of bronchiectasis.

RÉSUMÉ

La bronchectasie est une maladie obstructive chronique des voies respiratoires caractérisée par une dilatation anormale des bronches et l’accumulation de sécrétions endoluminales muco-purulentes. La détérioration progressive de la fonction pulmonaire, les épisodes fréquents d’infection respiratoire et la toux productive quotidienne ont des répercussions négatives sur la vie des personnes touchées. De multiples facteurs causaux peuvent êre à l’origine de la bronchectasie, mais les déficiences dans les mécanismes de défense de l’hôte représentent une caractéristique essentielle. La première ligne de défense des voies respiratoires contre les agents pathogènes inhalés est la clairance mucociliaire. Les déficiences systémiques des mécanismes de défense de l’hôte, en particulier celles qui affectent l’opsonophagocytose et la destruction des bactéries, sont tout aussi essentielles pour prévenir la bronchectasie. Des preuves considérables indiquent que l’activité protéasique des neutrophiles sans opposition dans la lumière des voies respiratoires entraîne une perte d’élasticité bronchique et une accumulation excessive de mucus. Cette revue se concentre sur les progrès dans notre compréhension des mécanismes pathogènes qui apportent de nouvelles thérapies ciblées pour les personnes souffrant des manifestations cliniques de la bronchectasie.

Introduction

Bronchiectasis is a chronic lung disease characterized by sustained, abnormally enlarged bronchi containing excessive stagnant, concentrated mucus that fuels an infamous “vicious circle” of infection, neutrophil elastase-dominated inflammation, airflow obstruction and tissue destruction.Citation1,Citation2 The causes of bronchiectasis are numerous, heterogeneous and vary considerably according to the age and geographic region of affected individuals ().Citation3 Characterization of genetic, microbiological and environmental features in individuals is fundamental to the management of bronchiectasis. Unfortunately for most individuals, a treatable cause of bronchiectasis cannot be identified. However, a better understanding of treatable traits that define endotypes as suggested by Flume et al.,Citation4 is likely to improve patient care and accelerate the discovery of effective new therapies that slow or stop disease progression.

Table 1. Causes of bronchiectasis.

Several morphological types of bronchiectasis are associated with bronchomalacia (airway wall destruction), including cylindrical, varicose and cystic or saccular bronchiectasis.Citation5 A distinct category of bronchiectasis is traction bronchiectasis in which bronchomalacia is not prominent, but dilatation is generated by an increase in the tension of peri-bronchial fibers acting on the airways. Clinical manifestations of traction bronchiectasis correlate with the severity of the underlying interstitial lung disease.Citation6 The focus of the current review is on bronchiectasis with bronchomalacia, not traction bronchiectasis.

The prevalence of bronchiectasis is highest in women and in the elderly. Etiologies vary according to sex, age and geography. Causes of bronchiectasis have changed due to improved public health policies and patient care. Data from the European bronchiectasis EMBARC registry suggests that in Europe bronchiectasis following infections with tuberculosis, non-tuberculous mycobacteria (NTM), childhood Bordetella pertussis and other viruses have declined while COPD, asthma, connective tissue disease and immunodeficiency are more prevalent causes of bronchiectasis.Citation7 Causes of bronchiectasis vary greatly between countries, with idiopathic and post-infectious accounting for between 40% and 70% of cases. In most areas of the world infections predisposing to bronchiectasis remain prominent.Citation3 Documentation reporting the causes of bronchiectasis in many parts of the world remains sparse or absent.

The airway obstruction associated with bronchiectasis largely results from a loss of tissue elasticity and the accumulation of viscous secretions. The increase in airway compliance and mucus are both closely linked to the unabated presence of neutrophil proteases—especially human leukocyte elastase (HLE).Citation8,Citation9

HLE present in neutrophil granules is essential for normal host defense against gram negative bacteria.Citation10 A potential pitfall of targeting cellular HLE could be an increase in susceptibility to gram negative sepsis.Citation11 However, extracellular HLE represents an attractive therapeutic target.

