Rapid Review
Progress in therapies for cystic fibrosis Kris De Boeck, Margarida D Amaral
Standard follow-up and symptomatic treatment have allowed most patients with cystic fibrosis to live to young adulthood. However, many patients still die prematurely from respiratory insufficiency. Hence, further investigations to improve these therapies are important and might have relevance for other diseases—eg, exploring how to increase airway hydration, how to safely downscale the increased inflammatory response in the lung, and how to better combat lung infections associated with cystic fibrosis. In parallel, development of modulators that target the underlying dysfunction in the cystic fibrosis transmembrane conductance regulator (CFTR) is fast moving forward. Existing treatments are specific to certain mutations, or mutation class, in CFTR. An effective, although not yet entirely corrective, treatment is available for patients with class III mutations, and a treatment with modest effectiveness is available for patients who are homozygous for Phe508del, albeit at a very high cost. Corrective treatments that are non-specific to mutation class and thus applicable to all patients—eg, gene therapy, cell-based therapies, and activation of alternative ion channels that bypass CFTR—are being explored, but they are still in early stages of development. In view of the large number of patients with very rare mutations, a plan to advance personalised biomarkers to predict treatment effect is also being investigated and validated.
Introduction Cystic fibrosis is the most common life-shortening rare disease, affecting around 32 000 individuals in Europe and about 85 000 individuals worldwide. In most European Union (EU) countries, adult patients now outnumber paediatric patients, but the median age at death remains low at roughly 28 years.1 However, for patients born in the past 15 years, median predicted survival in the UK is now greater than 50 years.2 The number of patients reported to disease registries worldwide rises steadily because of the widespread implementation of newborn screening, increased diagnosis in low-income and middle-income countries, and improved survival.3 Cystic fibrosis is caused by mutations in one gene— cystic fibrosis transmembrane conductance regulator (CFTR)—which encodes an epithelial chloride and bicarbonate ion channel.4,5 Most patients ultimately develop progressive lung disease with airway mucus obstruction, bacterial infection, and inflammation (figure 1) despite intense symptomatic (ie, standard) treatments, which do not treat the molecular cause of the disease (ie, defective CFTR protein). These symptomatic treatments include mucolytics to dissolve thick mucus, antibiotics to treat or prevent infections, and antiinflammatory agents to dampen chronic inflammation. However, treatments that act at the level of the CFTR molecular defect are necessary to block the series of events that lead to progressive lung disease. Patients have a large range of clinical phenotypes, which can be partly explained by the roughly 2000 CFTR gene mutations so far identified (see Cystic Fibrosis Mutation Database), of which only around 200 have been characterised in terms of disease liability (see CFTR2).7 Other genetic, cellular, and environmental factors, which remain largely unknown, also modify the clinical course of the disease and each individual’s response to therapy.8–11 Cystic fibrosis is often regarded as a model disease, since many pioneering studies in genetics, molecular
and cellular pathogenesis, and drug discovery that have been done in cystic fibrosis paved the way for other rare genetics disorders. Furthermore, evidence shows that the CFTR protein has a key role in several major respiratory conditions of high public health relevance that are rapidly becoming more prevalent in the EU, such as asthma and chronic obstructive pulmonary disease (COPD; estimated as the fourth leading cause of death worldwide), in which lack of functional CFTR at the cell surface has been demonstrated.12–14 The CFTR protein is even thought to have a role in smoking-related respiratory disease, since loss of CFTR at the plasma membrane is a major and early event in cells exposed to cigarette smoke and pollutants.15–18 These findings led some researchers to consider COPD and heavy smoking as so-called acquired CFTR deficiency, by contrast with genetic cystic fibrosis.
Lancet Respir Med 2016 Published Online April 1, 2016 http://dx.doi.org/10.1016/ S2213-2600(16)00023-0 Pediatric Pulmonology, Department of Pediatrics, University of Leuven, Leuven, Belgium (Prof K De Boeck PhD); and Faculty of Sciences, Biosystems and Integrative Sciences Institute (BioISI), University of Lisbon, Lisbon, Portugal (M D Amaral PhD) Correspondence to: Prof Kris De Boeck, Pediatric Pulmonology, Department of Pediatrics, University of Leuven, Leuven 3000, Belgium christiane.deboeck@uzleuven. be
For the Cystic Fibrosis Mutation Database see http://www.genet. sickkids.on.ca For more on CFTR2 see http://www.cftr2.org/
Key messages • 80 000 people worldwide have cystic fibrosis and will die prematurely from respiratory insufficiency • Standard follow-up and symptomatic treatments have allowed most patients to live to young adulthood • Improvement of conventional drugs continues to reduce disease complications by increasing airway hydration and ameliorate chronic lung inflammation and infection, thereby raising the expected age of survival • To further increase the lifespan and quality of life of patients, more effective treatments are needed and therapies correcting the underlying defect hold promise of achieving this goal • Modulators that target the underlying dysfunction in cystic fibrosis transmembrane conductance regulator (CFTR) in a mutation-specific or mutation-class-specific way are being developed at a steady pace • An effective treatment is available for patients with class III mutations (around 5% of all patients) and is becoming available for patients who are homozygous for Phe508del (roughly 40–45% of all patients) • So-called mutation agnostic corrective treatments—such as gene therapy, cell-based therapies, and activation of alternative ion channels to bypass CFTR—are in early stages of development
www.thelancet.com/respiratory Published online April 1, 2016 http://dx.doi.org/10.1016/S2213-2600(16)00023-0
1
Rapid Review
Symptomatic treatments
Molecular-defect treatments
6
Destructive cycle
7
Progressive loss of lung function
4 Decreased water content in airway surface liquid; thick mucus
3 Abnormal CI– permeability altered ionic transport
Inflammation and scarring
Bacterial infection
CI– 2 Defective CFTR protein
CFTR
5 Mucus obstruction and bronchiectasis
Na+
Na+
ENaC Na+
Na+
1 Two defective CFTR genes
Figure 1: Pathogenetic cascade that causes cystic fibrosis lung disease Cystic fibrosis is caused by mutations in the CFTR gene, which trigger a series of events that ultimately lead to severe lung deficiency. Most standard therapies treat the symptoms (eg, mucolytics to dissolve the thick mucus). However, treatments that target molecular defects acting earlier in this cascade of events (eg, CFTR modulators) can prevent progression to end-stage lung disease.6 CFTR=cystic fibrosis transmembrane conductance regulator. ENaC=epithelial sodium channel. Modified from Amaral.6
In this Review, we highlight progress in both standard symptomatic treatments and new therapies targeting the molecular defect in CFTR. In the fast-moving field of cystic fibrosis research, our discussion of these new therapies can be seen as an update to the 2013 Review in The Lancet Respiratory Medicine.19 Our focus here is to highlight recent and ongoing trials rather than past trials or preclinical studies.
Symptomatic therapies Understandably, most attention and resources are now focused on correction of the molecular defect with CFTR modulators—ie, correctors and potentiators. However, we should stress that none of these therapies are sufficiently effective to be used as stand-alone treatments at present. Even the most successful of these therapies (ie, ivacaftor in patients with a class III mutation) is used in addition to existing standard therapies.20 During treatment with ivacaftor, sweat chloride value, a marker of CFTR function, comes close to, but does not reach, the normal range.20,21 Additionally, although the clinical benefit of the drug is impressive (a 10% predicted improvement in forced expiratory volume in 1 s [FEV1]), disease progression is not stopped: the treatment is estimated to only halve the FEV1 rate of decline.22 Furthermore, during ivacaftor treatment, patients still have pulmonary exacerbations and other lung complications. Thus, until disease manifestations can be 2
prevented by eradicating the root cause and entirely blocking the pathophysiological cascade,23 the conventional symptomatic therapies, which enable most patients to live to adulthood, remain important.24 Further research to optimise these treatment modalities continues to be highly relevant for patients with cystic fibrosis and might also benefit patients with other chronic lung diseases, such as non-cystic fibrosis bronchiectasis and COPD. In this section, we discuss developments in the strategies to restore the airway surface liquid layer and improve mucociliary clearance, to dampen the excessive inflammatory response in the lung, and to control chronic lung infection.
Restoration of airway surface liquid and mucociliary clearance Central in the pathophysiology of cystic fibrosis lung disease are abnormally viscid secretions and deficient mucociliary clearance, leading to airway obstruction. The absence of chloride and bicarbonate secretion via the CFTR channel, coupled to excess sodium ion absorption via the epithelial sodium channel (ENaC, which is not downregulated by CFTR), leads to insufficient water secretion to the airway surface liquid layer. Whether this insufficiency results directly in decreased height of the periciliary layer, or whether the increased oncotic pressure in the overlying mucus layer pushes the
www.thelancet.com/respiratory Published online April 1, 2016 http://dx.doi.org/10.1016/S2213-2600(16)00023-0
Rapid Review
periciliary layer down (the new so-called gel-on-brush model),25 is under investigation. At present, inhalation of hypertonic saline and mannitol are used to improve airway hydration by altering osmolarity of the airway surface liquid.26,27 Dornase alfa (ie, recombinant human DNase-1) increases mucociliary clearance by decreasing sputum viscosity and is part of standard care for cystic fibrosis.24 As shown in the Epidemiologic Study of Cystic Fibrosis,28 the use of dornase alfa is associated with a reduction in the rate of FEV1 decline. However, around 30% of patients might be non-responders,29,30 probably because of insufficient sputum concentration of magnesium needed for DNase-1 activity or excessive sputum actin content, the naturally occurring inhibitor of DNase-1.30 When tested in vitro, PRX-110, a plant-cell-expressed recombinant form of human DNase-1, was more resistant than dornase alfa to the inhibitory effect of actin.31 Another strategy is to use actin depolymerising agents such as gelsolin or polyanions (eg, polyaspartate),32 which await proof-of-concept trials. To further restore mucociliary clearance, ENaC inhibitors are being considered as stand-alone therapy, in combination with existing hydrators such as hypertonic saline, or in combination with CFTR modulators. Indeed, ENaC inhibitors sit in between symptomatic therapy to improve mucociliary clearance and therapies aiming to improve the molecular defect by targeting non-CFTR channels (see below).
