Skip to main content
Chest logoLink to Chest
. 2015 Mar 12;148(2):533–542. doi: 10.1378/chest.14-1997

The Evolution of Cystic Fibrosis Care

Jessica E Pittman 1,, Thomas W Ferkol 1
PMCID: PMC4524331  PMID: 25764168

Abstract

Cystic fibrosis (CF) is the most common life-limiting inherited illness of whites. Most of the morbidity and mortality in CF stems from impaired mucociliary clearance leading to chronic, progressive airways obstruction and damage. Significant progress has been made in the care of patients with CF, with advances focused on improving mucociliary clearance, minimizing inflammatory damage, and managing infections; these advances include new antimicrobial therapies, mucolytic and osmotic agents, and antiinflammatory treatments. More recently, researchers have targeted disease-causing mutations using therapies to promote gene transcription and improve channel function, which has led to impressive physiologic changes in some patients. As we develop more advanced, allele-directed therapies for the management of CF, it will become increasingly important to understand the specific genetic and environmental interactions that cause the significant heterogeneity of lung disease seen in the CF population. This understanding of CF endotypes will allow for more targeted, personalized therapies for future patients. This article reviews the genetic and molecular basis of CF lung disease, the treatments currently available, and novel therapies that are in development.


Cystic fibrosis (CF), which is an autosomal recessive defect occurring in approximately one in 3,500 live births based on data from neonatal screening,1 is the most common, life-shortening inherited disease of whites. The life expectancy of a child born with CF has improved steadily, largely because of advances in disease surveillance and more aggressive treatment strategies. Nevertheless, patients with CF die too young, with much of the early morbidity and mortality from CF resulting from progressive airway involvement. Universal adoption of newborn screening in the United States1 has led to earlier diagnosis and treatment of CF, which has sparked hope that lung disease can be averted even before it begins, especially with the advent of newer agents that target the basic cellular defect and have the potential to radically change clinical outcomes. The use of defined endotypes, phenotypic subtypes that rely on a combination of genetics, biomarkers, environmental exposures, clinical outcome measures, and infectious and inflammatory factors to characterize disease, may better direct therapeutic interventions by targeting specific populations with CF. In this article, we review the pathophysiology of CF lung disease and describe how current and emerging therapies can both treat and possibly prevent this progressive lung disease.

Genetics and Pathophysiology of CF Airway Disease

To fully understand the current and newer therapies for CF, physicians must have a working knowledge of the basic pathophysiology of the disease. CF is clinically characterized by chronic sinopulmonary and GI manifestations, which are caused by abnormalities in the cystic fibrosis transmembrane conductance regulator (CFTR), a channel located at the surface of the cells lining the airway epithelium and in the submucosal glands that mediates cyclic adenosine monophosphate (cAMP)-regulated transport of chloride and other anions.26 The CFTR is functionally linked to the epithelial sodium channel (ENaC) and alternative apical chloride channels; thus, the CFTR defects lead to not only reduced chloride conductance but also dysregulation of ENaC activity. This failure of chloride secretion and sodium hyperabsorption leads to desiccation of the periciliary fluid layer and viscous mucus on the airway surface. The dehydrated secretions and excess solids in airway mucus impair mucociliary clearance, obstruct the airways, allow bacterial infection to become established, and subsequently incite an intense inflammatory response.7 Impaired activity of bactericidal proteins produced by airway epithelia related to altered bicarbonate secretion in the CF airway creates gaps in innate airway defenses and contributes to chronic infection (Fig 1).8

Figure 1 –

Figure 1 –

Epithelial pathophysiology in cystic fibrosis, characterized by altered anion secretion, sodium hyperabsorption, and dehydration of the apical surface fluid that leads to reduced periciliary fluid volume and pH, which interferes with mucociliary clearance and innate defenses, resulting in chronic infection. The decreased periciliary fluid volume also concentrates inflammatory mediators at the immediate epithelial surface. CFTR = cystic fibrosis transmembrane conductance regulator; Cl = chloride; ClCa = alternative chloride channel; ENaC = epithelial sodium channel; HCO3 = bicarbonate; H2O = water; Na+ = sodium.

Nearly 2,000 disease-causing mutations in CFTR, a 27-exon gene on chromosome 7,911 have been identified. Although racial and ethnic differences exist, the most common mutation is c.1521_1523delCTT, or delF508, which accounts for > 70% of mutant alleles in the CF population.11,12 Mutations are usually grouped into six classes based on the protein product. Class 1 mutations consist of frameshift or nonsense variants that cause altered or impaired CFTR transcription, producing either messenger RNA that decays before nuclear export or truncated, nonfunctional protein. Class 2 mutations, such as delF508, result in misfolded protein that is trafficked to degradation pathways, has abnormal function, and is more rapidly cleared from the cell membrane. Mutations from both of these classes typically lead to more severe lung disease and pancreatic insufficiency. Class 3 mutations, including G551D, commonly called gating mutations, lead to a CFTR that is poorly responsive or nonresponsive to ATP activation of the nucleotide-binding domains, resulting in defective chloride conductance across the apical cell membrane. The clinical phenotypes and severity associated with class 3 mutations vary. Class 4, or conducting, mutations have impaired chloride conductance or transport caused by alterations in size and ion selectivity of the channel pore. Class 5 mutations are frequently splice mutants that reduce the rate of synthesis of functional CFTR and include intron 8 polythymidine variants. Class 6 mutations involve genetic defects that are near the N- or C-terminus and lead to aberrant membrane insertion, stability, or trafficking (Fig 2).2,6,1318 Some mutations defy classification. For instance, delF508 acts as a class 2 (altered processing), class 3 (impaired gating), and class 6 (rapidly degraded) mutation, which potentially has implications for molecular therapy. Other mutations are yet to be classified, either because of rarity of incidence or unclear effect on CFTR function.

Figure 2 –

Figure 2 –

Classes of CFTR mutations. ER = endoplasmic reticulum; mRNA = messenger RNA. See Figure 1 legend for expansion of other abbreviations.

Pulmonary disease is the primary cause of mortality in patients with CF,19,20 with considerable variability in presentation, severity, and progression of pulmonary involvement, even among patients with the same CFTR mutation.11 Patient-level differences have been attributed to a variety of factors including genetic modifiers, sex, race, ethnicity, socioeconomic status, nutritional status, and exposure to airborne pollutants, including tobacco smoke.2125 There is mounting evidence suggesting that pulmonary disease begins early in infancy. The lungs of neonates with CF have long been considered histologically normal, but animal models now indicate that structural abnormalities in the CF lung may occur in utero.26,27 Infants with CF are typically born at lower birth weight than their non-CF counterparts, and poor nutrition has an impact throughout life, particularly in early childhood when somatic growth is linked to lung and alveolar growth.28,29 Although it is unusual for neonates to have respiratory symptoms, older infants can develop tachypnea, wheezing, and cough that is frequently associated with respiratory viral infections. Studies of infants and preschool children with CF have found abnormal pulmonary function, structural changes, and increased inflammatory markers in BAL fluid occurring as early as 6 months of age.3032 Moreover, there is significant disease heterogeneity within the lungs of a single patient, particularly in the early stages of disease.