The sustained presence of free, unopposed HLE in the extracellular compartment of the bronchial lumen is associated with worse infection, increased exacerbations and airway destruction in both cystic fibrosis (CF) and non-CF bronchiectasis.Citation12,Citation13 Several proteins involved in airway structural integrity and host defenses are cleaved by HLE (reviewed by Voynow and Shinbashi).Citation14 Proteases can cleave connective tissues thus increasing airway compliance.Citation15 Furthermore, active extracellular HLE degrades several opsonins including immunoglobulin IgG, SP-A, SP-D, the complement protein fragment C3bi and the complement receptor CR1, creating an opsonin-receptor mismatch that suppresses phagocytosis and killing of bacterial pathogens.Citation16,Citation17 The HLE-mediated phagocytosis and killing defect in bronchiectasis is reversed by in vivo inhibition of HLE using aerosolized alpha-1 antitrypsin.Citation18 HLE also increases mRNA transcripts of the secreted airway mucin gene MUC5AC and induces airway mucus hypersecretion, a cardinal feature of bronchiectasis.Citation19,Citation20 Other pro-inflammatory effects of HLE are the increased synthesis of the neutrophil chemoattractant IL-8 and cleavage of the macrophage phosphatidylserine receptor thus inhibiting macrophage clearance of apoptotic neutrophils (efferocytosis), which is essential for resolution of inflammation.Citation21,Citation22 HLE also proteolyzes the key airway anti-bacterial host defense proteins transferrin, lactoferrin and midkine.Citation14 Finally, HLE can cleave host airway antiproteases, alpha-1 antitrypsin and secretory leukoprotease inhibitor (SLPI).Citation23

Therapeutic attempts to directly inhibit HLE with alpha1 antitrypsin and other antiproteases have thus far been disappointing.Citation14 The failure to translate HLE inhibition into an approved therapy for bronchiectasis largely reflects the difficulty of conducting definitive specific interventions in such a heterogeneous and complex disease, indicating that targeted treatments should be trialed in subgroups of patients where there may be a stronger rationale for targeting one of the pathogenic pathways leading to bronchiectasis morbidity. It may also suggest that if we are to improve the lives of people living with bronchiectasis, we need to focus on more than a single target.

Our understanding of the pathogenesis of bronchiectasis has greatly evolved in recent years. The remarkable improvements observed in individuals with CF who have benefited from the advent of highly effective cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapy has sharpened our focus on mucociliary clearance as one of the major airway host defenses against chronic bacterial infection and tissue destruction.Citation24,Citation25 Defective CFTR function is not limited to CF and is observed in healthy smokers and individuals with chronic obstructive pulmonary disease (COPD).Citation26,Citation27 CFTR dysfunction also is induced by both host and pathogen-derived toxic products.Citation28,Citation29 However, translating therapies that have proven helpful in CF to non-CF bronchiectasis is not simple. DNase I (dornase alpha) therapy improves lung function and decreases respiratory exacerbations in CF but has no beneficial effect in non-CF bronchiectasis, and may even cause harm.Citation30 Similarly, inhaled tobramycin markedly increases lung function in CFCitation31 but has a more modest effect on lung function in non-CF bronchiectasis.Citation32 The reasons for the differences in clinical responses between CF and non-CF bronchiectasis are poorly understood and may relate to CFTR-specific mechanisms, the severity of the CF airway mucus clearance defect or both. Regardless, knowledge obtained from studies of CF help inform us about the pathogenesis of bronchiectasis and potential therapeutic targets.

Therapeutic targets

Major targets for the development of new therapies in non-CF bronchiectasis are listed in . Some of the mechanisms associated with each of these targets have been extensively studied in both CF and non-CF bronchiectasis. The therapeutic interventions addressing each target have been the object of informative clinical trials in CF and several of these novel approaches are being studied in non-CF bronchiectasis.

Table 2. Potential targets for therapeutic intervention in bronchiectasis.

Muco-ciliary clearance

Formal airway clearance techniques (ACT) remain the cornerstone of therapy in bronchiectasis. Clinical trials of ACT present challenges in study design, patient recruitment, placebo control, blinding and length of study. However, a randomized placebo-controlled study over 1 year of the ACT known as ETGOL (slow expiration with the glottis opened in the lateral posture) in bronchiectasis suggests that there are numerous potential benefits of ACT.Citation33 These included improvements in the number of respiratory exacerbations, quality of life (St. George’s Respiratory Questionnaire) and decreased cough (Leicester Cough Questionnaire).