Safe reduction of excessive lung inflammation The complex association between inflammation and cystic fibrosis lung disease has been reviewed elsewhere.33,34 The hallmark of cystic fibrosis lung disease is infection by bacteria and other pathogens. However, compared with lung infection in other disorders, in cystic fibrosis the associated inflammation is increased but is still ineffective to clear pathogens from the lung. Lung inflammation in cystic fibrosis is driven by neutrophils, which contribute to structural lung damage, for example via release of neutrophil elastase. Although the mechanism is not fully understood, another hallmark is reduced invasiveness of pathogens despite a high local bacterial burden.35,36 Therefore, the balance between excessive inflammation (which contributes to lung damage) and insufficient inflammation (which possibly allows progression to more invasive disease) needs to be maintained, and potent anti-inflammatory therapies have been shown to worsen cystic fibrosis lung disease.37 Therapeutic strategies that only modestly decrease the inflammatory response, promote resolution of inflammation, and increase local antiprotease or antioxidant activity might prove safer and are now being explored. In a 4 year clinical trial in patients aged 6–14 years,38 the administration of high-dose systemic steroids (2 mg prednisone per kg bodyweight) on alternating days was
discontinued prematurely because of severe safety concerns. The low-dose treatment (1 mg prednisone per kg bodyweight) on alternating days did show a small benefit on repeated measurements of FEV1 (% predicted) but was again associated with serious side-effects, such as growth retardation, cataract, and more frequent pseudomonas infection.38 Inhaled steroids have no proven benefit.39 Leukotriene B4 (LTB4) is a potent activator of inflammatory responses mediated by neutrophils, macrophages, and monocytes, and its concentration is increased in the lung of patients with cystic fibrosis. In a clinical trial with the LTB4 receptor antagonist BIIL 284,37 active treatment was associated with increased pulmonary exacerbations in adult patients. In an animal model, BIIL 284 increased the incidence of bacterial infections in the mouse lung.40 Therefore, existing anti-inflammatory strategies are mainly restricted to non-steroidal drugs. Oral high-dose ibuprofen decreases the rate of decline of lung function, but fear of serious side-effects and the need for frequent drug level monitoring severely limit its use.41,42 In 2015, ibuprofen was identified to have some CFTR corrector activity in a human bronchial epithelial cell assay and in a mouse model.43 With treatments aiming to decrease inflammation, a more realistic expectation is to decrease pulmonary exacerbations and stabilise lung function (ie, less decline) rather than acute improvement in lung function.44 Obviously, a longer treatment period is needed to measure these outcomes.44 Several new compounds are being studied in clinical trials (table 1). Acebilustat is a small molecule that blocks the enzyme leukotriene A4 hydrolase, thereby decreasing the production of LTB4. The aim of acebilustat is to reduce airway obstruction by blocking excessive neutrophil influx and activation. In a phase 1 clinical trial with oral acebilustat (50 mg and 100 mg once daily) for 15 days in 17 adult patients with cystic fibrosis and mild to moderate lung disease, positive trends were seen in blood and sputum biomarkers, including LTB4, without changes in sputum microbiology.45 The planned phase 2 trial, EMPIRE-CF (ClinicalTrials.gov identifier NCT02443688), will aim to show a reduction in pulmonary exacerbations and in FEV1 decline. The oral anti-inflammatory compound ajulemic acid (Resunab) is designed to enhance the resolution of chronic inflammation by binding and activating cannabinoid receptor type 2, which is present on immune cells such as monocytes, T cells, and B cells. Ajulemic acid induces apoptosis of T cells, inhibits leucocyte migration, reduces cytokine release (eg, interleukin-6 and interleukin-8), decreases production of LTB4, and increases production of the anti-inflammatory lipoxin A4. The drug had a favourable safety profile in a phase 1 trial46 and will progress to phase 2 trials in cystic fibrosis (NCT02465450) and dermatomyositis. Research into strategies that increase local antiprotease and antioxidant activity to combat damage by neutrophils
www.thelancet.com/respiratory Published online April 1, 2016 http://dx.doi.org/10.1016/S2213-2600(16)00023-0
For more on Resunab see http:// www.corbuspharma.com/ product-pipeline/resunab
3
Rapid Review
Trial description
Intervention
Sponsor
Improving airway hydration NCT01619657
Phase 2 pilot study of safety and efficacy in newborns and infants
Preventive inhalation of 6% hypertonic saline (4 mL) versus 0·9% isotonic saline (4 mL), twice daily for 52 weeks
Heidelberg University, Heidelberg, Germany
NCT02378467
Phase 3 study of safety and efficacy (improvement in lung clearance index) in children aged 3–5 years
Inhalation of 7% hypertonic saline versus 0·9% isotonic saline, twice daily for 48 weeks
University of Washington, Seattle, WA, USA
NCT02343445
Phase 2 placebo-controlled trial of safety and efficacy in patients aged 12 years or older
Inhalation of ENaC blocker (P-1037) with saline or Parion Sciences hypertonic saline versus saline or hypertonic saline
Safely combatting inflammation NCT01270074
Phase 3 placebo-controlled trial of safety and efficacy in infants to prevent development of bronchiectasis at age 3 years
Azithromycin (10 mg/kg bodyweight) three times Queensland Children’s Medical per week, from age 3 months until 3 years Research Institute, Brisbane, QLD, Australia
NCT02443688
Phase 2 placebo-controlled trial of safety, tolerability, and efficacy in adult patients
Oral acebilustat, once daily for 48 weeks
NCT02465450
Phase 2 placebo-controlled trial of safety, tolerability, Ajulemic acid (JBT-101) pharmacokinetics, and efficacy in adult patients
Celtaxsys Corbus Pharmaceuticals
Improving control of lung infection NCT01455675
Phase 3 trial of safety and efficacy for prevention of re-infection with Pseudomonas aeruginosa
Avian polyclonal anti-P aeruginosa antibodies versus placebo
Mukoviszidose Institut, Bonn, Germany
NCT02526004
Phase 3 trial of microbiome-determined antibiotic therapy in cystic fibrosis exacerbations
Microbiome-guided therapy versus standard therapy
University College Cork, Cork, Ireland
ENaC=epithelial sodium channel.
Table 1: Key ongoing clinical trials of standard therapies
has been active for more than a decade, but progress is slow. The continued interest is explained by the safety of these approaches; unfortunately, efficacy has been more difficult to prove. In an open-label multicentre study in Germany in 52 patients with cystic fibrosis, pseudomonas lung infection, and raised free elastase levels in induced sputum,47 daily inhalation of α1 antitrypsin (25 mg) for 4 weeks resulted in raised sputum α1 antitrypsin levels and reductions in free elastase levels and concentrations of interleukin-8, TNF-α, interleukin-1β, and LTB4; decreased proportion of neutrophils; and a reduction in Pseudomonas aeruginosa colony count. However, these changes were not associated with any change in lung function.46 In a doubleblind phase 2 study (NCT01684410),48 higher doses (ie, 100 mg and 200 mg) of aerosolised α1 antitrypsin (a liquid preparation of purified α1 proteinase inhibitor from pooled human plasma) inhaled daily via a nebuliser for 3 weeks were safe and well tolerated. Increasing the amount of the antioxidant glutathione in the lungs has been attempted, either by inhalation of glutathione (which is deficiently transported by CFTR) or by oral administration of the prodrug N-acetylcysteine, which is then converted to glutathione.49–54 Although some changes in inflammatory parameters were noted with inhaled glutathione,49–54 no sustained change in lung function or exacerbations was seen in a subsequent 6-month trial.52 Conversely, in a 24-week double-blind trial with oral N-acetylcysteine (n=70),54 no change in inflammatory markers (eg, sputum neutrophil elastase or plasma interleukin-8) was seen, but lung function was stabilised without a significant change in pulmonary 4
exacerbations. However, in view of the small sample size, the possibility of this being a chance finding cannot be excluded. The authors hypothesised other potential mechanisms of action, such as a positive effect of N-acetylcysteine on other pathways of inflammation or on CFTR trafficking.