The CF lungs are not infected at birth, but over time, various bacteria are intermittently found in airway secretions.31 As bronchial obstruction evolves, the airway becomes persistently infected.33 Acquisition of Burkholderia cepacia, Pseudomonas aeruginosa, methicillin-resistant Staphylococcus aureus, Stenotrophomonas maltophilia, and Mycobacterium abcessus has been associated with deteriorating lung function and poorer prognosis.3340 Newly emerging data on the CF respiratory and gut microbiome suggest a complex interplay of microbes that contribute to, and are altered by, progressive CF lung disease.41,42

The inflammatory response in CF airways is both protective and destructive, driven by local stimuli, mediators, and chemoattractants. The intense neutrophilic inflammation is remarkably compartmentalized, with disease limited to the airway.4345 Several lines of evidence suggest that airway inflammation in CF may be exaggerated, with higher epithelial production of proinflammatory cytokines, but not all CF cell models and clinical studies have revealed a dysregulated response.43 Indeed, several non-CFTR genetic modifiers, some of which are associated with inflammation, have been identified that can influence the severity of lung disease.11,4648 Inflammatory mediators, such as neutrophil elastase, have been used as biomarkers for CF lung disease.49 Neutrophil-derived proteases are released during phagocytosis and cell death and are central to the pathogenesis of CF lung disease by interference with nonspecific airway defenses, stimulation of epithelial chemokine secretion, and degradation of structural proteins, all of which lead to progressive bronchiectasis and bronchomalacia.50

Assessment of CF Lung Disease

CF care relies on a multidisciplinary, center-based team approach that involves physicians, nurses, nutritionists, respiratory therapists, and other ancillary staff. Comprehensive care has contributed to improvements in morbidity and mortality in the CF population.51 Despite recommendations for standardized care based on randomized controlled trials and the availability of clinical guidelines from the Cystic Fibrosis Foundation that outline “best” practices for diagnosis, routine monitoring, and interventions to slow progression of lung disease,1,5254 debate continues regarding optimal care, with the goal being early detection and aggressive treatment of disease. Routine surveillance studies are recommended, including regular spirometry to monitor the lung function of children ≥ 6 years of age, annual chest radiographs, and respiratory bacterial cultures to assess for pathogens in the lower respiratory tract. Studies have shown that respiratory culture results, particularly oropharyngeal swab and BAL fluid, may differ by source.55,56 Even cultures of lavage fluid from different lung segments can have different results.56 However, a 2011 randomized controlled study showed no apparent benefit in lavage-guided management of infection over standard oropharyngeal cultures in young children with CF.57

Pulmonary function testing is an important tool in the clinical evaluation of the patient with CF, and FEV1 has been linked directly to morbidity and mortality.58 However, as the clinical outcomes in CF have improved, the rate of pulmonary function decline has slowed, thus making spirometry a less sensitive tool for recognizing and monitoring disease in patients with mild disease,59 which has implications for both clinical care and trials. Children have considerable lung disease well before they are symptomatic or demonstrate either abnormal or deteriorating lung function.31,60 A growing number of centers reliably perform spirometry in preschool children. Infant pulmonary function testing using the raised volume rapid thoracoabdominal compression technique and plethysmography are considered useful adjuncts to standard care in centers with the necessary staffing, equipment, and expertise.61,62 Multiple breath washout testing, which assesses ventilation inhomogeneity, has been shown to be more sensitive than spirometry for the identification of early or mild CF lung disease.63,64 Despite a lack of normative data, some centers are beginning to use this approach clinically.

Sensitivity of the chest radiograph, particularly for early CF lung disease or minor changes, is relatively low. Findings can be inconsistent and evaluator dependent, even when using scoring systems.65 Although CT scanning has been shown to be a more sensitive test for the identification of structural abnormalities, including air trapping and bronchiectasis, concern about repeated radiation exposure has limited its routine use. MRI of the lung has shown promise for the evaluation of both structural and ventilation abnormalities without radiation risk, although further development of testing protocols is still needed.66

Current and Future Management of CF Lung Disease

Antimicrobial Therapies

Antibiotics are mainstays for the treatment of acute exacerbations and maintenance therapy for patients with chronic infection. At times, patients with CF will develop increasing respiratory signs and symptoms related to increasing bacterial burden, airway inflammation, and endobronchial obstruction, a phenomenon termed a pulmonary exacerbation. Although a precise definition of pulmonary exacerbation remains elusive,67 it is clinically manifested by a worsening cough, dyspnea, and sputum production, as well as systemic symptoms such as fatigue, anorexia, and weight loss.67,68 Frequent pulmonary exacerbations negatively affect prognosis.69 Pulmonary function measures typically fall during exacerbations, and the treatment goal is a return to best baseline measures, irrespective of the patient’s symptoms. However, many patients fail to reach this goal.70 Although a few studies have attempted to define the optimal length of treatment,71,72 pulmonary exacerbations are generally treated with antibiotic therapy for 10 to 21 days, guided by bacterial isolates from respiratory secretions and aggressive airway clearance techniques.73,74

The eradication of P aeruginosa from the lower respiratory tract has become a cornerstone of CF treatment in the United States during the past 2 decades. Several different eradication strategies, which typically consist of single agents or combinations of systemic and inhaled antibiotics, have been tested.75 Although strategies to clear initial P aeruginosa infection have been shown to delay chronic colonization and slow lung function decline, Pseudomonas eventually becomes established in the airways in most older children with CF.76 Chronic suppressive therapy with inhaled antibiotics such as tobramycin and aztreonam has been shown to decrease the frequency of pulmonary exacerbations; the impact of these therapies on mortality remains unknown.77,78 Other antibiotics currently being developed for inhaled administration include vancomycin, ciprofloxacin, and levofloxacin.79,80