Airway mucus is predominantly comprised of the secreted airway mucins Muc5B, Muc5AC and small amounts of Muc2. Airway secreted mucins play 2 major host defense roles. First mucins, particularly Muc5B, are key to the trapping and clearing of inhaled pathogens and harmful particles through the mucociliary escalator.Citation34 Second, the anionic charge of airway mucins provides airway cells with an effective barrier protecting against oxidants and proteases.Citation35–37 These 2 roles of mucins are in opposition of one another since adequate airway clearance is associated with more fluid mucus whereas effective barrier protection requires thicker, viscous mucus. The successful switch back and forth from fluid to viscous mucus is a dynamic process dictated by numerous airway epithelial chloride and sodium channels of which CFTR plays a leading role. CFTR responds to environmental queues such as the chronic oxidative stress of repeated cigarette smoke exposure that decreases CFTR function and increases the protective mucus barrier. However, sustained accumulation of concentrated mucins in the airways as observed in certain CFTR deficiency states (CF, COPD), non-CF bronchiectasis and ciliary dyskinesia can also protect inhaled pathogens against host defenses and favor the proliferation of airway bacteria.Citation38–41 Recent data also suggest that increased mucin concentration and viscoelastic properties of spontaneously produced sputum predict sputum eosinophilia.Citation42 These observations point to the importance of several potential interventions aimed at restoring healthy mucus transport in non-CF bronchiectasis such as smoking cessation, CFTR modulators and effective mucolytic agents to reduce mucus viscosity and improve mucociliary clearance.Citation43,Citation44

The advent of highly effective CFTR modulator therapy (HEMT) in CF renders the clinical course of CF more like that of non-CF bronchiectasis and raises the question of whether HEMT-treated CF patients should be included in future bronchiectasis clinical trials. The answer is probably no. First, CFTR deficiency affects systemic immune responses which may not be fully normalized by HEMT. Second, many CF individuals have persistent severe pancreatic insufficiency, diabetes and liver abnormalities that can impact their ability to respond to infectious respiratory exacerbations, making them clearly distinct from those with non-CF bronchiectasis. Finally, the acquisition of unusual pathogens such as Burkholderia cepacia at an early age in CF is a distinguishing feature that can make comparisons with non-CF bronchiectasis imperfect.

Host immune defects

Immune deficiencies associated with reduced amounts of IgG, IgA and/or IgM immunoglobulins are clearly linked to an increased risk of developing bronchiectasis.Citation45 An increase in serum IgE may point to specific entities, particularly allergic bronchopulmonary aspergillosis or an immune dysregulation causing a hypereosinophilic syndrome. Both primary and secondary humoral immune deficiencies are significant risk factors for bronchiectasis, and serum immunoglobins should be assessed early after a diagnosis of bronchiectasis of unknown cause. Therapeutic interventions to compensate for immune deficiencies have shown significant benefit and novel Ig-replacement therapies continue to be explored.Citation46

Neutrophilic inflammation

The neutrophil serine proteases (NSPs) HLE, cathepsin G and proteinase III are activated through cathepsin C-dependent (CatC, also known as dipeptidyl peptidase 1, DPP-1) cleavage of an amino terminal dipeptide during the promyelocyte phase of neutrophil maturation.Citation47 Inhibitors of CatC block NSP activation. The CatC inhibitor AZD7986 (brensocatib) has shown a favorable early safety and tolerability profile in healthy humans.Citation48 Furthermore, a phase 2 clinical trial demonstrated that brensocatib prolongs the time to first exacerbation in non-CF bronchiectasis.Citation49 However, a potential pitfall of CatC inhibition is a decrease in host defense as reported in studies of olfactomedin-4. Olfactomedin-4 (OLFM4) is a matrix glycoprotein of neutrophil-specific granules which interacts with CatC and decreases the maturation of NSPs.Citation50 OLFM4 is present in 20–30% of circulating neutrophils from healthy individuals, and is more highly expressed during sepsis, acute respiratory distress syndrome and shock.Citation51 Neutrophils from OLFM4-/- mice have an increased killing capacity against Staphylococcus aureus and E. coli. In contrast, individuals with an elevated percentage of OLFM4 positive neutrophils (≥37.6%) had a higher 60-day mortality than those with a lower (<37.6%) percentage of OLFM4-positive neutrophils (60-day mortality: 56% in the high group, vs 18% in the low group, p < 0.001, n = 120).Citation52 A possible interpretation of these results is that OLFM4 inhibits CatC-dependent activation of key NSPs critical to innate immune responses, thus increasing the risk of mortality during sepsis.