Efficient control of chronic lung infection: treatments beyond antibiotics Intensive use of oral, inhaled, and systemic antibiotics has undoubtedly improved survival of patients with cystic fibrosis and is still one of the mainstay therapies.24 Ultimately, however, Pseudomonas species will develop widespread antibiotic resistance, and pathogens that are increasingly more difficult to treat—such as Burkholderia cepacia complex, Achromobacter species, atypical mycobacteria, and fungi—might also emerge. The development of new inhaled antibiotics and better or faster inhalation devices has been the topic of several recent reviews.55–57 Therefore, we focus on strategies other than antibiotics to combat chronic lung infection in cystic fibrosis. Biofilm production is a common adaptation mechanism during chronic infection, and is typical for chronic pseudomonas infection in the cystic fibrosis lung. Disruption of the biofilm could thus help to eradicate Pseudomonas species, and several strategies that are being explored include saccharides such as liposomal β glycan58 and OligoG,59,60 an oligosaccharide derived from brown algae. Safety and tolerability of nebulised OligoG have been proven in healthy volunteers and in patients,61 and patterns
www.thelancet.com/respiratory Published online April 1, 2016 http://dx.doi.org/10.1016/S2213-2600(16)00023-0
Rapid Review
of lung deposition after inhalation are now being studied. Another compound that has been reported to disrupt the biofilm (by a yet-unknown mechanism), oral cysteamine, is being tested in a phase 1 trial for tolerability and pharmacokinetics.62 Interestingly, this compound, in combination with a tea flavonoid, has been reported to rescue traffic of Phe508del-CFTR by restoring autophagy.63,64 Nitric oxide concentrations are decreased in exhaled air from patients with cystic fibrosis compared with healthy individuals.65 The importance of this finding is not entirely clear but has been linked to the increased susceptibility to infection.66 Attempts to increase nitric oxide production by inhalation of interferon-γ to stimulate nitric oxide synthase proved unsafe, since more exacerbations were seen in patients given the high dose; neither high-dose treatment nor low-dose treatment demonstrated benefit on lung function, sputum imflammatory markers, or bacterial density.67 Strategies to increase nitric oxide concentration without interfering with complex inflammatory cell signalling seem safer, but the effectiveness of these approaches is still to be proven. Direct inhalation of nitric oxide effectively decreased bacterial load in a rat model of P aeruginosa pneumonia.68 The safety and tolerability of nitric oxide inhalation (160 ppm for 30 min three times per day) have also been assessed in patients with cystic fibrosis, and at least transient reductions in bacterial load were noted.69 Nitric oxide concentrations in the airways can also be increased by repeated inhalations of L-arginine, the substrate for nitric oxide synthase. In a pilot trial,70 inhalation of 500 mg L-arginine twice daily for 2 weeks was well tolerated, and FEV1 increased by 56 mL on average, although this increase was not significant; no change in inflammatory markers in sputum was reported. Interest in treatment with bacteriophages—ie, viruses that infect and replicate in specific strains of bacteria— is mounting.71 Especially for patients colonised with multiresistant organisms, controlling the infection with bacteriophage treatment could be a last resort. At present, no clinical trial has started, but preparatory work specific for cystic fibrosis is advancing. AntiPseudomonas bacteriophages have been shown to retain their activity when nebulised.72 Strict safety regulations imposed by health authorities are one of the major hurdles to assess the efficacy of this therapy in the clinic. Finally, a new and rapidly expanding research area is the lung microbiota, but a full discussion of this topic is beyond the scope of this Review. Knowledge of lung microbiota has considerably changed the way lung infection in cystic fibrosis is viewed. The healthy airways are colonised by a wide spectrum of bacteria. By contrast, in the airways of patients with cystic fibrosis, bacterial diversity and species richness decrease as the disease progresses and lung function decreases.73 However, this improved knowledge of the microbial community has not yet translated into more judicious or efficacious use
of antibiotics during exacerbations. An EU-funded project and clinical trial have started to address these issues and fill this knowledge gap (see CF Matters).
For more on CF Matters see http://www.cfmatters.eu/
Therapies to correct molecular defects CFTR modification therapies: classes of CFTR mutations The roughly 2000 CFTR gene alterations that have so far been described consist of missense (39·6%), frameshift (15·6%), splicing (11·4%), and nonsense (8·3%) mutations; large (2·6%) and in-frame (2·0%) deletions or insertions; promoter mutations (0·7%); and presumed non-pathological variants (15·0%).6,74 However, the 3 base-pair deletion of phenylalanine 508 (Phe508del) is present in around 85% of patients worldwide, with a higher frequency reported in northern Europeans than in southern Europeans.75 All mutations that cause cystic fibrosis ultimately lead to a defect in cAMP-regulated chloride and bicarbonate secretion by epithelial cells, but many reasons exist for the different causative mechanisms.76 Not only is elucidation of the molecular and cellular effects brought about by CFTR mutations informative for structure–function studies, but it can also provide the scientific basis for mutation-specific corrective therapies.23 Therefore, these mutations can be grouped according to their functional defect into seven classes (figure 2).6,77,78 Class I mutations affect protein production and include mostly nonsense mutations (ie, those with premature stop codons), thus often causing degradation of mRNA by nonsense-mediated decay. Class II mutations include Phe508del and affect CFTR protein traffic as a result of protein misfolding and retention at the endoplasmic reticulum (ER) by the ER quality control mechanism. Such retention is followed by premature degradation, which prevents the protein from trafficking to the cell surface, thus severely reducing CFTR function.79,80 Class III mutations impair gating of the CFTR channel. Class IV mutations cause a substantial decrease in CFTR channel conductance (ie, flow) of chloride and bicarbonate ions. Class V mutations lead to a major reduction in the levels of normal CFTR protein, often because of alternative splicing that generates both aberrant and normal mRNA species, the proportion between which might vary among patients and in different organs of each patient. Class VI mutations destabilise CFTR at the cell surface, either by increasing CFTR endocytosis or by decreasing its recycling back to the cell surface. Finally, class VII mutations are so-called unrescuable mutations, because they cannot be pharmacologically rescued per se—eg, large deletions such as the dele2,3(21kb) mutation.81 However, promising so-called one-size-fits-all therapeutic strategies (also known as mutation agnostic; ie, independent of the mutation class) will also be suitable for this class of mutations (see below). Novel emerging drugs targeting the underlying defect—ie, mutant CFTR—open up new opportunities
www.thelancet.com/respiratory Published online April 1, 2016 http://dx.doi.org/10.1016/S2213-2600(16)00023-0
5
Rapid Review
CI– CI– CI– – – CI CI– CI
CI–
CI–
CI–
CFTR
Wild-type CFTR Class I
Class II
Class III
Class IV
Class V
Class VI
Class VII
CFTR defect
No protein
No traffic
Impaired gating
Decreased conductance
Less protein
Less stable
No mRNA
Mutation examples
GLy542X, Trp1282X
Phe508del, Asn1303Lys, Ala561Glu
Gly551Asp, Ser549Arg, Gly1349Asp
Arg117His, Arg334Trp, Ala455Glu
Ala455Glu, 3272-26A→G, 3849+10 kg C→T
c. 120del23, rPhe508del
dele2,3(21 kb), 1717-1G→A
Corrective therapy
Rescue synthesis
Rescue traffic
Restore channel activity
Restore channel activity
Correct splicing
Promote stability
Unrescuable
Drug (approved)
Read-through compounds (no)
Correctors (yes)
Potentiators (yes)
Potentiators (no)
Antisense oligonucleotides, correctors, potentiators? (no)
Stabilisers (no)
Bypass therapies (no)
Figure 2: Classes of gene mutations and respective therapeutic strategies CFTR mutations are grouped into seven functional classes, with the expectation that the same modulators will be applicable to all the defects in one class. However, class VII mutations are not expected to be rescuable by any modulator. A therapeutic strategy for class VII mutations could involve stimulation of alternative chloride channels (ie, bypass therapies). CFTR=cystic fibrosis transmembrane conductance regulator. rPhe508del=rescued Phe508del. Modified from Amaral.6
for so-called curative treatment. At present, two of these drugs have been approved for clinical use: ivacaftor benefits only patients with gating mutations (around 5% of all patients) and, to some extent, patients with the Arg117His mutation; lumacaftor is beneficial for patients with the homozygous Phe508del genotype (ie, roughly 40–45% of all patients with cystic fibrosis; table 2).20,21,82–84 Ivacaftor was shown to decrease sweat chloride concentration by a mean of 50 mmol/L, augment predicted FEV1 by a mean of 10%, reduce the number of pulmonary exacerbations, and improve body-mass index (BMI), Z scores, and quality of life, not only in patients with the Gly551Asp mutation but also in patients with other class III mutations.20,21 Ivacaftor has now been used in patients with the Gly551Asp mutation for more than 5 years. Across age and disease severity groups, a robust benefit was seen after marketing authorisation by both the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA).85–87 Long-term benefits are starting to become apparent87—ie, less frequent infections with pseudomonas,88 reduced rate of lung function decline,21 improved glucose tolerance,89 sustained weight gain, and better growth in children.90 In vitro, ivacaftor potentiates not only class III CFTR 6
mutants but also the normal CFTR channel and some mutant proteins of classes IV and V.91 In a phase 3 trial,82 a 5% improvement in lung function was reported in adult patients with the class IV mutation Arg117His; however, this improvement was not seen in children with the same mutation. In the entire cohort with this mutation, improvements in sweat chloride (−24 mmol/L) and quality-of-life score were seen.82 However, clinical benefit in patients with other class IV or V mutations with residual function still needs to be firmly proven. New strategies are emerging for class V mutations that affect splicing, and a particular area of interest is the correction of alternative splicing with antisense oligonucleotides.92 The introduction of ivacaftor led to great optimism, even though this drug is indicated in only up to 10% of patients with cystic fibrosis. Ivacaftor provides proof of concept that small-molecule therapy that interferes with the molecular defect is possible, which encouraged patients to participate in clinical trials with CFTR modulators. Yet, many challenges remain to ensure that patients have access to new treatments. Ivacaftor is very expensive (around US$311 000 per patient per year), and some health authorities are slow to approve reimbursement, since the need for lifelong treatment poses a substantial burden on health-care budgets.93,94
www.thelancet.com/respiratory Published online April 1, 2016 http://dx.doi.org/10.1016/S2213-2600(16)00023-0
Rapid Review
Several companies are now developing other drugs that potentiate CFTR channel function, and clinical trials are ongoing (table 3). Results from phase 3 trials in patients homozygous for Phe508del showed that combination therapy of lumacaftor (a corrector that partly rescues Phe508del-CFTR traffic) and ivacaftor (which potentiates channel function) induced a significant but modest (2·6–4·0%) improvement in lung function and a Frequency
30–40% reduction in pulmonary exacerbations— defined as treatments with additional antibiotics because of an increase in pulmonary symptoms—over the 24 week treatment period.83 Nonetheless, this combination therapy has already been approved by the FDA and EMA for patients homozygous for Phe508del aged 12 years or older (table 2). Again, cost (around US$250 000 per patient per year) is a major issue, since 40–50% of patients qualify for this treatment.
