You are on page 1of 17

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/326527106

The epithelial sodium channel (ENaC) as a therapeutic target for cystic


fibrosis lung disease

Article  in  Expert Opinion on Therapeutic Targets · July 2018


DOI: 10.1080/14728222.2018.1501361

CITATIONS READS

11 1,459

2 authors, including:

Patrick Moore
University of Minnesota Twin Cities
12 PUBLICATIONS   73 CITATIONS   

SEE PROFILE

All content following this page was uploaded by Patrick Moore on 04 December 2018.

The user has requested enhancement of the downloaded file.


Expert Opinion on Therapeutic Targets

ISSN: 1472-8222 (Print) 1744-7631 (Online) Journal homepage: http://www.tandfonline.com/loi/iett20

The epithelial sodium channel (ENaC) as a


therapeutic target for cystic fibrosis lung disease

Patrick J. Moore & Robert Tarran

To cite this article: Patrick J. Moore & Robert Tarran (2018) The epithelial sodium channel (ENaC)
as a therapeutic target for cystic fibrosis lung disease, Expert Opinion on Therapeutic Targets, 22:8,
687-701, DOI: 10.1080/14728222.2018.1501361

To link to this article: https://doi.org/10.1080/14728222.2018.1501361

Accepted author version posted online: 20


Jul 2018.
Published online: 26 Jul 2018.

Submit your article to this journal

Article views: 221

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=iett20
EXPERT OPINION ON THERAPEUTIC TARGETS
2018, VOL. 22, NO. 8, 687–701
https://doi.org/10.1080/14728222.2018.1501361

REVIEW

The epithelial sodium channel (ENaC) as a therapeutic target for cystic fibrosis lung
disease
Patrick J. Moorea and Robert Tarrana,b
a
Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA; bDepartment of Cell Biology & Physiology, University of North Carolina,
Chapel Hill, NC, USA

ABSTRACT ARTICLE HISTORY


Introduction: Cystic fibrosis is an autosomal recessive disorder caused by mutations in the cystic Received 15 March 2018
fibrosis transmembrane conductance regulator (CFTR) gene that codes for the CFTR anion channel. In Accepted 13 July 2018
the absence of functional CFTR, the epithelial Na+ channel is also dysregulated. Airway surface liquid KEYWORDS
(ASL) hydration is maintained by a balance between epithelial sodium channel (ENaC)-led Na+ absorp- Cystic fibrosis; CFTR; ENaC;
tion and CFTR-dependent anion secretion. This finely tuned homeostatic mechanism is required to SPLUNC1; amiloride; channel
maintain sufficient airway hydration to permit the efficient mucus clearance necessary for a sterile lung activating protease;
environment. In CF airways, the lack of CFTR and increased ENaC activity lead to ASL/mucus dehydra- mucociliary clearance
tion that causes mucus obstruction, neutrophilic infiltration, and chronic bacterial infection. Rehydration
of ASL/mucus in CF airways can be achieved by inhibiting Na+ absorption with pharmacological
inhibitors of ENaC.
Areas covered: In this review, we discuss ENaC structure and function and its role in CF lung disease
and focus on ENaC inhibition as a potential therapeutic target to rehydrate CF mucus. We also discuss
the failure of the first generation of pharmacological inhibitors of ENaC and recent alternate strategies
to attenuate ENaC activity in the CF lung.
Expert opinion: ENaC is an attractive therapeutic target to rehydrate CF ASL that may serve as a
monotherapy or function in parallel with other treatments. Given the increased number of strategies
being employed to inhibit ENaC, this is an exciting and optimistic time to be in this field.