HLE-rich neutrophil extracellular traps (NET) have also been associated with non-CF bronchiectasis and are decreased with chronic macrolide therapy (discussed in a subsequent section) in the subgroup of patients with elevated markers of NETosis.Citation53 Finally, HLE inhibitors continue to garner interest but have yet to demonstrate sustained benefit in clinically meaningful outcomes.Citation54

Bacterial load

Individuals with bronchiectasis and chronic Pseudomonas aeruginosa (PA) infection of their airways are at a much higher risk of respiratory exacerbations, an accelerated rate of decline of lung function and increased hospitalizations.Citation55 Chronic PA suppressive therapy is recognized as a desirable objective in the treatment of bronchiectasis. Attempts to eradicate the first appearance of PA in sputum may also be of benefit.Citation56,Citation57 Strategies to achieve these goals include inhaled antibiotics including tobramycin and colistimethate sodium. Inhaled tobramycin in bronchiectasis decreased the PA airway burden and, in some studies, reduced the number of exacerbations and hospital admissions (reviewed in Elborn et al.Citation32) The soon-to-be announced PROMIS trials (NCT03093974, NCT03460704) aim at assessing the use of a colistimethate sodium nebulization solution in non-CF bronchiectasis. Early positive clinical outcomes have been reported although definitive results of the 2 trials are yet to be published.Citation58 Preclinical studies of a phage anti-PA cocktail have led to the initiation of a clinical trial of this novel approach to suppress airway PA.Citation59

Macrolide therapy, particularly oral azithromycin, has been extensively studied in non-CF bronchiectasis as is discussed in a recent Cochrane review.Citation60 The review suggests that chronic azithromycin use in non-CF bronchiectasis is generally safe, reduces the number of respiratory exacerbations and improves quality of life while having limited or no effect on the number of hospitalizations. Several mechanisms by which chronic macrolide therapy may provide benefit have been proposed, including a decrease in the density of sensitive bacteria, a decrease in biofilm formation, and anti-inflammatory effects related to NETosis, but a single clear mechanism has not been identified. Potential adverse effects of macrolide therapy include cardiac (prolonged QT interval, arrythmias), otic (deafness and tinnitus) and microbiological (increased in pathogen, particularly NTM, antibiotic resistance) events.

Prevention of respiratory viral infections

During the COVID-19 pandemic one of the striking observations was that individuals with non-CF and CF bronchiectasis had a marked reduction in the number of respiratory exacerbations when compared to the period prior to the pandemic.Citation61 Social distancing, immunization, wearing masks and rapid detection of respiratory viral infections likely contributed to the reduction in exacerbations. These observations should encourage us to explore and implement prevention of respiratory viral infections as a priority in the care of people with bronchiectasis. Host airway protease inhibition in SARS-CoV-2 and possibly influenza have shown promise in pre-clinical work.Citation62

Perspectives

In the past 3 years we have witnessed evidence that prevention of respiratory virus transmission is associated with a reduced number of infectious exacerbations in individuals with bronchiectasis. Further benefits likely include a decrease in post-viral airway damage and worsening of bronchiectasis. These observations reinforce the importance of an integrated approach to the prevention of respiratory viral transmission in our society. The recently observed dramatic improvements in lung function and quality of life in CF has also sharpened our focus on the importance of effective mucociliary clearance.

Clinical trials of CFTR modulator therapy, novel mucolytic drugs and airway clearance techniques represent important ongoing areas of investigation in non-CF bronchiectasis. New approaches to improve host immune deficiencies and decrease bacterial load are ongoing and will likely be added to the bronchiectasis treatment toolkit. The targeting of neutrophil-derived proteases (eg, by blocking DPP-1)Citation49 has the potential to decrease the loss of bronchial wall elasticity and the excessive production of mucus, 2 key goals in the effective management of bronchiectasis.

Finally, we live in an era in which gene editing and correction are rapidly evolving. Gene targeting may represent a promising area of future therapeutic translation for rare individuals with a single genetic defect causing bronchiectasis. A preclinical study revealed that correction of the dynein heavy chain 11 (DNAH11) gene restores ciliary function in vitro.Citation63 Translation to clinical trials remains elusive but may one day become a feasible option for specific individuals.