Corrective therapy Limitations
Status
Gly551Asp and eight other gating mutations (class III)
4–5%
Ivacaftor
Very high cost (around US$311 000 per patient per year)
Approved in the EU and USA for patients aged 2 years or above
Arg117His (class IV)
1–2%
Ivacaftor
Very high cost; variable response
Approved in the EU for patients aged 18 years or above, and in the USA for patients aged 6 years or above
Lumacaftor plus ivacaftor
Very high cost (around US$250 000 per patient per year); low efficacy; variable response
Approved in the EU and USA for patients aged 12 years or above
Homozygous Phe508del
40–45%
Heterozygous Phe508del
40%
None
¨
¨
Nonsense mutations
10%
Ataluren
Benefit uncertain
Phase 3 trial (NCT02139306) ongoing in patients who are not using inhaled aminoglycosides
Other mutations (including class VII mutations)
10–15%
None
¨
¨
EU=European Union.
Table 2: Corrective therapies targeting the molecular defect in cystic fibrosis, by mutation type
Mechanism of action
Stage of development
Sponsor
Ataluren
Rescue CFTR protein synthesis by read-through of nonsense mutations
Phase 3 confirmatory study (NCT02139306) of efficacy and safety is ongoing in patients with nonsense CFTR mutations who are not taking inhaled aminoglycosides
PTC Therapeutics
VX-661 (corrector) plus ivacaftor (potentiator)
Rescue Phe508del-CFTR protein to the cell surface with a corrector and stimulate channel function with a potentiator
Phase 3 studies (NCT02347657, NCT02392234, and NCT02412111) of efficacy and safety are ongoing in patients aged 12 years or above with homozygous and heterozygous Phe508del-CFTR mutation
Vertex Pharmaceuticals
N91115 (proteostasis regulator)
Stabilise Phe508del-CFTR protein after being rescued to the cell surface
Phase 2 study (NCT02589236) is ongoing in adults who are homozygous for Phe508del and receiving lumacaftor–ivacaftor combination treatment
Nivalis Therapeutics
Riociguat
Improve CFTR expression
Phase 2 trial (NCT02170025) of safety, tolerability, and efficacy is underway in adults homozygous for Phe508del
Bayer
QR-010 (antisense oligonucleotide)
In-vivo correction of the defect in Phe508del-CFTR mRNA
Phase 1b trial (NCT02532764) is ongoing to assess the safety of single and multiple ascending doses of inhaled QR-010 in adult patients with homozygous Phe508del; phase 1 open-label exploratory study (NCT02564354) is underway to assess the efficacy of intranasal administration on nasal potential difference (a measure of CFTR function) in patients with homozygous and heterozygous Phe508del
ProQR Therapeutics
GLPG1837 (potentiator) Rescue CFTR function by stimulating channel function
Phase 2a trial of safety and tolerability is ongoing in adults with at least one class III mutation
Galapagos NV
QBW251 (potentiator)
Rescue CFTR function by stimulating channel function
Phase 1 (NCT02190604) and phase 2a (NCT02190604) trials are underway to assess safety, tolerability, and pharmacokinetics in healthy volunteers and adults with at least one class III–VI mutations
Novartis Pharmaceuticals
FDL169 (potentiator)
Rescue CFTR function by stimulating channel function
Phase 1 trial (NCT02359357) of safety, tolerability, and pharmacokinetics of single and repeat oral doses in healthy male volunteers is underway
Flatley Discovery Laboratory LLC
C-10355 and C-10358 (potentiators)
Rescue CFTR function by stimulating channel function
Phase 1 trial (NCT02392702) of safety, tolerability, and pharmacokinetics in healthy volunteers, with a pharmacokinetic comparison with ivacaftor, completed.
Concert Pharmaceuticals
Other correctors and potentiators
In the pipeline of several companies, pending results from preclinical and phase 1 studies of Vertex Pharmaceuticals, Rescue CFTR function at the cell surface by correctors with complementary action Galapagos NV, and Flatley two correctors and stimulate channel Discovery Laboratory LLC function with a potentiator in patients homozygous or heterozygous for Phe508del
CFTR=cystic fibrosis transmembrane conductance regulator.
Table 3: Treatments targeting molecular defects in CFTR and relevant clinical trials
www.thelancet.com/respiratory Published online April 1, 2016 http://dx.doi.org/10.1016/S2213-2600(16)00023-0
7
Rapid Review
For more on the Cystic Fibrosis Trust see http://www. cysticfibrosis.org.uk/
At present, combination therapy has not shown efficacy in patients heterozygous for Phe508del. Few treatment options are available for patients with the predominant Phe508del mutation (present in roughly 85% of all patients),19,78,95 since the rescue of Phe508del by one corrector is indeed inefficient. An improved understanding of how the complex three-dimensional folding of the Phe508del-CFTR protein is disturbed points to the need for a combination of correctors with different mechanisms of action, together with a potentiator to allow more efficient rescue of the mutant protein.96,97 Ivacaftor potentiates normal CFTR, class III mutants, and several mutated CFTR proteins associated with residual function.91 However, different corrector–potentiator combinations are probably needed to rescue different misfolded class II mutant proteins.98 As has been shown for class V splicing defects,92 antisense oligonucleotides can also be used to correct the Phe508del mutation. Single-stranded antisense RNA-based oligonucleotides can act as guide sequences to repair the targeted abnormal mRNA. The repaired mRNA is then translated into a wild-type CFTR protein.99 For one such therapy (QR-010), clinical trials are underway to assess the safety, tolerability, and pharmacokinetics of single and multiple increasing doses of the inhaled drug in patients homozygous for Phe508del (NCT02532764 and NCT02564354). These studies will also investigate changes in sweat chloride, weight, Cystic Fibrosis Questionnaire Revised Respiratory Symptom Score, and FEV1. The so-called read-through strategy for patients with premature termination codons (ie, class I mutations) remains an important concept because it has potential in all diseases caused by a nonsense mutation. The EMA has granted conditional approval for ataluren in patients with Duchenne’s muscular dystrophy caused by a premature stop codon. For patients with cystic fibrosis, the 48 week double-blind phase 3 trial of ataluren100 did not lead to a firm conclusion; indeed, the primary endpoint of improvement in FEV1 was not met. However, a prespecified subgroup analysis showed that patients who were not using inhaled aminoglycosides had less decline in lung function than had those given placebo. In view of this finding, a confirmatory phase 3 trial in patients who are not using inhaled aminoglycosides (NCT02139306) is ongoing. Moreover, the number of new read-through drugs in preclinical development is growing.