Mucolytic and Osmotic Agents

Because the primary physiologic manifestation of CF in the lungs is defective mucociliary clearance, it is only logical that several therapies have been developed to target this defect. To maintain lung health, airway secretions need to be mobilized to not only relieve airway obstruction but also reduce infection and airway inflammation. Effective airway clearance techniques are essential components of CF therapy, although few data have clearly demonstrated the long-term effect of airway clearance on lung function and virtually no studies have compared the efficacy of different methods.81,82 Aerosolized mucolytic agents have been incorporated into CF care to clear airway secretions. Dornase alfa (recombinant human deoxyribonuclease I) cleaves the DNA released in high concentrations by degraded neutrophils present in CF mucus, thus reducing sputum viscosity and leading to slower lung function decline and fewer pulmonary exacerbations.68,83,84 N-acetylcysteine, which acts through hydrolysis of disulfide bonds in mucin, has a long history of use as a mucolytic agent for patients with CF, although few studies support its effectiveness.85

Other inhaled therapies have targeted fluid and ion balance in the airways as a means of improving mucociliary clearance. Inhaled hypertonic saline, which leads to a temporary increase in mucociliary clearance by increasing the depth of the periciliary fluid space and lowering mucus osmolality, has been widely adopted in children and adults with CF.86,87 Its efficacy in younger subjects has yet to be established.88 More recently, inhaled mannitol has been proposed as an osmotic agent designed to rehydrate the airway surface layer, and several studies suggest inhalation of mannitol may improve FEV1.89,90 However, there are concerns about possible adverse effects. While this therapy is available for adults in Europe, Australia, and New Zealand, it has not been approved by the US Food and Drug Administration.

Another therapeutic strategy to increase periciliary fluid depth is to alter the function of other non-CFTR ion channels at the apical surface. A uridine triphosphate nucleotide analog (denufosol) activates non-CFTR chloride channels and inhibits sodium absorption by stimulating P2Y2 receptors on epithelial cells,91 but a large phase 3, placebo-controlled study in patients with CF failed to show an effect.92,93 Several studies have examined agents that block ENaC, but have shown little benefit to date. Studies examining newer ENaC inhibitors are in the early stages.94

Antiinflammatory Therapies

Chronic neutrophilic inflammation is central to the pathogenesis of CF lung disease. Neutrophilic inflammation is present from early infancy,95 even before the child is symptomatic. Inflammation in the CF lung is highly compartmentalized and confined to the airway lumen, whereas the alveolar space is relatively spared. Various antiinflammatory agents have been applied to CF lung disease.96,97 High-dose ibuprofen has been shown to significantly decrease the rate of lung function decline in younger children with CF,98 but its use is somewhat limited because of compliance and potential renal and gastric toxicities. Alternate-day treatment regimens with systemic corticosteroids have also been shown to significantly slow lung function decline, although there is even greater concern for adverse effects.99 Inhaled corticosteroids have shown some benefit in terms of preservation of lung function, but long-term use has been associated with increased risk of glucose intolerance and reduced growth velocity.100

Azithromycin has gained acceptance for its antiinflammatory effects, although the precise mechanism of action in CF is unclear. In a large multicenter study in the United States, patients with CF who were > 6 years of age and infected with P aeruginosa who were treated with azithromycin three times weekly had better lung function and fewer pulmonary exacerbations when compared with those who were given a placebo. A subsequent clinical trial in subjects who were not chronically infected with P aeruginosa did not reveal a difference in lung function, though there was a benefit in terms of a reduction in pulmonary exacerbations and improved nutritional status.101,102 Few adverse effects have been reported, which may explain some of the popularity of this agent. There are contraindications to its use, specifically nontuberculous mycobacterial infections, where use of azithromycin has the potential to induce antibiotic resistance, and prolonged QT interval. Other oral antiinflammatory agents currently under study include N-acetylcysteine, docosahexaenoic acid, and sildenafil.103 Their effects on lung function and airway inflammation have yet to be established.

CFTR Genotype-Specific Therapies

Historically, treatment of CF has focused on the downstream effects of CFTR dysfunction—impaired mucociliary clearance, chronic infection, and chronic inflammation.45 The ideal therapy for CF lung disease would directly treat the disease proximate to the gene or protein defect, allowing for normal or near-normal CFTR function. Despite its early promise, gene therapy has yet to be successful in patients.104 One of the most exciting advances in CF therapeutics is the discovery of small molecules that alter mutant CFTR function.105

Through the Cystic Fibrosis Foundation Therapeutic Development Network, a nonprofit clinical trials network established to facilitate the development and conduct of all phases of clinical trials,106 several novel therapies are being tested or introduced to CF care (Fig 3). Some agents have allele- or mutation-specific effects.

Figure 3 –

Figure 3 –

Recently approved and newer agents that are being tested for cystic fibrosis through the Therapeutics Development Network. FDA = US Food and Drug Administration. See Figure 1 legend for expansion of other abbreviation. (Modified with permission from the Cystic Fibrosis Foundation.106)

Class 1 mutations, which include nonsense mutations that result in premature termination of CFTR messenger RNA, account for roughly 10% of CFTR mutations. Several agents allow the ribosome to read through the premature stop codon but not through normal termination codons. Despite promising early trials, a large phase 3 clinical trial with ataluren (PTC124), an oral medication that promotes ribosomal read-through, showed no improvement in FEV1 compared with placebo, though the results may have been confounded by aminoglycoside use.107

In contrast, a Food and Drug Administration-approved oral medication, ivacaftor, acts as a CFTR potentiator, improving protein function in patients with class 3 mutations, and has been shown to dramatically improve lung function and quality of life and reduce sweat chloride levels and pulmonary exacerbations, in patients with the G551D mutation (Fig 4).108 Its indication was recently expanded to several other gating mutations,109 which unfortunately still represents only a minority of patients with CF.

Figure 4 –

Figure 4 –

Correcting the molecular defect in cystic fibrosis. Newer agents have mutation-specific effects on defective CFTR proteins. Class 3 or gating mutations, such as G551D, result in a defective CFTR that is unable to regulate chloride flow because of impaired ATP activation of the nucleotide-binding domains, which is corrected by the potentiator, ivacaftor. Class 2 mutations, such as delF508, result in misfolded protein that is trafficked to degradation pathways, but the corrector, lumicaftor, acts as a chaperone and traffics the defective protein to an apical surface. Nevertheless, the delF508 protein still has impaired channel function and will require addition of a potentiator to correct the chloride conductance. See Figure 1 and 2 legends for expansion of abbreviations.