Conclusion

In summary key observations in CF and non-CF bronchiectasis have markedly improved our understanding of potential therapeutic targets. The challenge now is to conduct robust clinical trials that will convert this knowledge into effective strategies that rapidly improve the quality of life for all patients living with bronchiectasis.

Author contributions

A.M. Cantin conceived, wrote and edited the manuscript.

Acknowledgments

The author thanks Simon Couillard for editorial assistance. A.M. Cantin is a member of the Centre de Recherche Clinique Etienne-LeBel of the CHUS.

Disclosure statement

A.M. Cantin has received honoraria from Vertex Pharmaceuticals (Canada) Incorporated and from Boehringer-Ingelheim for research and presentations outside of the submitted work.

Additional information

Funding

The author reported there is no funding associated with the work featured in this article.

References

  • Cole PJ. Inflammation: a two-edged Sword–the model of bronchiectasis. Eur J Respir Dis Suppl. 1986;147:6–15.
  • O’Donnell AE. Bronchiectasis – a Clinical Review. N Engl J Med. 2022;387:533–545.
  • Chandrasekaran R, Mac Aogain M, Chalmers JD, Elborn SJ, Chotirmall SH. Geographic variation in the aetiology, epidemiology and microbiology of bronchiectasis. BMC Pulm Med. 2018;18(1):83. doi:10.1186/s12890-018-0638-0.
  • Flume PA, Chalmers JD, Olivier KN. Advances in bronchiectasis: endotyping, genetics, microbiome, and disease heterogeneity. Lancet. 2018;392(10150):880–890. doi:10.1016/S0140-6736(18)31767-7.
  • Juliusson G, Gudmundsson G. Diagnostic imaging in adult non-cystic fibrosis bronchiectasis. Breathe. 2019;15(3):190–197. doi:10.1183/20734735.0009-2019.
  • Hata A, Hino T, Putman RK, et al. Traction Bronchiectasis/Bronchiolectasis on CT scans in relationship to clinical outcomes and mortality: the COPDGene study. Radiology. 2022;304(3):694–701. doi:10.1148/radiol.212584.
  • Chalmers JD, Polverino E, Crichton ML, et al. Bronchiectasis in Europe: data on disease characteristics from the European Bronchiectasis registry (EMBARC). Lancet Respir Med. 2023;11(7):637–649. doi:10.1016/S2213-2600(23)00093-0.
  • Turino GM, Lourenco RV, McCracken GH. Role of connective tissues in large pulmonary airways. J Appl Physiol. 1968;25(6):645–653. doi:10.1152/jappl.1968.25.6.645.
  • Fahy JV, Schuster A, Ueki I, Boushey HA, Nadel JA. Mucus hypersecretion in bronchiectasis. The role of neutrophil proteases. Am Rev Respir Dis. 1992;146(6):1430–1433. doi:10.1164/ajrccm/146.6.1430.
  • Belaaouaj A, Kim KS, Shapiro SD. Degradation of outer membrane protein A in Escherichia coli killing by neutrophil elastase. Science. 2000;289(5482):1185–1188. doi:10.1126/science.289.5482.1185.
  • Belaaouaj A, McCarthy R, Baumann M, et al. Mice lacking neutrophil elastase reveal impaired host defense against gram negative bacterial sepsis. Nat Med. 1998;4(5):615–618. doi:10.1038/nm0598-615.
  • Shoemark A, Cant E, Carreto L, et al. A point-of-care neutrophil elastase activity assay identifies bronchiectasis severity, airway infection and risk of exacerbation. Eur Respir J. 2019;53(6):1900303. doi:10.1183/13993003.00303-2019.
  • Sly PD, Gangell CL, Chen L, et al. Risk factors for bronchiectasis in children with cystic fibrosis. N Engl J Med. 2013;368(21):1963–1970. doi:10.1056/NEJMoa1301725.
  • Voynow JA, Shinbashi M. Neutrophil Elastase and chronic lung disease. Biomolecules. 2021;11(8):1065. doi:10.3390/biom11081065.