1. Introduction are born with normal lungs and pathology develops over time
[6]. CF lung disease is characterized by the accumulation of
1.1. CF lung disease
dehydrated mucus that serves as a reservoir for chronic bac-
Cystic fibrosis (CF) is a recessive genetic disease that is com- terial infections which result in unremitting cycles of chronic
mon in Caucasians. In this population, CF occurs in ~1:3000 infection and neutrophilia/inflammation [7,8]. Colonization of
live births and affects ~70,000 individuals worldwide, with a the CF lung occurs in the first years of life with pathogens
predicted survival age that is now close to 43 years [1]. CF is including Staphylococcus aureus and Haemophilus influenza,
caused by mutations in the cystic fibrosis transmembrane followed by the acquisition of Pseudomonas aeruginosa, typi-
regulator (CFTR) gene which codes for a cAMP/protein kinase cally by 5 years of age [9–11]. The dehydrated mucus present
A (PKA)-regulated apical membrane anion channel. CF-causing in the CF lung is thought to enhance P. aeruginosa pathogeni-
mutations lead to reduced or absent anion secretion across city through promotion of biofilm formation and antibiotic
affected epithelia, including the intestines, pancreas, repro- resistance [12]. Chemical factors such as high levels of reactive
ductive tracts, sweat glands, and lungs [2]. To date, over oxygen species, high iron levels, and lower pH may also con-
1700 CFTR mutations have been found that fall into 6 classes: tribute to persistent microbial infection [13–15]. Similarly, the
defective protein production mutations (Class 1), protein pro- dehydrated mucus is hypoxic which promotes P. aeruginosa
cessing mutations (Class 2), gating mutations (Class 3), con- biofilm formation [16]. Elevated levels of interleukin (IL)-8
duction mutations (Class 4), insufficient protein mutations [17,18], IL-6 [19], and tumor necrosis factor α [20–22], along
(Class 5), and accelerated degradation of CFTR (Class 6). The with reduced levels of anti-inflammatory cytokines, contribute
different classes of CFTR mutations are summarized in to necrosis of the lung [23]. Neutrophil elastase levels are also
Figure 1 [3]. elevated which can degrade host antimicrobial peptides such
Whilst some of the first manifestations of CF occur in the as defensins, LL-37, and short palate lung and nasal epithelial
pancreas, due to altered CFTR-dependent bicarbonate secre- clone 1 (SPLUNC1) which may further impede bacterial clear-
tion and a failure to secrete digestive enzymes, to date, lung ance [24,25].
disease accounts for the vast majority of CF morbidity [4]. CF Mucus rehydration is a desired therapeutic approach to
pancreatic disease occurs in utero [5]. However, CF patients prevent mucus plugging and airflow obstruction in the CF

CONTACT Robert Tarran robert_tarran@med.unc.edu Department of Cell Biology & Physiology, University of North Carolina, 7102 Marsico Hall, 125 Mason
Farm Road, Chapel Hill, NC 27599, USA
© 2018 Informa UK Limited, trading as Taylor & Francis Group
688 P. J. MOORE AND R. TARRAN

two membrane-spanning domains connected by a large extra-


Article highlights cellular domain and intracellular N- and C-termini. The extracel-
● The role of ENaC in CF lung disease.
lular, pre-M2 region near the second transmembrane domain of
● SPLUNC1 is an allosteric modulator of ENaC activity. α, β, and γ ENaC subunits affects the channel conductance and is
● Strategies to attenuate Na+ absorption in the CF lung. where amiloride binds [31] (Figure 2). The HG motif, so named
● Failure of first generation of ENaC antagonists.
● Targeting ENaC inhibition may alleviate CF symptoms.
because it possesses a glycine and a histidine in the amino
● Small molecules and peptides demonstrate promising results for terminus, is present in every ENaC/degenerin family member
inhibiting ENaC activity. known to conduct ions and is involved in channel opening [32].
This box summarizes key points contained in the article. Mutation of these residues in the β ENaC subunit causes pseu-
dohypoaldosteronism, a disease characterized by increased Na+
excretion (natriuresis) due to decreased renal ENaC activity and
increased mucus clearance rates due to decreased pulmonary
lung. Since mucus hydration is directly proportional to mucus ENaC activity [33,34]. According to the ASICs and ENaC structural
clearance rates [26], such an approach is predicted to increase models, the DEG gating is located within the highly conserved
mucus clearance and reduce mucus accumulation. TM2 region [30,35] and mutations of residues in the gating
Rehydrating the airway surface liquid (ASL) may be achieved region results in channel activation and subsequently an increase
by (1) direct addition of osmolytes (e.g. inhaled hypertonic in open probability (PO) [32,36]. αENaC was the first subunit to be
saline), (2) restoring CFTR function, (3) activating an alternate cloned and when αENaC is injected into Xenopus oocytes, ~2% of
Cl− secretory pathway, or (4) inhibiting the epithelial Na+ current is seen relative to α, β, γ ENaC co-expression. In contrast,
channel (ENaC). This review focuses on the role of ENaC in the β or γ subunits alone do not produce currents when
maintaining ASL hydration and summarizes the current ther- expressed alone. When the α, β or α, γ subunit combinations
apeutic approaches to modulate ENaC and Na+ absorption for are co-injected, up to 15% of αβγENaC activity occurs [37]. A
the treatment of patients with CF. fourth subunit, δENaC, has been shown to assemble with β and
γ-subunits and contributed to 50% of amiloride-sensitive salt
transport across primary human nasal epithelial cells [38,39].
1.2. ENaC structure However, its relevance to lower airways transepithelial Na+
absorption is not fully understood.
ENaC is expressed at the apical surface of epithelia of the lung,
colon, kidney, salivary ducts, and sweat ducts. Here, ENaC is the
rate limiting step for transepithelial Na+ absorption [27–29].
1.3. ENaC regulation
Homology studies using the crystal structure of the acid-sensing
ion channel (ASIC) have shown that ENaC is a heterotrimeric Like most plasma membrane ion channels, ENaC is regulated
transmembrane (TM) protein composed of α, β, and γ-subunits by either altering PO or by altering the number of channels at
that form a pore with high selectivity for Na+ and Li+ over K+ and the cell surface (N). ENaC gating kinetics are typically charac-
sensitivity to the small molecule antagonist amiloride [30]. Each terized by long opening and closing times. However, ENaC can
subunit is encoded by a different gene, i.e. SCNN1A (alpha), be posttranslationally modified to affect both channel density
SCNN1B (beta), and SCNN1G (gamma). These subunits contain (N) and PO. For example, three different classes of proteases