Targeting non-CFTR channels: the bypass approach For patients with so-called unrescuable mutations (class VII), the most straightforward approach is to target alternative non-CFTR anion channels, such that the loss of CFTR-mediated chloride transport in airway epithelia can be bypassed to restore ion transport.21 These therapies, which are sometimes called mutation agnostic, might compensate not only for the loss of CFTR-mediated anion 8
conductance but also for other cellular defects, including ENaC hyperactivity and the reduced height and pH of airway surface liquid, which in turn impair bacterial clearance and killing,101 enhance sodium absorption,102 and reduce mucus fluidity.103–105 Many alternative ion channels and transporters are promising drug targets. Anoctamin 1 (ANO1; also known as TMEM16A) is a calcium-activated chloride channel present in many epithelial cells,106–110 and ANO6 (also known as TMEM16F) is an essential component of the outwardly rectifying chloride channel.111 Genetic variants of SLC9A3, which encodes a sodium–proton exchanger, have been associated with pseudomonas infections and decline in pulmonary function.110,112 The chloride–bicarbonate transporter SLC26A9 is associated with susceptibility to meconium ileus113 and diabetes,114 which are frequent complications in cystic fibrosis. However, the logical strategy of stimulating alternative chloride channels has been unsuccessful so far. Inhalation of denufosol, which stimulates calciumactivated chloride channels through the P2Y2 subtype of purinergic receptors, was promising in phase 2 investigation, but robust clinical benefit was lacking.115 Nevertheless, innovative approaches to find new pathways to stimulate these channels are underway (eg, INOVCF project116 and Cystic Fibrosis Trust in the UK). Another approach is to target ENaC, a major regulator of salt and water re-absorption. Since ENaC is downregulated by CFTR, the absence of functional CFTR leads to ENaC hyperactivity in cystic fibrosis,117,118 which in turn results in water depletion from the airway surface liquid and reduction in its height and fluidity.119 Mice that overexpressed β ENaC mimicked various aspects of human respiratory disease in cystic fibrosis,120 whereas mice with CFTR knockout or mutations did not develop lung disease.121 In view of the major role of ENaC in the airway, ENaC inhibitors have been developed to reduce ENaCmediated sodium hyperabsorption and increase hydration of airway surface liquid. These include specific inhibitors such as amiloride (the short-lived prototype ENaC blocker) and its derivatives benzamil or PS552, and activators of purinergic receptors (ATP, UTP, or denufosol) that inhibit ENaC through stimulation of calcium-activated chloride channels.23 However, repeated inhalations of amiloride did not show benefits in clinical trials.122 Development of longacting blockers was halted because of hyperkalaemia or other side-effects. Newer ENaC blockers, such as P-1037 and GS-9411 (NCT00999531), are being developed. The safety and tolerability of P-1037 (NCT02343445) are being studied as monotherapy and in combination with hypertonic saline. The inhaled combination had a transitory effect on mucus hydration and mucociliary clearance in healthy individuals and could lead to a greater or more prolonged benefit than that with inhaled hypertonic saline alone.123 Along these lines, a systems biology screen of more than 6000 genes identified a key ENaC regulator, DGK1,
www.thelancet.com/respiratory Published online April 1, 2016 http://dx.doi.org/10.1016/S2213-2600(16)00023-0
Rapid Review
which normalises (but does not totally block) ENaC function and restores fluid absorption to normal levels in primary lung cells from patients with cystic fibrosis.124 However, excessive blocking of ENaC might cause severe harm due to hyperkalaemia or undesirable accumulation of fluid in the lungs (ie, pulmonary oedema).125
Gene, cell-based, and other mutation agnostic therapies After cloning the CFTR gene, gene therapy was originally the priority for therapy, but gene transfer into lung cells in vivo soon proved more challenging than anticipated. Nevertheless, research into gene therapy led to a phase 2b clinical trial (NCT01621867)126 of DNA plasmid containing the CFTR cDNA (ie, a copy of the CFTR mRNA) complexed with cationic liposomes (ie, a non-viral vector), which was delivered once a month to the lungs of patients with cystic fibrosis. Results from this double-blind randomised trial showed a significant, albeit modest, effect in the treatment group versus placebo at 12-month follow-up. The 3·7% difference in FEV1 resulted from stabilisation of lung function in treated patients and a decline in lung function in the placebo group.126 Despite the fact that the placebo was only 0·9% saline (ie, no empty vector, CFTR DNA, or liposomes), efficacy data are at least encouraging, and improved versions of this therapy might become a treatment option in the future. Cell-based therapy is another mutation agnostic therapeutic approach with high potential. Indeed, cellular reprogramming and genome editing technologies (based on targeting of nucleases) have made possible the correction of patient-specific pluripotent stem cells and induction of their differentiation into a specific cell type. This approach has been shown to be feasible for cystic fibrosis in intestinal organoids and induced pluripotent stem cells.127,128 Therefore, the disease-target tissues could potentially be repopulated with these corrected cells. However, safety issues still need to be resolved regarding the clinical use of stem cells before cell-based approaches can become a therapeutic option. Another class of emerging therapies focus on systems approaches to target both mechanisms of disease and their therapeutic correction.11 One such strategy targets proteostasis (ie, maintenance of the cellular proteome in a folded, functionally competent state) and can be applied not only for cystic fibrosis but also for a group of so-called protein misfolding diseases.129 A successful example is suberolyanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor and regulator of the chaperone protein Hsp90, which has been shown to restore the cell surface expression and channel activity of Phe508del-CFTR to 28% of wild-type levels in human primary airway epithelial cells.130
Repurposing existing drugs for so-called orphan mutations Despite the great breakthrough in drug development, the first CFTR modulator, ivacaftor, targets only 5–10% of patients at best, and its combination with lumacaftor
benefits another 40–45% of patients (ie, those who are homozygous for Phe508del). Thus, effective treatment for the remaining 50% of patients remains an unmet need. Indeed, the wide range of CFTR mutations consists of many uncommon variants (so-called orphan mutations), for which prediction of disease outcome is difficult, since the respective functional defect has not been defined. Although phenotypes vary widely, many of these mutations in classes IV–VI result in partial CFTR function and milder, so-called atypical, forms of disease.131,132 Since each of these mutations affect very few patients worldwide, they pose considerable challenges not only in establishing the diagnosis but also in assessing the potential of the new mutation-based therapies (so-called theranostics—ie, the use of molecular or functional diagnostic tests and targeted therapeutics in an interdependent, collaborative manner, which aims to individualise treatment by targeting therapy to an individual’s disease subtype and genetic or functional profile).131–133 Some of these patients with orphan mutations are likely to respond to existing CFTR modulators. Therefore, there is a need to functionally characterise these orphan mutations and to establish validated biomarkers for theranostics. Without any preselection of suitable candidates, the N-of-1 clinical trial that considers an individual patient as the sole unit of observation is an inefficient, costly, and time-consuming approach.
Using ex-vivo response to predict in-vivo response For many CFTR mutations, how they disturb the CFTR pathway is unknown, and most are very rare. For these rare mutations, use of ex-vivo models is a promising approach to predict in-vivo response. Bioelectric measurements of CFTR dysfunction in native or cultured tissues from patients that are already in use for cystic fibrosis diagnosis and prognosis134,135 have also been proposed for personalised therapies.6,136 The most promising models for treatment response are intestinal organoids and primary cultures of epithelial cells derived from bronchi or nasal scrapings.98,136,137 Primary intestinal organoids are grown from rectum crypts obtained via suction biopsy and contain stem cells; therefore, they can be expanded indefinitely to obtain a continuous source of patient-specific material. Forskolin induces swelling of organoids from healthy individuals via stimulation of the CFTR channel, but not in organoids from patients with cystic fibrosis, and the magnitude of swelling correlates with CFTR activity.137 Organoids from patients can be incubated with drug candidates to assess their effect on CFTR function. If CFTR function and organoid swelling are restored by a specific modulator in a patient’s tissue ex vivo, then testing the clinical benefit in vivo is the next logical step. There is therefore an unmet need to test compounds that improve organoid function ex vivo in patients via N-of-1 trials, so as to prove that a positive ex-vivo organoid response indeed correlates with a good in-vivo clinical response. Therefore, the research community
www.thelancet.com/respiratory Published online April 1, 2016 http://dx.doi.org/10.1016/S2213-2600(16)00023-0
9
Rapid Review
Search strategy and selection criteria We searched PubMed for articles published in English from Jan 1, 2014, to Sept 30, 2015, with a focus on new areas of research for symptomatic therapies and therapies aiming to correct the molecular defect, and on recent treatments that are in or close to clinical development. We used the search terms “cystic fibrosis” and “novel therapies”, “novel drugs”, “treatments”, “correctors”, “potentiators”, “basic defect therapies”, or “innovative therapeutic strategies”. To complement our search on new drugs that are being developed, we also reviewed abstracts from the 38th European Cystic Fibrosis Conference and the 28th Annual North American Cystic Fibrosis Conference, which were both held in 2015.
has to move towards more standardised and accessible formats for the generalised use of these innovative approaches as surrogate markers in clinical trials and in personalised medicine. These major technological advances offer new possibilities for targeted therapies by precisely predicting which patients will or will not respond to a medical therapy.138
Conclusion On multiple fronts, treatments that improve outcome in patients with cystic fibrosis are emerging. Studies of many symptomatic treatments aiming to improve the effects of CFTR dysfunction (eg, abnormal mucociliary clearance and chronic lung infection; table 1) and treatments that target the underlying defect (table 3) are underway. Therapies targeting the molecular defect can be mutation agnostic (eg, gene therapy) or can be specific to mutation class or even particular mutations. With more than 2000 different CFTR mutations causing the disease, a precision medicine approach could offer great benefit. Contributors KDB reviewed articles on standard therapies, and MDA reviewed articles on treating the basic defect. Both authors wrote the manuscript. Declaration of interests KDB is a member of Steering Committee, an Advisory Board Member, and a principal investigator in studies funded by Vertex Pharmaceuticals; a member of the data monitoring committee in studies funded by Aptalis; a consultant and principal investigator in studies funded by Galapagos, PTC Therapeutics, and Boehringer; an Advisory Board Member and principal investigator in studies funded by Pharmaxis; and a consultant for Ablynx, Protalix, and Raptor during the conduct of the study. MDA reports personal fees from Vertex Pharmaceuticals, PTC Therapeutics, Gilead Sciences, and Galapagos; and grants from Gilead Génese outside of the submitted work. Acknowledgments KDB is supported by Instituut voor Wetenschap en Technologie (IWT) grant ZL356704 and Interne Fondsen KULeuven (IF) C32/15/027. Work in KDB and MDA’s laboratory is supported by UID/MULTI/04046/2013 centre grant (to BioISI) and research grants from FCT/MCTES Portugal (PTDC/BIM-MEC/2131/2014), Cystic Fibrosis Foundation USA (AMARAL15XX0 and AMARAL15XX1), Gilead GÉNESE-Portugal Programme (PGG/008/2015), and Cystic Fibrosis Trust UK (SRC 003).We thank Luka Clarke for revising this manuscript and to Els Aertgeerts for secretarial assistance.