Correction of the class 2 mutation delF508 is the holy grail of CF therapeutics, because it could potentially change the clinical course of 90% of all affected individuals in the United States. Numerous small molecule correctors are in the therapeutic pipeline,106 including lumicaftor, which has been shown to act as a chaperone for the defective CFTR protein and partially restored cyclic adenosine monophosphate-mediated chloride secretion in delF508 human bronchial epithelial cells in vitro. Early experience with lumicaftor alone showed a modest drug response only,110 which is not surprising because the rescued delF508 protein is still a defective channel (Fig 3). Recent clinical trials combining lumicaftor with ivacaftor to improve mutant channel gating reportedly have yielded promising results. However, studies using airway epithelial cell models have shown that ivacaftor interfered with the effect of lumicaftor on delF508 CFTR, indicating that this combination of agents may have limitations.111,112

Shift Toward Primary Prevention/Treatment of Infants and Young Children

Although advances over the past 20 years have led to tremendous progress in the treatment of CF lung disease, these therapies are currently aimed at treating existing lung disease and slowing disease progression, which is often described as secondary or tertiary disease prevention. It is clear that lung disease begins very early in life, and CF may provide a striking opportunity for the primary prevention-initiation of therapies before measurable disease is present.113 Universal implementation of prenatal and neonatal screening programs in the United States means that infants are usually diagnosed before manifesting respiratory symptoms. The emergence of novel genotype-, mutation-, or class-specific therapeutics that target the basic defects allows for personalized treatments. These discoveries have raised the hope that CF lung disease can be prevented before it starts, well before children develop respiratory symptoms.

For these primary prevention strategies to succeed, more sensitive outcome measures that can identify the earliest changes in lung disease are needed for use in infants and young children. Very few interventional studies have examined treatments in children < 6 years of age, largely because of the difficulty of measuring a treatment effect. Early physiologic and structural changes in the CF lung are relatively subtle, which makes demonstration of a significant treatment response challenging. In addition, there are few adequately sensitive, low-risk techniques to measure disease in this population. Further development of methods that can detect the earliest changes of CF lung disease while being minimally invasive, and ideally imparting no risk from radiation or sedation, is critical for the identification and longitudinal following of disease in young children.

As we move forward toward an era of personalized medicine, the primary prevention of CF lung disease will likely be accomplished by comprehensively defining endotypes of patients with CD, using genotype (including CFTR and modifier genes) and phenotype, combined with knowledge of environmental exposures and social factors, to stratify patients by risk and likely disease manifestations. Endotyping may also improve outcomes in therapeutic trials by better classifying those subjects most likely to benefit from interventions and adding the power to identify specific responder groups. This approach has the potential to improve clinical care by identifying those patients at highest risk, in whom a particular therapy (or combination of therapies) will likely have the greatest impact while minimizing risk.

Conclusions

Several novel therapies are being tested or have recently been introduced to CF care, with even more treatments in the pipeline. These new therapies have brought the exciting possibility of prevention of CF lung disease, an unprecedented opportunity to profoundly change the morbidities and mortality associated with CF, forever changing the patients’ lives. The goal of managing patients with CF without measureable lung disease is potentially within our grasp.

The adoption of many of these therapeutic approaches has and will continue to improve the health of patients with CF, but the growing number of treatments is beginning to present clinical dilemmas. What therapies should be given to which patients? When do we begin therapies in young children? How do we minimize risk as well as the physical, financial, emotional, and social burdens to the patients while providing the most aggressive care possible? As therapeutic advances continue, particularly with the move toward genotype-directed treatments, better classification of patients with CF from infancy onwards will play an important role in characterizing disease progression and response to therapies. Endotyping of patients will need to become the norm and will likely have a significant impact in both future research studies and clinical care.

Acknowledgments

Financial/nonfinancial disclosures: The authors have reported to CHEST that no potential conflicts of interest exist with any companies/organizations whose products or services may be discussed in this article.

Role of sponsors: The views expressed do not necessarily reflect the official policies of the Department of Health and Human Services, nor does mention of trade names, commercial practices, or organizations imply endorsement by the US Government.

ABBREVIATIONS

CF

cystic fibrosis

CFTR

cystic fibrosis transmembrane conductance regulator

ENaC

epithelial sodium channel

Footnotes

FUNDING/SUPPORT: The authors were supported by the Cystic Fibrosis Foundation Therapeutics (Dr Pittman), the Children’s Discovery Institute (Dr Ferkol), and the National Institutes of Health (NIH) [Grants HL101465 (Dr Ferkol), HL116211 (Drs Pittman and Ferkol), HL096458 (Dr Ferkol), and K12 HL120002 (Dr Pittman)].

Reproduction of this article is prohibited without written permission from the American College of Chest Physicians. See online for more details.