Figure 1. Different classes of CFTR mutations.


EXPERT OPINION ON THERAPEUTIC TARGETS 689

Figure 2. Structural features of the epithelial Na+ channel (ENaC). ENaC exists as a heterotrimer consisting of α, β, and γ subunits. Each subunit contains two
membrane spanning domains with intracellular N- and C-termini. The extracellular domains contain sites for proteolytic cleavage while the PY motif in the C-termini
is the site for ubquination.

have been shown to activate ENaC: (1) intracellular, conver- transition to a high PO state [42]. Prostasin, or CAP1, is a GPI-
tase-type proteases such as furin [40]; (2) extracellular, cell- anchored plasma membrane protein that was the first pro-
attached proteases, which can be membrane spanning or GPI- tease to be identified that could cleave ENaC, an affect that
anchored such as prostasin [41]; and (3) soluble/secreted pro- was reprised with trypsin [47]. Human transmembrane pro-
teases including trypsin and neutrophil elastase [42,43]. All of tease serine 4 (CAP2) and matriptase (CAP3) are also known to
these proteases cleave the extracellular domains of α- and γ- regulate ENaC [48]. However, in contrast to prostasin, both
ENaC to increase the channel open probability. Many of these CAP2 and CAP3 must possess catalytic activity to stimulate
proteases predominantly belong to a family of serine pro- ENaC [49]. Serine proteases can also cleave ENaC and have
teases which target mainly R and K residues on α- and been demonstrated to increase amiloride sensitive currents in
γ-subunits of ENaC [44]. That is, cleavage occurs at multiple Xenopus laevis oocytes and in airway epithelia [43,50]. Harris
sites, including at conserved basic residues located at α-K561, et al. observed that a brief exposure of ENaC expressed in
β-R503, and γ-R515. However R-138 on γ-ENaC is the only Xenopus oocytes to neutrophil elastase generated a new frag-
cleavage site that is required for activation [45]. ment of γ-ENaC in the surface pool, consistent with cleavage
ENaC is cleaved intracellularly by furin-type convertases downstream of the γ-ENaC furin site [51]. In addition, the
and can then be further cleaved by transmembrane or extra- specific residues V182 and V193 in human γ-ENaC were iden-
cellular channel activating proteases (CAPs) [40]. Furin, which tified as being essential for ENaC stimulation by neutrophil
is primarily present in the trans-Golgi networks, cleaves α and elastase [45].
γ-ENaC subunits through a consensus sequence R/S-XX-R As well as being cleaved by numerous proteases, ENaC can
where X is any amino acid. Cleavage of the α-subunit occurs also be regulated by intracellular second messengers. For
twice. A furin site is also present on γ-ENaC; however, a second example, the cAMP/PKA complex regulates ENaC by phos-
protease at a distal site to the furin site is required for ENaC phorylation and inhibition of NEDD4-2 [52]. Stutts et al. pre-
activation [40,46]. For reasons that are not fully understood, viously studied fibroblasts stably transduced with αβγ-ENaC
some ENaCs can bypass the furin cleavage steps and traffic to subunits ± CFTR. Here, they found that the presence/absence
the plasma membrane as ‘silent’ or ‘near-silent’ channels, of CFTR changed ENaC’s sensitivity to cAMP/PKA and whilst
which can then be fully activated by other proteases. cAMP/PKA inhibited ENaC in the presence of CFTR, they acti-
However, regardless of whether or not ENaC is cleaved intra- vated ENaC in CFTR’s absence [53,54]. Whilst the nature of this
cellularly or extracellularly, the end result of cleavage is the interaction is not fully understood, the intracellular domains of
690 P. J. MOORE AND R. TARRAN