10
References 1 Zolin A, McKone EF, van Rens J et al. ECFS Patient Registry Annual Data Report 2010. Karup: European Cystic Fibrosis Society, 2014. 2 Dodge JA, Lewis PA, Stanton M, Wilsher J. Cystic fibrosis mortality and survival in the UK: 1947–2003. Eur Respir J 2007; 29: 522–26. 3 Hurley MN, McKeever TM, Prayle AP, Fogarty AW, Smyth AR. Rate of improvement of CF life expectancy exceeds that of general population—observational death registration study. J Cyst Fibros 2014; 13: 410–15. 4 Riordan JR. CFTR function and prospects for therapy. Annu Rev Biochem 2008; 77: 701–26. 5 Shamsuddin AK, Quinton PM. Native small airways secrete bicarbonate. Am J Respir Cell Mol Biol 2014; 50: 796–804. 6 Amaral MD. Novel personalized therapies for cystic fibrosis: treating the basic defect in all patients. J Intern Med 2015; 277: 155–66. 7 Sosnay PR, Siklosi KR, Van Goor F, et al. Defining the disease liability of variants in the cystic fibrosis transmembrane conductance regulator gene. Nat Genet 2013; 45: 1160–67. 8 Cutting GR. Modifier genes in Mendelian disorders: the example of cystic fibrosis. Ann N Y Acad Sci 2010; 1214: 57–69. 9 Guillot L, Beucher J, Tabary O, Le Rouzic P, Clement A, Corvol H. Lung disease modifier genes in cystic fibrosis. Int J Biochem Cell Biol 2014; 52: 83–93. 10 Corvol H, Thompson KE, Tabary O, le Rouzic P, Guillot L. Translating the genetics of cystic fibrosis to personalized medicine. Transl Res 2016; 168: 40–49. 11 Amaral MD, Balch WE. Hallmarks of therapeutic management of the cystic fibrosis functional landscape. J Cyst Fibros 2015; 14: 687–99. 12 Schraufnagel DE, ed. Breathing in America: diseases, progress, and hope. New York: American Thoracic Society, 2010: 47–56. http://www.thoracic.org/patients/patient-resources/breathing-inamerica/resources/chapter-5-chronic-obstructive-pulmonarydisease.pdf (accessed Sept 29, 2015). 13 Johannesson B, Hirtz S, Schatterny J, Schultz C, Mall MA. CFTR regulates early pathogenesis of chronic obstructive lung disease in βENaC-overexpressing mice. PLoS One 2012; 7: e44059. 14 Bodas M, Tran I, Vij N. Therapeutic strategies to correct proteostasis-imbalance in chronic obstructive lung diseases. Curr Mol Med 2012; 12: 807–14. 15 Sandford A. The role of CFTR mutations in asthma. Can Respir J 2012; 19: 44–45. 16 Tzetis M, Efthymiadou A, Strofalis S, et al. CFTR gene mutations— including three novel nucleotide substitutions—and haplotype background in patients with asthma, disseminated bronchiectasis and chronic obstructive pulmonary disease. Hum Genet 2001; 108: 216–21. 17 Clunes LA, Davies CM, Coakley RD, et al. Cigarette smoke exposure induces CFTR internalization and insolubility, leading to airway surface liquid dehydration. FASEB J 2012; 26: 533–45. 18 Sloane PA, Shastry S, Wilhelm A, et al. A pharmacologic approach to acquired cystic fibrosis transmembrane conductance regulator dysfunction in smoking related lung disease. PLoS One 2012; 7: e39809. 19 Boyle MP, De Boeck K. A new era in the treatment of cystic fibrosis: correction of the underlying CFTR defect. Lancet Respir Med 2013; 1: 158–63. 20 Ramsey BW, Davies J, McElvaney NG, et al, and the VX08-770-102 Study Group. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med 2011; 365: 1663–72. 21 De Boeck K, Munck A, Walker S, et al. Efficacy and safety of ivacaftor in patients with cystic fibrosis and a non-G551D gating mutation. J Cyst Fibros 2014; 13: 674–80. 22 Sawicki GS, McKone E, Pasta DJ, Wagener J, Johnson C, Konstan MW. The effect of ivacaftor on the rate of lung function decline in CF patients with G551D-CFTR mutation. 37th European Cystic Fibrosis Conference; Gothenburg, Sweden; June 11–14, 2014. WS3.1 23 Amaral MD, Kunzelmann K. Molecular targeting of CFTR as a therapeutic approach to cystic fibrosis. Trends Pharmacol Sci 2007; 28: 334–41. 24 Smyth AR, Bell SC, Bojcin S, et al, and the European Cystic Fibrosis Society. European Cystic Fibrosis Society standards of care: best practice guidelines. J Cyst Fibros 2014; 13 (suppl 1): S23–42.
www.thelancet.com/respiratory Published online April 1, 2016 http://dx.doi.org/10.1016/S2213-2600(16)00023-0
Rapid Review
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
Button B, Cai LH, Ehre C, et al. A periciliary brush promotes the lung health by separating the mucus layer from airway epithelia. Science 2012; 337: 937–41. Elkins MR, Robinson M, Rose BR, et al, and the 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: 229–40. Bilton D, Daviskas E, Anderson SD, et al, and the B301 Investigators. Phase 3 randomized study of the efficacy and safety of inhaled dry powder mannitol for the symptomatic treatment of non-cystic fibrosis bronchiectasis. Chest 2013; 144: 215–25. Konstan MW, Wagener JS, Pasta DJ, et al, and the 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: 545–53. Cobos N, Danés I, Gartner S, González M, Liñán S, Arnau JM. DNase use in the daily care of cystic fibrosis: who benefits from it and to what extent? Results of a cohort study of 199 patients in 13 centres. DNase National Study Group. Eur J Pediatr 2000; 159: 176–81. Sanders NN, Franckx H, De Boeck K, Haustraete J, De Smedt SC, Demeester J. Role of magnesium in the failure of rhDNase therapy in patients with cystic fibrosis. Thorax 2006; 61: 962–66. Protalix BioTherapeutics Announces AIR DNase(TM) Data Presented at the 38th European Cystic Fibrosis Conference. Protalix Biotherapeutics. June 12, 2015. http://phx.corporate-ir.net/ phoenix.zhtml?c=101161&p=irol-newsArticle&ID=2058840 (accessed Sept 29, 2015). Bucki R, Cruz K, Pogoda K, et al. Enhancement of Pulmozyme activity in purulent sputum by combination with poly-aspartic acid or gelsolin. J Cyst Fibros 2015; 14: 587–93. Cantin AM, Hartl D, Konstan MW, Chmiel JF. Inflammation in cystic fibrosis lung disease: pathogenesis and therapy. J Cyst Fibros 2015; 14: 419–30. Cohen-Cymberknoh M, Kerem E, Ferkol T, Elizur A. Airway inflammation in cystic fibrosis: molecular mechanisms and clinical implications. Thorax 2013; 68: 1157–62. Govan JR, Deretic V. Microbial pathogenesis in cystic fibrosis: mucoid Pseudomonas aeruginosa and Burkholderia cepacia. Microbiol Rev 1996; 60: 539–74. Staudinger BJ, Muller JF, Halldórsson S, et al. Conditions associated with the cystic fibrosis defect promote chronic Pseudomonas aeruginosa infection. Am J Respir Crit Care Med 2014; 189: 812–24. Konstan MW, Döring G, Heltshe SL, et al, and the Investigators and Coordinators of BI Trial 543.45. A randomized double blind, placebo controlled phase 2 trial of BIIL 284 BS (an LTB4 receptor antagonist) for the treatment of lung disease in children and adults with cystic fibrosis. J Cyst Fibros 2014; 13: 148–55. Eigen H, Rosenstein BJ, FitzSimmons S, Schidlow DV, and the Cystic Fibrosis Foundation Prednisone Trial Group. A multicenter study of alternate-day prednisone therapy in patients with cystic fibrosis. J Pediatr 1995; 126: 515–23. Balfour-Lynn IM, Lees B, Hall P, et al, and the CF WISE (Withdrawal of Inhaled Steroids Evaluation) Investigators. Multicenter randomized controlled trial of withdrawal of inhaled corticosteroids in cystic fibrosis. Am J Respir Crit Care Med 2006; 173: 1356–62. Döring G, Bragonzi A, Paroni M, et al. BIIL 284 reduces neutrophil numbers but increases P aeruginosa bacteremia and inflammation in mouse lungs. J Cyst Fibros 2014; 13: 156–63. Konstan MW, Schluchter MD, Xue W, Davis PB. Clinical use of Ibuprofen is associated with slower FEV1 decline in children with cystic fibrosis. Am J Respir Crit Care Med 2007; 176: 1084–89. Konstan MW, VanDevanter DR, Rasouliyan L, et al, and the Scientific Advisory Group, and the Investigators and Coordinators of the Epidemiologic Study of Cystic Fibrosis. Trends in the use of routine therapies in cystic fibrosis: 1995–2005. Pediatr Pulmonol 2010; 45: 1167–72. Carlile GW, Robert R, Goepp J, et al. Ibuprofen rescues mutant cystic fibrosis transmembrane conductance regulator trafficking. J Cyst Fibros 2015; 14: 16–25. De Boeck K, Fajac I, Ratjen F. End-points and biomarkers for clinical trials in cystic fibrosis. Eur Respir Monogr 2014; 64: 104–115.