References

  • 1.Farrell PM, Rosenstein BJ, White TB, et al. ; Cystic Fibrosis Foundation. Guidelines for diagnosis of cystic fibrosis in newborns through older adults: Cystic Fibrosis Foundation consensus report. J Pediatr. 2008;153(2):S4-S14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Drumm ML, Ziady AG, Davis PB. Genetic variation and clinical heterogeneity in cystic fibrosis. Annu Rev Pathol. 2012;7:267-282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Quinton PM. Chloride impermeability in cystic fibrosis. Nature. 1983;301(5899):421-422. [DOI] [PubMed] [Google Scholar]
  • 4.Knowles M, Gatzy J, Boucher R. Relative ion permeability of normal and cystic fibrosis nasal epithelium. J Clin Invest. 1983;71(5):1410-1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wine JJ. The genesis of cystic fibrosis lung disease. J Clin Invest. 1999;103(3):309-312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Quinton PM. Cystic fibrosis: impaired bicarbonate secretion and mucoviscidosis. Lancet. 2008;372(9636):415-417. [DOI] [PubMed] [Google Scholar]
  • 7.Knowles MR, Boucher RC. Mucus clearance as a primary innate defense mechanism for mammalian airways. J Clin Invest. 2002;109(5):571-577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Pezzulo AA, Tang XX, Hoegger MJ, et al. Reduced airway surface pH impairs bacterial killing in the porcine cystic fibrosis lung. Nature. 2012;487(7405):109-113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Rommens JM, Iannuzzi MC, Kerem B, et al. Identification of the cystic fibrosis gene: chromosome walking and jumping. Science. 1989;245(4922):1059-1065. [DOI] [PubMed] [Google Scholar]
  • 10.Kerem B, Rommens JM, Buchanan JA, et al. Identification of the cystic fibrosis gene: genetic analysis. Science. 1989;245(4922):1073-1080. [DOI] [PubMed] [Google Scholar]
  • 11.Knowles MR, Drumm M. The influence of genetics on cystic fibrosis phenotypes. Cold Spring Harb Perspect Med. 2012;2(12):a009548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Mickle JE, Cutting GR. Genotype-phenotype relationships in cystic fibrosis. Med Clin North Am. 2000;84(3):597-607. [DOI] [PubMed] [Google Scholar]
  • 13.Welsh MJ, Smith AE. Molecular mechanisms of CFTR chloride channel dysfunction in cystic fibrosis. Cell. 1993;73(7):1251-1254. [DOI] [PubMed] [Google Scholar]
  • 14.Zielenski J, Tsui LC. Cystic fibrosis: genotypic and phenotypic variations. Annu Rev Genet. 1995;29:777-807. [DOI] [PubMed] [Google Scholar]
  • 15.Cheng SH, Gregory RJ, Marshall J, et al. Defective intracellular transport and processing of CFTR is the molecular basis of most cystic fibrosis. Cell. 1990;63(4):827-834. [DOI] [PubMed] [Google Scholar]
  • 16.Cutting GR, Kasch LM, Rosenstein BJ, et al. A cluster of cystic fibrosis mutations in the first nucleotide-binding fold of the cystic fibrosis conductance regulator protein. Nature. 1990;346(6282):366-369. [DOI] [PubMed] [Google Scholar]
  • 17.Drumm ML, Wilkinson DJ, Smit LS, et al. Chloride conductance expressed by delta F508 and other mutant CFTRs in Xenopus oocytes. Science. 1991;254(5039):1797-1799. [DOI] [PubMed] [Google Scholar]
  • 18.Kiesewetter S, Macek M, Jr, Davis C, et al. A mutation in CFTR produces different phenotypes depending on chromosomal background. Nat Genet. 1993;5(3):274-278. [DOI] [PubMed] [Google Scholar]
  • 19.Stern RC, Boat TF, Doershuk CF, Tucker AS, Primiano FP, Jr, Matthews LW. Course of cystic fibrosis in 95 patients. J Pediatr. 1976;89(3):406-411. [DOI] [PubMed] [Google Scholar]
  • 20.Morgan WJ, Wagener JS, Yegin A, Pasta DJ, Millar SJ, Konstan MW; Scientific Advisory Group, investigators; coordinators of the Epidemiologic Study of Cystic Fibrosis. Probability of treatment following acute decline in lung function in children with cystic fibrosis is related to baseline pulmonary function. J Pediatr. 2013;163(4):1152-1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wolfenden LL, Schechter MS. Genetic and non-genetic determinants of outcomes in cystic fibrosis. Paediatr Respir Rev. 2009;10(1):32-36. [DOI] [PubMed] [Google Scholar]
  • 22.Schechter MS. Non-genetic influences on cystic fibrosis lung disease: the role of sociodemographic characteristics, environmental exposures, and healthcare interventions. Semin Respir Crit Care Med. 2003;24(6):639-652. [DOI] [PubMed] [Google Scholar]
  • 23.Schaedel C, de Monestrol I, Hjelte L, et al. Predictors of deterioration of lung function in cystic fibrosis. Pediatr Pulmonol. 2002;33(6):483-491. [DOI] [PubMed] [Google Scholar]
  • 24.Goss CH, Newsom SA, Schildcrout JS, Sheppard L, Kaufman JD. Effect of ambient air pollution on pulmonary exacerbations and lung function in cystic fibrosis. Am J Respir Crit Care Med. 2004;169(7):816-821. [DOI] [PubMed] [Google Scholar]
  • 25.Rosenfeld M, Emerson J, McNamara S, et al. ; EPIC Study Group Participating Clinical Sites. Baseline characteristics and factors associated with nutritional and pulmonary status at enrollment in the cystic fibrosis EPIC observational cohort. Pediatr Pulmonol. 2010;45(9):934-944. [DOI] [PubMed] [Google Scholar]
  • 26.Adam RJ, Michalski AS, Bauer C, et al. Air trapping and airflow obstruction in newborn cystic fibrosis piglets. Am J Respir Crit Care Med. 2013;188(12):1434-1441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hoegger MJ, Fischer AJ, McMenimen JD, et al. Cystic fibrosis. Impaired mucus detachment disrupts mucociliary transport in a piglet model of cystic fibrosis. Science. 2014;345(6198):818-822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ranganathan SC, Parsons F, Gangell C, Brennan S, Stick SM, Sly PD; Australian Respiratory Early Surveillance Team for Cystic Fibrosis. Evolution of pulmonary inflammation and nutritional status in infants and young children with cystic fibrosis. Thorax. 2011;66(5):408-413. [DOI] [PubMed] [Google Scholar]
  • 29.Gaskin KJ. Nutritional care in children with cystic fibrosis: are our patients becoming better? Eur J Clin Nutr. 2013;67(5):558-564. [DOI] [PubMed] [Google Scholar]
  • 30.Stick SM, Brennan S, Murray C, et al. ; Australian Respiratory Early Surveillance Team for Cystic Fibrosis (AREST CF). Bronchiectasis in infants and preschool children diagnosed with cystic fibrosis after newborn screening. J Pediatr. 2009;155(5):623-628. [DOI] [PubMed] [Google Scholar]