  • Mariano CA, Sattari S, Ramirez GO, Eskandari M. Effects of tissue degradation by collagenase and elastase on the biaxial mechanics of porcine airways. Respir Res. 2023;24(1):105. doi:10.1186/s12931-023-02376-8.
  • Tosi MF, Zakem H, Berger M. Neutrophil elastase cleaves C3bi on opsonized pseudomonas as well as CR1 on neutrophils to create a functionally important opsonin receptor mismatch. J Clin Invest. 1990;86(1):300–308. doi:10.1172/JCI114699.
  • Berger M, Sorensen RU, Tosi MF, Dearborn DG, Doring G. Complement receptor expression on neutrophils at an inflammatory site, the Pseudomonas-infected lung in cystic fibrosis. J Clin Invest. 1989;84(4):1302–1313. doi:10.1172/JCI114298.
  • McElvaney NG, Hubbard RC, Birrer P, et al. Aerosol alpha 1-antitrypsin treatment for cystic fibrosis. Lancet. 1991;337(8738):392–394. doi:10.1016/0140-6736(91)91167-s.
  • Voynow JA, Young LR, Wang Y, Horger T, Rose MC, Fischer BM. Neutrophil elastase increases MUC5AC mRNA and protein expression in respiratory epithelial cells. Am J Physiol. 1999;276(5):L835–43. doi:10.1152/ajplung.1999.276.5.L835.
  • Park JA, He F, Martin LD, Li Y, Chorley BN, Adler KB. Human neutrophil elastase induces hypersecretion of mucin from well-differentiated human bronchial epithelial cells in vitro via a protein kinase C{delta}-mediated mechanism. Am J Pathol. 2005;167(3):651–661. doi:10.1016/s0002-9440(10)62040-8.
  • Nakamura H, Yoshimura K, McElvaney NG, Crystal RG. Neutrophil elastase in respiratory epithelial lining fluid of individuals with cystic fibrosis induces interleukin-8 gene expression in a human bronchial epithelial cell line. J Clin Invest. 1992;89(5):1478–1484. doi:10.1172/JCI115738.
  • Vandivier RW, Fadok VA, Hoffmann PR, et al. Elastase-mediated phosphatidylserine receptor cleavage impairs apoptotic cell clearance in cystic fibrosis and bronchiectasis. J Clin Invest. 2002;109(5):661–670. doi:10.1172/JCI0213572.
  • Weldon S, McNally P, McElvaney NG, et al. Decreased levels of secretory leucoprotease inhibitor in the Pseudomonas-infected cystic fibrosis lung are due to neutrophil elastase degradation. J Immunol. 2009;183(12):8148–8156. doi:10.4049/jimmunol.0901716.
  • Keating D, Marigowda G, Burr L, et al. VX-445-tezacaftor-ivacaftor in patients with cystic fibrosis and one or two Phe508del alleles. N Engl J Med. 2018;379(17):1612–1620. doi:10.1056/NEJMoa1807120.
  • Middleton PG, Mall MA, Dřevínek P, et al. Elexacaftor-tezacaftor-ivacaftor for cystic fibrosis with a single Phe508del Allele. N Engl J Med. 2019;381(19):1809–1819. doi:10.1056/NEJMoa1908639.
  • Cantin AM, Hanrahan JW, Bilodeau G, et al. Cystic fibrosis transmembrane conductance regulator function is suppressed in cigarette smokers. Am J Respir Crit Care Med. 2006;173(10):1139–1144. doi:10.1164/rccm.200508-1330OC.
  • Dransfield MT, Wilhelm AM, Flanagan B, et al. Acquired cystic fibrosis transmembrane conductance regulator dysfunction in the lower airways in COPD. Chest. 2013;144(2):498–506. doi:10.1378/chest.13-0274.
  • Le Gars M, Descamps D, Roussel D, et al. Neutrophil elastase degrades cystic fibrosis transmembrane conductance regulator via calpains and disables channel function in vitro and in vivo. Am J Respir Crit Care Med. 2013;187(2):170–179. doi:10.1164/rccm.201205-0875OC.
  • Trinh NT, Bilodeau C, Maille E, et al. Deleterious impact of Pseudomonas aeruginosa on cystic fibrosis transmembrane conductance regulator function and rescue in airway epithelial cells. Eur Respir J. 2015;45(6):1590–1602. doi:10.1183/09031936.00076214.