45
46
47
48
49 50
51
52
53
54
55 56 57 58
59
60
61
62
63
64
65
Springman E, Grosswald R, Philpot E, et al. A phase 1 clinical study of CTX-4430 in cystic fibrosis patients. 38th European Cystic Fibrosis Conference; Brussels, Belgium; June 10–13, 2015. 126. Corbus Pharma. http://www.corbuspharma.com/news/pressreleases/detail/199/corbus-pharmaceuticals-announces-fda-orphandrug (accessed March 25, 2016). Griese M, Latzin P, Kappler M, et al. alpha1-Antitrypsin inhalation reduces airway inflammation in cystic fibrosis patients. Eur Respir J 2007; 29: 240–50. Nichols D, Donaldson SH, Dorkin H, et al. A 3 week dose escalation, randomized, double-blind, placebo-controlled trial to assess the safety, tolerability, and possible efficacy of 100 mg or 200 mg of once daily inhaled alpha-1 HC in cystic fibrosis (CF). 28th Annual North American Cystic Fibrosis Conference; Atlanta, GA, USA; Oct 9–11, 2014. 281. Kogan I, Ramjeesingh M, Li C, et al. CFTR directly mediates nucleotide-regulated glutathione flux. EMBO J 2003; 22: 1981–89. Bishop C, Hudson VM, Hilton SC, Wilde C. A pilot study of the effect of inhaled buffered reduced glutathione on the clinical status of patients with cystic fibrosis. Chest 2005; 127: 308–17. Hartl D, Starosta V, Maier K, et al. Inhaled glutathione decreases PGE2 and increases lymphocytes in cystic fibrosis lungs. Free Radic Biol Med 2005; 39: 463–72. Griese M, Kappler M, Eismann C, et al, and the Glutathione Study Group. Inhalation treatment with glutathione in patients with cystic fibrosis. A randomized clinical trial. Am J Respir Crit Care Med 2013; 188: 83–89. Tirouvanziam R, Conrad CK, Bottiglieri T, Herzenberg LA, Moss RB, Herzenberg LA. High-dose oral N-acetylcysteine, a glutathione prodrug, modulates inflammation in cystic fibrosis. Proc Natl Acad Sci USA 2006; 103: 4628–33. Conrad C, Lymp J, Thompson V, et al. Long-term treatment with oral N-acetylcysteine: affects lung function but not sputum inflammation in cystic fibrosis subjects. A phase II randomized placebo-controlled trial. J Cyst Fibros 2015; 14: 219–27. Tiddens HA, Bos AC, Mouton JW, Devadason S, Janssens HM. Inhaled antibiotics: dry or wet? Eur Respir J 2014; 44: 1308–18. Agent P, Parrott H. Inhaled therapy in cystic fibrosis: agents, devices and regimens. Breathe (Sheff) 2015; 11: 110–18. Waters V, Smyth A. Cystic fibrosis microbiology: advances in antimicrobial therapy. J Cyst Fibros 2015; 14: 551–60. Halwani M, Balkhy HH, Khiyami MA, Omri A. Antimicrobial activity of liposomal β-glycan against Pseudomonas aeruginosa isolated from cystic fibrosis patients. 38th European Cystic Fibrosis Conference; Brussels, Belgium; June 10–13, 2015. 130. Pritchard MF, Ferguson LC, Powell E, et al. OligoG fluorescent conjugation for localisation in a mucoid pseudomonal biofilm. 38th European Cystic Fibrosis Conference; Brussels, Belgium; June 10–13, 2015. 139. Hodges L, MacGregor G, Stevens H, et al. An open label, randomised, two-way crossover scintigraphic study to investigate lung deposition of radiolabelled alginate oligosaccharide delivered as a dry powder and as a nebulised solution in cystic fibrosis patients. 28th Annual North American Cystic Fibrosis Conference; Atlanta, GA, USA; Oct 9–11, 2014. 251. Pritchard MF, Powell LC, Menzies GE, at al. A new class of safe oligosaccharide polymer therapy to modify the mucus barrier of chronic respiratory disease. Mol Pharm 2016; 13: 863–72. Devereux G, Steele S, Griffiths KJ, et al. An open label investigation of the tolerability and pharmacokinetics or oral cysteamine in adults with cystic fibrosis. 38th European Cystic Fibrosis Conference; Brussels, Belgium; June 10–13, 2015. A141. De Stefano D, Villella VR, Esposito S, et al. Restoration of CFTR function in patients with cystic fibrosis carrying the F508del-CFTR mutation. Autophagy 2014; 10: 2053–74. Luciani A, Villella VR, Esposito S, et al. Defective CFTR induces aggresome formation and lung inflammation in cystic fibrosis through ROS-mediated autophagy inhibition. Nat Cell Biol 2010; 12: 863–75. Grasemann H, Michler E, Wallot M, Ratjen F. Decreased concentration of exhaled nitric oxide (NO) in patients with cystic fibrosis. Pediatr Pulmonol 1997; 24: 173–77.
www.thelancet.com/respiratory Published online April 1, 2016 http://dx.doi.org/10.1016/S2213-2600(16)00023-0
11
Rapid Review
66
67
68
69
70
71
72
73 74 75
76 77 78
79
80
81
82
83
84 85
86
87
12
Meng QH, Springall DR, Bishop AE, et al. Lack of inducible nitric oxide synthase in bronchial epithelium: a possible mechanism of susceptibility to infection in cystic fibrosis. J Pathol 1998; 184: 323–31. Moss RB, Mayer-Hamblett N, Wagener J, et al. Randomized, double-blind, placebo-controlled, dose-escalating study of aerosolized interferon gamma-1b in patients with mild to moderate cystic fibrosis lung disease. Pediatr Pulmonol 2005; 39: 209–18. Miller CC, Hergott CA, Rohan M, Arsenault-Mehta K, Döring G, Mehta S. Inhaled nitric oxide decreases the bacterial load in a rat model of Pseudomonas aeruginosa pneumonia. J Cyst Fibros 2013; 12: 817–20. Blau H, Aviram M, Mussaffi H, et al. Safety and tolerability of nitric oxide given intermittently via inhalation to subjects with cystic fibrosis in a prospective open-label phase IIa trial. 28th Annual North American Cystic Fibrosis Conference; Atlanta, GA, USA; Oct 9–11, 2014. 226. Grasemann H, Tullis E, Ratjen F. A randomized controlled trial of inhaled L-arginine in patients with cystic fibrosis. J Cyst Fibros 2013; 12: 468–74. Hraiech S, Brégeon F, Rolain JM. Bacteriophage-based therapy in cystic fibrosis-associated Pseudomonas aeruginosa infections: rationale and current status. Drug Des Devel Ther 2015; 9: 3653–63. Sahota JS, Smith CM, Radhakrishnan P, et al. Bacteriophage delivery by nebulization and efficacy against phenotypically diverse Pseudomonas aeruginosa from cystic fibrosis patients. J Aerosol Med Pulm Drug Deliv 2015; 28: 353–60. Coburn B, Wang PW, Diaz Caballero J, et al. Lung microbiota across age and disease stage in cystic fibrosis. Sci Rep 2015; 5: 10241. Cystic fibrosis mutation database statistics. http://www.genet. sickkids.on.ca/StatisticsPage.html (accessed March 25, 2016). De Boeck K, Zolin A, Cuppens H, Olesen HV, Viviani L. The relative frequency of CFTR mutation classes in European patients with cystic fibrosis. J Cyst Fibros 2014; 13: 403–09. Welsh MJ, Smith AE. Molecular mechanisms of CFTR chloride channel dysfunction in cystic fibrosis. Cell 1993; 73: 1251–54. Zielenski J, Tsui LC. Cystic fibrosis: genotypic and phenotypic variations. Annu Rev Genet 1995; 29: 777–807. Bell SC, De Boeck K, Amaral MD. New pharmacological approaches for cystic fibrosis: promises, progress, pitfalls. Pharmacol Ther 2015; 145: 19–34. Rich DP, Anderson MP, Gregory RJ, et al. Expression of cystic fibrosis transmembrane conductance regulator corrects defective chloride channel regulation in cystic fibrosis airway epithelial cells. Nature 1990; 347: 358–63. 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: 827–34. Dörk T, Macek M Jr, Mekus F, et al. Characterization of a novel 21-kb deletion, CFTRdele2,3(21 kb), in the CFTR gene: a cystic fibrosis mutation of Slavic origin common in Central and East Europe. Hum Genet 2000; 106: 259–68. Moss RB, Flume PA, Elborn JS, et al, and the VX11-770-110 (KONDUCT) Study Group. Efficacy and safety of ivacaftor in patients with cystic fibrosis who have an Arg117His-CFTR mutation: a double-blind, randomised controlled trial. Lancet Respir Med 2015; 3: 524–33. Wainwright CE, Elborn JS, Ramsey BW, et al, and the TRAFFIC Study Group, and the TRANSPORT Study Group. Lumacaftor–ivacaftor in patients with cystic fibrosis homozygous for Phe508del CFTR. N Engl J Med 2015; 373: 220–31. Bosch B, De Boeck K. Searching for a cure for cystic fibrosis. A 25-year quest in a nutshell. Eur J Pediatr 2016; 175: 1–8. Davies JC, Wainwright CE, Canny GJ, et al, and the VX08-770-103 (ENVISION) Study Group. Efficacy and safety of ivacaftor in patients aged 6 to 11 years with cystic fibrosis with a G551D mutation. Am J Respir Crit Care Med 2013; 187: 1219–25. Hebestreit H, Sauer-Heilborn A, Fischer R, Käding M, Mainz JG. Effects of ivacaftor on severely ill patients with cystic fibrosis carrying a G551D mutation. J Cyst Fibros 2013; 12: 599–603. Sawicki GS, McKone EF, Pasta DJ, et al. Sustained benefit from ivacaftor demonstrated by combining clinical trial and cystic fibrosis patient registry data. Am J Respir Crit Care Med 2015; 192: 836–42.