  • 31.Sly PD, Brennan S, Gangell C, et al. ; Australian Respiratory Early Surveillance Team for Cystic Fibrosis (AREST-CF). Lung disease at diagnosis in infants with cystic fibrosis detected by newborn screening. Am J Respir Crit Care Med. 2009;180(2):146-152. [DOI] [PubMed] [Google Scholar]
  • 32.Linnane B, Robinson P, Ranganathan S, Stick S, Murray C. Role of high-resolution computed tomography in the detection of early cystic fibrosis lung disease. Paediatr Respir Rev. 2008;9(3):168-174. [DOI] [PubMed] [Google Scholar]
  • 33.Emerson J, Rosenfeld M, McNamara S, Ramsey B, Gibson RL. Pseudomonas aeruginosa and other predictors of mortality and morbidity in young children with cystic fibrosis. Pediatr Pulmonol. 2002;34(2):91-100. [DOI] [PubMed] [Google Scholar]
  • 34.Li Z, Kosorok MR, Farrell PM, et al. Longitudinal development of mucoid Pseudomonas aeruginosa infection and lung disease progression in children with cystic fibrosis. JAMA. 2005;293(5):581-588. [DOI] [PubMed] [Google Scholar]
  • 35.Nixon GM, Armstrong DS, Carzino R, et al. Clinical outcome after early Pseudomonas aeruginosa infection in cystic fibrosis. J Pediatr. 2001;138(5):699-704. [DOI] [PubMed] [Google Scholar]
  • 36.Sun L, Jiang RZ, Steinbach S, et al. The emergence of a highly transmissible lineage of cbl+ Pseudomonas (Burkholderia) cepacia causing CF centre epidemics in North America and Britain. Nat Med. 1995;1(7):661-666. [DOI] [PubMed] [Google Scholar]
  • 37.Waters V, Atenafu EG, Salazar JG, et al. Chronic Stenotrophomonas maltophilia infection and exacerbation outcomes in cystic fibrosis. J Cyst Fibros. 2012;11(1):8-13. [DOI] [PubMed] [Google Scholar]
  • 38.Waters V, Atenafu EG, Lu A, Yau Y, Tullis E, Ratjen F. Chronic Stenotrophomonas maltophilia infection and mortality or lung transplantation in cystic fibrosis patients. J Cyst Fibros. 2013;12(5):482-486. [DOI] [PubMed] [Google Scholar]
  • 39.Dasenbrook EC, Merlo CA, Diener-West M, Lechtzin N, Boyle MP. Persistent methicillin-resistant Staphylococcus aureus and rate of FEV1 decline in cystic fibrosis. Am J Respir Crit Care Med. 2008;178(8):814-821. [DOI] [PubMed] [Google Scholar]
  • 40.Esther CR, Jr, Esserman DA, Gilligan P, Kerr A, Noone PG. Chronic Mycobacterium abscessus infection and lung function decline in cystic fibrosis. J Cyst Fibros. 2010;9(2):117-123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Rabin HR, Surette MG. The cystic fibrosis airway microbiome. Curr Opin Pulm Med. 2012;18(6):622-627. [DOI] [PubMed] [Google Scholar]
  • 42.Li L, Somerset S. The clinical significance of the gut microbiota in cystic fibrosis and the potential for dietary therapies. Clin Nutr. 2014;33(4):571-580. [DOI] [PubMed] [Google Scholar]
  • 43.Callaghan M, McClean S. Bacterial host interactions in cystic fibrosis. Curr Opin Microbiol. 2012;15(1):71-77. [DOI] [PubMed] [Google Scholar]
  • 44.Gibson RL, Burns JL, Ramsey BW. Pathophysiology and management of pulmonary infections in cystic fibrosis. Am J Respir Crit Care Med. 2003;168(8):918-951. [DOI] [PubMed] [Google Scholar]
  • 45.Cohen-Cymberknoh M, Kerem E, Ferkol T, Elizur A. Airway inflammation in cystic fibrosis: molecular mechanisms and clinical implications. Thorax. 2013;68(12):1157-1162. [DOI] [PubMed] [Google Scholar]
  • 46.Cutting GR. Modifier genes in Mendelian disorders: the example of cystic fibrosis. Ann N Y Acad Sci. 2010;1214:57-69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Accurso FJ, Sontag MK. Gene modifiers in cystic fibrosis. J Clin Invest. 2008;118(3):839-841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Drumm ML, Konstan MW, Schluchter MD, et al. ; Gene Modifier Study Group. Genetic modifiers of lung disease in cystic fibrosis. N Engl J Med. 2005;353(14):1443-1453. [DOI] [PubMed] [Google Scholar]
  • 49.Sly PD, Gangell CL, Chen L, et al. ; AREST CF Investigators. Risk factors for bronchiectasis in children with cystic fibrosis. N Engl J Med. 2013;368(21):1963-1970. [DOI] [PubMed] [Google Scholar]
  • 50.Elizur A, Cannon CL, Ferkol TW. Airway inflammation in cystic fibrosis. Chest. 2008;133(2):489-495. [DOI] [PubMed] [Google Scholar]
  • 51.Matthews LW, Doershuk CF, Wise M, Eddy G, Nudelman H, Spector S. A therapeutic regimen for patients with cystic fibrosis. J Pediatr. 1964;65:558-575. [DOI] [PubMed] [Google Scholar]
  • 52.Flume PA, Mogayzel PJ, Jr, Robinson KA, et al. ; Clinical Practice Guidelines for Pulmonary Therapies Committee. Cystic fibrosis pulmonary guidelines: treatment of pulmonary exacerbations. Am J Respir Crit Care Med. 2009;180(9):802-808. [DOI] [PubMed] [Google Scholar]
  • 53.Flume PA, Mogayzel PJ, Jr, Robinson KA, Rosenblatt RL, Quittell L, Marshall BC; Clinical Practice Guidelines for Pulmonary Therapies Committee; Cystic Fibrosis Foundation Pulmonary Therapies Committee. Cystic fibrosis pulmonary guidelines: pulmonary complications: hemoptysis and pneumothorax. Am J Respir Crit Care Med. 2010;182(3):298-306. [DOI] [PubMed] [Google Scholar]
  • 54.Mogayzel PJ, Jr, Naureckas ET, Robinson KA, et al. ; Pulmonary Clinical Practice Guidelines Committee. Cystic fibrosis pulmonary guidelines. Chronic medications for maintenance of lung health. Am J Respir Crit Care Med. 2013;187(7):680-689. [DOI] [PubMed] [Google Scholar]
  • 55.Rosenfeld M, Emerson J, Accurso F, et al. Diagnostic accuracy of oropharyngeal cultures in infants and young children with cystic fibrosis. Pediatr Pulmonol. 1999;28(5):321-328. [DOI] [PubMed] [Google Scholar]
  • 56.Davis SD, Fordham LA, Brody AS, et al. Computed tomography reflects lower airway inflammation and tracks changes in early cystic fibrosis. Am J Respir Crit Care Med. 2007;175(9):943-950. [DOI] [PubMed] [Google Scholar]
  • 57.Wainwright CE, Vidmar S, Armstrong DS, et al. ; ACFBAL Study Investigators. Effect of bronchoalveolar lavage-directed therapy on Pseudomonas aeruginosa infection and structural lung injury in children with cystic fibrosis: a randomized trial. JAMA. 2011;306(2):163-171. [DOI] [PubMed] [Google Scholar]
  • 58.Kerem E, Reisman J, Corey M, Canny GJ, Levison H. Prediction of mortality in patients with cystic fibrosis. N Engl J Med. 1992;326(18):1187-1191. [DOI] [PubMed] [Google Scholar]