  • O’Donnell AE, Barker AF, Ilowite JS, Fick RB. Treatment of idiopathic bronchiectasis with aerosolized recombinant human DNase I. rhDNase Study Group. Chest. 1998;113(5):1329–1334. doi:10.1378/chest.113.5.1329.
  • Ramsey BW, Pepe MS, Quan JM, et al. Intermittent administration of inhaled tobramycin in patients with cystic fibrosis. Cystic Fibrosis Inhaled Tobramycin Study Group. N Engl J Med. 1999;340(1):23–30. doi:10.1056/NEJM199901073400104.
  • Elborn JS, Blasi F, Haworth CS, et al. Bronchiectasis and inhaled tobramycin: a literature review. Respir Med. 2022;192:106728. doi:10.1016/j.rmed.2021.106728.
  • Munoz G, de Gracia J, Buxo M, Alvarez A, Vendrell M. Long-term benefits of airway clearance in bronchiectasis: a randomised placebo-controlled trial. Eur Respir J. 2018;51(1):1701926. doi:10.1183/13993003.01926-2017.
  • Roy MG, Livraghi-Butrico A, Fletcher AA, et al. Muc5b is required for airway defence. Nature. 2014;505(7483):412–416. doi:10.1038/nature12807.
  • Cross CE, Halliwell B, Allen A. Antioxidant protection: a function of tracheobronchial and gastrointestinal mucus. Lancet. 1984;1(8390):1328–1330. doi:10.1016/s0140-6736(84)91822-1.
  • Nadziejko C, Finkelstein I. Inhibition of neutrophil elastase by mucus glycoprotein. Am J Respir Cell Mol Biol. 1994;11(1):103–107. doi:10.1165/ajrcmb.11.1.7912511.
  • Cantin AM, Ouellet C, Cloutier A, McDonald PP. Airway mucins inhibit oxidative and non-oxidative bacterial killing by human neutrophils. Front Pharmacol. 2020;11:554353. doi:10.3389/fphar.2020.554353.
  • Batson BD, Zorn BT, Radicioni G, et al. Cystic fibrosis airway mucus hyperconcentration produces a vicious cycle of Mucin, pathogen, and inflammatory interactions that promotes disease persistence. Am J Respir Cell Mol Biol. 2022;67(2):253–265. doi:10.1165/rcmb.2021-0359OC.
  • Kesimer M, Ford AA, Ceppe A, et al. Airway Mucin concentration as a marker of chronic bronchitis. N Engl J Med. 2017;377(10):911–922. doi:10.1056/NEJMoa1701632.
  • Hill DB, Button B, Rubinstein M, Boucher RC. Physiology and pathophysiology of human airway mucus. Physiol Rev. 2022;102(4):1757–1836. doi:10.1152/physrev.00004.2021.
  • Ramsey KA, Chen ACH, Radicioni G, et al. Airway mucus hyperconcentration in non-cystic fibrosis bronchiectasis. Am J Respir Crit Care Med. 2020;201(6):661–670. doi:10.1164/rccm.201906-1219OC.
  • Volpato M, Vialaret J, Hirtz C, et al. Rheology predicts sputum eosinophilia in patients with muco-obstructive lung diseases. Biochem Biophys Res Commun. 2022;622:64–71. doi:10.1016/j.bbrc.2022.07.025.
  • Mall MA, Danahay H, Boucher RC. Emerging concepts and therapies for mucoobstructive lung disease. Ann Am Thorac Soc. 2018;15(Suppl 3):S216–S226. doi:10.1513/AnnalsATS.201806-368AW.
  • Ehre C, Rushton ZL, Wang B, et al. An improved inhaled mucolytic to treat airway muco-obstructive diseases. Am J Respir Crit Care Med. 2019;199(2):171–180. doi:10.1164/rccm.201802-0245OC.
  • Gupta S, Pattanaik D, Krishnaswamy G. Common variable immune deficiency and associated complications. Chest. 2019;156(3):579–593. doi:10.1016/j.chest.2019.05.009.
  • Vonarburg C, Loetscher M, Spycher MO, et al. Topical application of nebulized human IgG, IgA and IgAM in the lungs of rats and non-human primates. Respir Res. 2019;20(1):99. doi:10.1186/s12931-019-1057-3.
  • Adkison AM, Raptis SZ, Kelley DG, Pham CT. Dipeptidyl peptidase I activates neutrophil-derived serine proteases and regulates the development of acute experimental arthritis. J Clin Invest. 2002;109(3):363–371. doi:10.1172/JCI0213462.