88
89
90
91
92
93 94
95
96
97
98
99
100
101
102 103 104
105
106
107
108 109
110 111
Rowe SM, Heltshe SL, Gonska T, et al, and the GOAL Investigators of the Cystic Fibrosis Foundation Therapeutics Development Network. Clinical mechanism of the cystic fibrosis transmembrane conductance regulator potentiator ivacaftor in G551D-mediated cystic fibrosis. Am J Respir Crit Care Med 2014; 190: 175–84. Bellin MD, Laguna T, Leschyshyn J, et al. Insulin secretion improves in cystic fibrosis following ivacaftor correction of CFTR: a small pilot study. Pediatr Diabetes 2013; 14: 417–21. Borowitz D, Lubarsky B, Wilschanski M, et al. Nutritional status improved in cystic fibrosis patients with the G551D mutation after treatment with ivacaftor. Dig Dis Sci 2016; 61: 198–207. Van Goor F, Yu H, Burton B, Hoffman BJ. Effect of ivacaftor on CFTR forms with missense mutations associated with defects in protein processing or function. J Cyst Fibros 2014; 13: 29–36. Igreja S, Clarke LA, Botelho HM, Marques L, Amaral MD. Correction of a cystic fibrosis splicing mutation by antisense oligonucleotides. Hum Mutat 2016; 37: 209–15. Bush A, Simmonds NJ. Hot off the breath: ‘I’ve a cost for’— the 64 million dollar question. Thorax 2012; 67: 382–84. Whiting P, Al M, Burgers L, et al. Ivacaftor for the treatment of patients with cystic fibrosis and the G551D mutation: a systematic review and cost-effectiveness analysis. Health Technol Assess 2014; 18: 1–106. Flume PA, Liou TG, Borowitz DS, et al, and the VX 08-770-104 Study Group. Ivacaftor in subjects with cystic fibrosis who are homozygous for the F508del-CFTR mutation. Chest 2012; 142: 718–24. Okiyoneda T, Veit G, Dekkers F et al. Mechanism-based corrector combination restores ΔF508-CFTR folding and function. Nat Chem Biol 2013; 9: 444–54. Farinha CM, King-Underwood J, Sousa M, et al. Revertants, low temperature, and correctors reveal the mechanism of F508del-CFTR rescue by VX-809 and suggest multiple agents for full correction. Chem Biol 2013; 20: 943–55. Awatade NT, Uliyakina I, Farinha CM, et al. Measurements of functional responses in human primary lung cells as a basis for personalized therapy for cystic fibrosis. EBioMedicine 2014; 2: 147–53. PROQR THERAPEUTICS N.V. (PRQR) IPO. http://www.nasdaq. com/markets/ipos/company/proqr-therapeutics-nv-942327-76207 (accessed Sept 29, 2015). Kerem E, Konstan MW, De Boeck K, et al, and the 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: 539–47. Pezzulo AA, Tang XX, Hoegger MJ, et al. Reduced airway surface pH impairs bacterial killing in the porcine cystic fibrosis lung. Nature 2012; 487: 109–13. Boucher, RC. Cystic fibrosis: a disease of vulnerability to airway surface dehydration. Trends Mol Med 2007; 13: 231–40. Quinton PM. Cystic fibrosis: impaired bicarbonate secretion and mucoviscidosis. Lancet 2008; 372: 415–17. Chen EY, Yang N, Quinton PM, Chin WC. A new role for bicarbonate in mucus formation. Am J Physiol Lung Cell Mol Physiol 2010; 299: L542–49. Gustafsson JK, Ermund A, Ambort D, et al. Bicarbonate and functional CFTR channel are required for proper mucin secretion and link cystic fibrosis with its mucus phenotype. J Exp Med 2012; 209: 1263–72. Caputo A, Caci E, Ferrera L, et al. TMEM16A, a membrane protein associated with calcium-dependent chloride channel activity. Science 2008; 322: 590–94. Schroeder BC, Cheng T, Jan YN, Jan LY. Expression cloning of TMEM16A as a calcium-activated chloride channel subunit. Cell 2008; 134: 1019–29. Yang YD, Cho H, Koo JY, et al. TMEM16A confers receptor-activated calcium-dependent chloride conductance. Nature 2008; 455: 1210–15. Sondo E, Caci E, Galietta LJ. The TMEM16A chloride channel as an alternative therapeutic target in cystic fibrosis. Int J Biochem Cell Biol 2014; 52: 73–76. Mall MA, Galietta LJ. Targeting ion channels in cystic fibrosis. J Cyst Fibros 2015; 14: 561–70. Martins JR, Faria D, Kongsuphol P, Reisch B, Schreiber R, Kunzelmann K. Anoctamin 6 is an essential component of the outwardly rectifying chloride channel. Proc Natl Acad Sci USA 2011; 108: 18168–72.
www.thelancet.com/respiratory Published online April 1, 2016 http://dx.doi.org/10.1016/S2213-2600(16)00023-0
Rapid Review
112 Dorfman R, Taylor C, Lin F, et al, and the Members of Canadian Consortium for CF Genetic Studies. Modulatory effect of the SLC9A3 gene on susceptibility to infections and pulmonary function in children with cystic fibrosis. Pediatr Pulmonol 2011; 46: 385–92. 113 Sun L, Rommens JM, Corvol H, et al. Multiple apical plasma membrane constituents are associated with susceptibility to meconium ileus in individuals with cystic fibrosis. Nat Genet 2012; 44: 562–69. 114 Blackman SM, Commander CW, Watson C, et al. Genetic modifiers of cystic fibrosis-related diabetes. Diabetes 2013; 62: 3627–35. 115 Moss RB. Pitfalls of drug development: lessons learned from trials of denufosol in cystic fibrosis. J Pediatr 2013; 162: 676–80. 116 Strategic Research Centre 2. INOVCF: innovative non-CFTR approaches for cystic fibrosis therapy. London: Cystic Fibrosis Trust. http://www.cysticfibrosis.org.uk/research-care/research/aboutcystic-fibrosis-research/how-we-invest-in-research/strategicresearch-centres/src-2 (accessed Sept 29, 2015). 117 Knowles MR, Stutts MJ, Spock A, Fischer N, Gatzy JT, Boucher RC. Abnormal ion permeation through cystic fibrosis respiratory epithelium. Science 1983; 221: 1067–70. 118 Mall M, Bleich M, Greger R, Schreiber R, Kunzelmann K. The amiloride-inhibitable Na+ conductance is reduced by the cystic fibrosis transmembrane conductance regulator in normal but not in cystic fibrosis airways. J Clin Invest 1998; 102: 15–21. 119 Boucher RC. Regulation of airway surface liquid volume by human airway epithelia. Pflugers Arch 2003; 445: 495–98. 120 Mall M, Grubb BR, Harkema JR, O’Neal WK, Boucher RC. Increased airway epithelial Na+ absorption produces cystic fibrosis-like lung disease in mice. Nat Med 2004; 10: 487–93. 121 Snouwaert JN, Brigman KK, Latour AM, et al. An animal model for cystic fibrosis made by gene targeting. Science 1992; 257: 1083–88. 122 Pons G, Marchand MC, d’Athis P, et al, and the Amiloride-AFLM Collaborative Study Group. French multicenter randomized double-blind placebo-controlled trial on nebulized amiloride in cystic fibrosis patients. Pediatr Pulmonol 2000; 30: 25–31. 123 Bennett WD, Wu J, Fuller F, et al. Duration of action of hypertonic saline on mucociliary clearance in the normal lung. J Appl Physiol 2015; 118: 1483–90. 124 Almaça J, Faria D, Sousa M, et al. High-content siRNA screen reveals global ENaC regulators and potential cystic fibrosis therapy targets. Cell 2013; 154: 1390–400. 125 Althaus M, Clauss WG, Fronius M. Amiloride-sensitive sodium channels and pulmonary edema. Pulm Med 2011; 2011: 830320.
126 Alton EW, Armstrong DK, Ashby D, et al, and the UK Cystic Fibrosis Gene Therapy Consortium. Repeated nebulisation of non-viral CFTR gene therapy in patients with cystic fibrosis: a randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Respir Med 2015; 3: 684–91. 127 Schwank G, Koo BK, Sasselli V, et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell 2013; 13: 653–58. 128 Crane AM, Kramer P, Bui JH, et al. Targeted correction and restored function of the CFTR gene in cystic fibrosis induced pluripotent stem cells. Stem Cell Rev 2015; 4: 569–77. 129 Balch WE, Morimoto RI, Dillin A, Kelly JW. Adapting proteostasis for disease intervention. Science 2008; 319: 916–19. 130 Hutt DM, Herman D, Rodrigues AP, et al. Reduced histone deacetylase 7 activity restores function to misfolded CFTR in cystic fibrosis. Nat Chem Biol 2010; 6: 25–33. 131 Goubau C, Wilschanski M, Skalická V, et al. Phenotypic characterisation of patients with intermediate sweat chloride values: towards validation of the European diagnostic algorithm for cystic fibrosis. Thorax 2009; 64: 683–91. 132 Noone PG, Knowles MR. ‘CFTR-opathies’: disease phenotypes associated with cystic fibrosis transmembrane regulator gene mutations. Respir Res 2001; 2: 328–32. 133 De Boeck K, Wilschanski M, Castellani C, et al, and the Diagnostic Working Group. Cystic fibrosis: terminology and diagnostic algorithms. Thorax 2006; 61: 627–35. 134 Hirtz S, Gonska T, Seydewitz HH, et al. CFTR Cl– channel function in native human colon correlates with the genotype and phenotype in cystic fibrosis. Gastroenterology 2004; 127: 1085–95. 135 Sousa M, Servidoni MF, Vinagre AM, et al. Measurements of CFTR-mediated Cl– secretion in human rectal biopsies constitute a robust biomarker for cystic fibrosis diagnosis and prognosis. PLoS One 2012; 7: e47708. 136 Beekman JM, Sermet-Gaudelus I, de Boeck K, et al. CFTR functional measurements in human models for diagnosis, prognosis and personalized therapy: report on the pre-conference meeting to the 11th ECFS Basic Science Conference, Malta, 26–29 March 2014. J Cyst Fibros 2014; 13: 363–72. 137 Dekkers JF, Wiegerinck CL, de Jonge HR, et al. A functional CFTR assay using primary cystic fibrosis intestinal organoids. Nat Med 2013; 19: 939–45. 138 Chang EH, Zabner J. Precision genomic medicine in cystic fibrosis. Clin Transl Sci 2015; 8: 606–10.
www.thelancet.com/respiratory Published online April 1, 2016 http://dx.doi.org/10.1016/S2213-2600(16)00023-0
13