  • 59.Davis SD, Brody AS, Emond MJ, Brumback LC, Rosenfeld M. Endpoints for clinical trials in young children with cystic fibrosis. Proc Am Thorac Soc. 2007;4(4):418-430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mott LS, Park J, Murray CP, et al. ; AREST CF. Progression of early structural lung disease in young children with cystic fibrosis assessed using CT. Thorax. 2012;67(6):509-516. [DOI] [PubMed] [Google Scholar]
  • 61.Borowitz D, Robinson KA, Rosenfeld M, et al. ; Cystic Fibrosis Foundation. Cystic Fibrosis Foundation evidence-based guidelines for management of infants with cystic fibrosis. J Pediatr. 2009;155(suppl 6):S73-S93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Rosenfeld M, Allen J, Arets BH, et al. ; American Thoracic Society Assembly on Pediatrics Working Group on Infant and Preschool Lung Function Testing. An official American Thoracic Society workshop report: optimal lung function tests for monitoring cystic fibrosis, bronchopulmonary dysplasia, and recurrent wheezing in children less than 6 years of age. Ann Am Thorac Soc. 2013;10(2):S1-S11. [DOI] [PubMed] [Google Scholar]
  • 63.Gustafsson PM, De Jong PA, Tiddens HA, Lindblad A. Multiple-breath inert gas washout and spirometry versus structural lung disease in cystic fibrosis. Thorax. 2008;63(2):129-134. [DOI] [PubMed] [Google Scholar]
  • 64.Owens CM, Aurora P, Stanojevic S, et al. ; London Cystic Fibrosis Collaboration. Lung Clearance Index and HRCT are complementary markers of lung abnormalities in young children with CF. Thorax. 2011;66(6):481-488. [DOI] [PubMed] [Google Scholar]
  • 65.Sawyer SM, Carlin JB, DeCampo M, Bowes G. Critical evaluation of three chest radiograph scores in cystic fibrosis. Thorax. 1994;49(9):863-866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Puderbach M, Eichinger M, Haeselbarth J, et al. Assessment of morphological MRI for pulmonary changes in cystic fibrosis (CF) patients: comparison to thin-section CT and chest x-ray. Invest Radiol. 2007;42(10):715-725. [DOI] [PubMed] [Google Scholar]
  • 67.Rosenfeld M, Emerson J, Williams-Warren J, et al. Defining a pulmonary exacerbation in cystic fibrosis. J Pediatr. 2001;139(3):359-365. [DOI] [PubMed] [Google Scholar]
  • 68.Fuchs HJ, Borowitz DS, Christiansen DH, et al. ; The Pulmozyme Study Group. Effect of aerosolized recombinant human DNase on exacerbations of respiratory symptoms and on pulmonary function in patients with cystic fibrosis. N Engl J Med. 1994;331(10):637-642. [DOI] [PubMed] [Google Scholar]
  • 69.Ferkol T, Rosenfeld M, Milla CE. Cystic fibrosis pulmonary exacerbations. J Pediatr. 2006;148(2):259-264. [DOI] [PubMed] [Google Scholar]
  • 70.Sanders DB, Bittner RC, Rosenfeld M, Hoffman LR, Redding GJ, Goss CH. Failure to recover to baseline pulmonary function after cystic fibrosis pulmonary exacerbation. Am J Respir Crit Care Med. 2010;182(5):627-632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Sequeiros IM, Jarad NA. Extending the course of intravenous antibiotics in adult patients with cystic fibrosis with acute pulmonary exacerbations. Chron Respir Dis. 2012;9(4):213-220. [DOI] [PubMed] [Google Scholar]
  • 72.VanDevanter DR, O’Riordan MA, Blumer JL, Konstan MW. Assessing time to pulmonary function benefit following antibiotic treatment of acute cystic fibrosis exacerbations. Respir Res. 2010;11:137-142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Fodor AA, Klem ER, Gilpin DF, et al. The adult cystic fibrosis airway microbiota is stable over time and infection type, and highly resilient to antibiotic treatment of exacerbations. PLoS ONE. 2012;7(9):e45001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Aaron SD, Ramotar K, Ferris W, et al. Adult cystic fibrosis exacerbations and new strains of Pseudomonas aeruginosa. Am J Respir Crit Care Med. 2004;169(7):811-815. [DOI] [PubMed] [Google Scholar]
  • 75.Høiby N, Frederiksen B, Pressler T. Eradication of early Pseudomonas aeruginosa infection. J Cyst Fibros. 2005;4(suppl 2):49-54. [DOI] [PubMed] [Google Scholar]
  • 76.Frederiksen B, Koch C, Høiby N. Antibiotic treatment of initial colonization with Pseudomonas aeruginosa postpones chronic infection and prevents deterioration of pulmonary function in cystic fibrosis. Pediatr Pulmonol. 1997;23(5):330-335. [DOI] [PubMed] [Google Scholar]
  • 77.Ramsey BW, Pepe MS, Quan JM, et al. ; Cystic Fibrosis Inhaled Tobramycin Study Group. Intermittent administration of inhaled tobramycin in patients with cystic fibrosis. N Engl J Med. 1999;340(1):23-30. [DOI] [PubMed] [Google Scholar]
  • 78.Máiz L, Girón RM, Olveira C, et al. Inhaled antibiotics for the treatment of chronic bronchopulmonary Pseudomonas aeruginosa infection in cystic fibrosis: systematic review of randomised controlled trials. Expert Opin Pharmacother. 2013;14(9):1135-1149. [DOI] [PubMed] [Google Scholar]
  • 79.Quon BS, Goss CH, Ramsey BW. Inhaled antibiotics for lower airway infections. Ann Am Thorac Soc. 2014;11(3):425-434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Goss CH, Ratjen F. Update in cystic fibrosis 2012. Am J Respir Crit Care Med. 2013;187(9):915-919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Main E, Prasad A, Schans C. Conventional chest physiotherapy compared to other airway clearance techniques for cystic fibrosis. Cochrane Database Syst Rev. 2005;(1):CD002011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Weller PH, Bush E, Preece MA, Matthew DJ. Short-term effects of chest physiotherapy on pulmonary function in children with cystic fibrosis. Respiration. 1980;40(1):53-56. [DOI] [PubMed] [Google Scholar]
  • 83.Quan JM, Tiddens HA, Sy JP, et al. ; Pulmozyme Early Intervention Trial Study Group. A two-year randomized, placebo-controlled trial of dornase alfa in young patients with cystic fibrosis with mild lung function abnormalities. J Pediatr. 2001;139(6):813-820. [DOI] [PubMed] [Google Scholar]
  • 84.Konstan MW, Wagener JS, Pasta DJ, et al. ; Scientific Advisory Group and Investigators and Coordinators of Epidemiologic Study of Cystic Fibrosis. Clinical use of dornase alpha is associated with a slower rate of FEV1 decline in cystic fibrosis. Pediatr Pulmonol. 2011;46(6):545-553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Duijvestijn YC, Brand PL. Systematic review of N-acetylcysteine in cystic fibrosis. Acta Paediatr. 1999;88(1):38-41. [DOI] [PubMed] [Google Scholar]