  • Palmer R, Maenpaa J, Jauhiainen A, et al. Dipeptidyl peptidase 1 inhibitor AZD7986 induces a sustained, exposure-dependent reduction in neutrophil elastase activity in healthy subjects. Clin Pharmacol Ther. 2018;104(6):1155–1164. doi:10.1002/cpt.1053.
  • Chalmers JD, Haworth CS, Metersky ML, et al. Phase 2 trial of the DPP-1 inhibitor brensocatib in bronchiectasis. N Engl J Med. 2020;383(22):2127–2137. doi:10.1056/NEJMoa2021713.
  • Liu W, Yan M, Liu Y, McLeish KR, Coleman WG, Jr., Rodgers GP. Olfactomedin 4 inhibits cathepsin C-mediated protease activities, thereby modulating neutrophil killing of Staphylococcus aureus and Escherichia coli in mice. J Immunol. 2012;189(5):2460–2467. doi:10.4049/jimmunol.1103179.
  • Kangelaris KN, Prakash A, Liu KD, et al. Increased expression of neutrophil-related genes in patients with early sepsis-induced ARDS. Am J Physiol Lung Cell Mol Physiol. 2015;308(11):L1102–13. doi:10.1152/ajplung.00380.2014.
  • Kangelaris KN, Clemens R, Fang X, et al. A neutrophil subset defined by intracellular olfactomedin 4 is associated with mortality in sepsis. Am J Physiol Lung Cell Mol Physiol. 2021;320(5):L892–L902. doi:10.1152/ajplung.00090.2020.
  • Keir HR, Shoemark A, Dicker AJ, et al. Neutrophil extracellular traps, disease severity, and antibiotic response in bronchiectasis: an international, observational, multicohort study. Lancet Respir Med. 2021;9(8):873–884. doi:10.1016/S2213-2600(20)30504-X.
  • Oriano M, Amati F, Gramegna A, et al. Protease-antiprotease imbalance in bronchiectasis. Int J Mol Sci. 2021;22:5996.
  • Finch S, McDonnell MJ, Abo-Leyah H, Aliberti S, Chalmers JD. A comprehensive analysis of the impact of Pseudomonas aeruginosa colonization on prognosis in adult bronchiectasis. Annals of the American Thoracic Society. 2015;12:1602–1611.
  • Polverino E, Goeminne PC, McDonnell MJ, et al. European Respiratory Society guidelines for the management of adult bronchiectasis. Eur Respir J. 2017;50(3):1700629. doi:10.1183/13993003.00629-2017.
  • Pieters A, Bakker M, Hoek RAS, et al. The clinical impact of Pseudomonas aeruginosa eradication in bronchiectasis in a Dutch referral centre. Eur Respir J. 2019;53(4):1802081. doi:10.1183/13993003.02081-2018.
  • de la Rosa-Carrillo D, Suarez-Cuartin G, Golpe R, Maiz Carro L, Martinez-Garcia MA. Inhaled colistimethate sodium in the management of patients with bronchiectasis infected by Pseudomonas aeruginosa: A narrative review of current evidence. Infect Drug Resist. 2022;15:7271–7292. doi:10.2147/IDR.S318173.
  • Tamma PD, Souli M, Billard M, et al. Safety and microbiological activity of phage therapy in persons with cystic fibrosis colonized with Pseudomonas aeruginosa: study protocol for a phase 1b/2, multicenter, randomized, double-blind, placebo-controlled trial. Trials. 2022;23(1):1057. doi:10.1186/s13063-022-07047-5.
  • Kelly C, Chalmers JD, Crossingham I, et al. Macrolide antibiotics for bronchiectasis. Cochrane Database Syst Rev. 2018;3:CD012406.
  • Crichton ML, Shoemark A, Chalmers JD. The impact of the COVID-19 pandemic on exacerbations and symptoms in bronchiectasis: a prospective study. Am J Respir Crit Care Med. 2021;204(7):857–859. doi:10.1164/rccm.202105-1137LE.
  • Shapira T, Monreal IA, Dion SP, et al. A TMPRSS2 inhibitor acts as a pan-SARS-CoV-2 prophylactic and therapeutic. Nature. 2022;605(7909):340–348. doi:10.1038/s41586-022-04661-w.
  • Lai M, Pifferi M, Bush A, et al. Gene editing of DNAH11 restores normal cilia motility in primary ciliary dyskinesia. J Med Genet. 2016;53(4):242–249. doi:10.1136/jmedgenet-2015-103539.