  • 86.Donaldson SH, Bennett WD, Zeman KL, Knowles MR, Tarran R, Boucher RC. Mucus clearance and lung function in cystic fibrosis with hypertonic saline. N Engl J Med. 2006;354(3):241-250. [DOI] [PubMed] [Google Scholar]
  • 87.Elkins MR, Robinson M, Rose BR, et al. ; National Hypertonic Saline in Cystic Fibrosis (NHSCF) Study Group. A controlled trial of long-term inhaled hypertonic saline in patients with cystic fibrosis. N Engl J Med. 2006;354(3):229-240. [DOI] [PubMed] [Google Scholar]
  • 88.Rosenfeld M, Ratjen F, Brumback L, et al. ; ISIS Study Group. Inhaled hypertonic saline in infants and children younger than 6 years with cystic fibrosis: the ISIS randomized controlled trial. JAMA. 2012;307(21):2269-2277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Jaques A, Daviskas E, Turton JA, et al. Inhaled mannitol improves lung function in cystic fibrosis. Chest. 2008;133(6):1388-1396. [DOI] [PubMed] [Google Scholar]
  • 90.Daviskas E, Anderson SD, Jaques A, Charlton B. Inhaled mannitol improves the hydration and surface properties of sputum in patients with cystic fibrosis. Chest. 2010;137(4):861-868. [DOI] [PubMed] [Google Scholar]
  • 91.Deterding RR, Lavange LM, Engels JM, et al. ; Cystic Fibrosis Therapeutics Development Network and the Inspire 08-103 Working Group. Phase 2 randomized safety and efficacy trial of nebulized denufosol tetrasodium in cystic fibrosis. Am J Respir Crit Care Med. 2007;176(4):362-369. [DOI] [PubMed] [Google Scholar]
  • 92.Ratjen F, Durham T, Navratil T, et al. ; TIGER-2 Study Investigator Group. Long term effects of denufosol tetrasodium in patients with cystic fibrosis. J Cyst Fibros. 2012;11(6):539-549. [DOI] [PubMed] [Google Scholar]
  • 93.Moss RB. Pitfalls of drug development: lessons learned from trials of denufosol in cystic fibrosis. J Pediatr. 2013;162(4):676-680. [DOI] [PubMed] [Google Scholar]
  • 94.Althaus M. ENaC inhibitors and airway re-hydration in cystic fibrosis: state of the art. Curr Mol Pharmacol. 2013;6(1):3-12. [DOI] [PubMed] [Google Scholar]
  • 95.Armstrong DS, Hook SM, Jamsen KM, et al. Lower airway inflammation in infants with cystic fibrosis detected by newborn screening. Pediatr Pulmonol. 2005;40(6):500-510. [DOI] [PubMed] [Google Scholar]
  • 96.Konstan MW, Davis PB. Pharmacological approaches for the discovery and development of new anti-inflammatory agents for the treatment of cystic fibrosis. Adv Drug Deliv Rev. 2002;54(11):1409-1423. [DOI] [PubMed] [Google Scholar]
  • 97.Lands LC, Stanojevic S. Oral non-steroidal anti-inflammatory drug therapy for lung disease in cystic fibrosis. Cochrane Database Syst Rev. 2013;6:CD001505. [DOI] [PubMed] [Google Scholar]
  • 98.Konstan MW, Byard PJ, Hoppel CL, Davis PB. Effect of high-dose ibuprofen in patients with cystic fibrosis. N Engl J Med. 1995;332(13):848-854. [DOI] [PubMed] [Google Scholar]
  • 99.Cheng K, Ashby D, Smyth RL. Oral steroids for long-term use in cystic fibrosis. Cochrane Database Syst Rev. 2013;6:CD000407. [DOI] [PubMed] [Google Scholar]
  • 100.Ren CL, Pasta DJ, Rasouliyan L, Wagener JS, Konstan MW, Morgan WJ; Scientific Advisory Group and the Investigators and Coordinators of the Epidemiologic Study of Cystic Fibrosis. Relationship between inhaled corticosteroid therapy and rate of lung function decline in children with cystic fibrosis. J Pediatr. 2008;153(6):746-751. [DOI] [PubMed] [Google Scholar]
  • 101.Saiman L, Marshall BC, Mayer-Hamblett N, et al. ; Macrolide Study Group. Azithromycin in patients with cystic fibrosis chronically infected with Pseudomonas aeruginosa: a randomized controlled trial. JAMA. 2003;290(13):1749-1756. [DOI] [PubMed] [Google Scholar]
  • 102.Saiman L, Anstead M, Mayer-Hamblett N, et al. ; AZ0004 Azithromycin Study Group. Effect of azithromycin on pulmonary function in patients with cystic fibrosis uninfected with Pseudomonas aeruginosa: a randomized controlled trial. JAMA. 2010;303(17):1707-1715. [DOI] [PubMed] [Google Scholar]
  • 103.Hoffman LR, Ramsey BW. Cystic fibrosis therapeutics: the road ahead. Chest. 2013;143(1):207-213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Kennedy MJ. Current status of gene therapy for cystic fibrosis pulmonary disease. Am J Respir Med. 2002;1(5):349-360. [DOI] [PubMed] [Google Scholar]
  • 105.Verkman AS, Galietta LJ. Chloride channels as drug targets. Nat Rev Drug Discov. 2009;8(2):153-171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Drug development pipeline. Cystic Fibrosis Foundation website. http://www.cff.org/research/drugdevelopmentpipeline.Accessed June 19, 2014.
  • 107.Kerem E, Konstan MW, De Boeck K, et al. ; Cystic Fibrosis Ataluren Study Group. Ataluren for the treatment of nonsense-mutation cystic fibrosis: a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Respir Med. 2014;2(7):539-547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Ramsey BW, Davies J, McElvaney NG, et al. ; VX08-770-102 Study Group. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med. 2011;365(18):1663-1672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.KALYDECO. US Food and Drug Administration website. http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/203188s008lbl.pdf. Accessed August 9, 2014.
  • 110.Clancy JP, Rowe SM, Accurso FJ, et al. Results of a phase IIa study of VX-809, an investigational CFTR corrector compound, in subjects with cystic fibrosis homozygous for the F508del-CFTR mutation. Thorax. 2012;67(1):12-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Cholon DM, Quinney NL, Fulcher ML, et al. Potentiator ivacaftor abrogates pharmacological correction of ΔF508 CFTR in cystic fibrosis. Sci Transl Med. 2014;6(246):246ra96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Veit G, Avramescu RG, Perdomo D, et al. Some gating potentiators, including VX-770, diminish ΔF508-CFTR functional expression. Sci Transl Med. 2014;6(246):246ra97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Pittman JE, Cutting G, Davis SD, Ferkol T, Boucher R. Cystic fibrosis: NHLBI Workshop on the Primary Prevention of Chronic Lung Diseases. Ann Am Thorac Soc. 2014;11(suppl 3):S161-S168. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Chest are provided here courtesy of American College of Chest Physicians

RESOURCES