CFTR were also shown to alter ENaC’s sensitivity to cAMP/ found that SPLUNC1 binding led to a NEDD4.2-dependent
PKA [55–57]. The interaction between CFTR and ENaC may ubiquitination of αENaC and internalization of channel subu-
also vary according to their location, and in the sweat glands, nits [76]. Thus, we propose that the information of SPLUNC1
where Cl− absorption rather than Cl− secretion occurs, ENaC is binding extracellularly was transmitted through ENaC, leading
activated in parallel with CFTR rather than being inactivated to a conformational change and increased accessibility of
[58], so more studies are required to fully understand this αENaC’s intracellular termini to NEDD4-2 and ubiquitination.
phenomenon. A reduction in the intracellular Cl− concentra- Indeed, co-expression of a concatemer where α–β–γ were
tion following stimulation of either CFTR or ClC Cl− channels linked N–C in order to constrain the channel abolished
has also been reported to lower ENaC activity [57,59,60]. SPLUNC1-induced ENaC internalization [76]. These data sug-
However, intracellular Cl− may not change so much during gest a previously undescribed way of regulating ENaC, where
absorption/secretion and the physiological relevance of ENaC serves as the receptor for SPLUNC1 that regulates chan-
this mode of action remains to be verified [61]. Stimulation nel density (N). Such findings lead us to postulate that
of P2Y2 receptors on the apical membrane of airway epithelial SPLUNC1’s ability to negatively regulate ENaC could be devel-
cells with purine nucleotides such as ATP activated oped as a potential therapeutic.
phospholipase C which in turn cleaves phosphatidylinositol
4,5-bisphosphate (PIP2) into diacyl glycerol and inositol 1,4,5-
1.5. The role of ENaC in CF lung disease
trisphosphate (IP3) [62]. The fall in PIP2 leads to a decrease in
ENaC PO [63,64]. Unlike cAMP regulation, PIP2 depletion inhi- Electrolyte transport across pulmonary epithelia, together with
bits ENaC activity in both normal lung and CF airways [65]. accompanying osmotic water flow, regulates ASL volume. In
Both PKA and PIP2 affect ENaC Po [66]. However, ENaC N is turn, ASL volume has been shown to directly affect mucus
also precisely regulated. ENaC can be ubiquitinated on all three rheology and mucus clearance rates [26]. Simply put, the more
subunits by the ubiquitin ligase neural precursor cell expressed fluid in the lung, the more quickly mucus is cleared. For
developmentally downregulated protein 4 (NEDD4-2) also example, patients with pseudohypoaldosteronism have
known as NEDD4L [67]. Each ENaC subunit has a PY motif (L/ reduced ENaC activity, increased airway secretions, and incred-
PPXY) that can bind to NEDD4-2 [68]. The subsequent ubiqui- ibly fast mucus clearance rates [77]. During normal mucus
tination of ENaC subunits leads to their rapid internalization clearance, ASL/mucus moves from the distal to the proximal
and degradation [34]. Butterworth et al. demonstrated that airways. However, with every successive generation, there is a
inhibition of deubiquitinating enzymes increased ubiquitination reduction in pulmonary surface area that leads to an abun-
and decreased surface αβγENaC [69]. Interestingly, deletions of dance of ASL. Thus, excess ASL needs to be absorbed in a
the PY motifs cause spontaneous upregulation of ENaC and controlled fashion, without causing dehydration, in order to
cause Liddle’s syndrome [66], which is characterized by maintain a constant ASL volume and prevent ASL pooling. In
increased renal ENaC activity, K+ excretion and Na+/H2O reten- normal airways, Cl− secretion via CFTR and Na+ absorption
tion that lead to hypertension and metabolic alkalosis [70]. In through ENaC are balanced to control ASL volume homeosta-
addition to the mutations observed in the PY motifs, sis and to achieve this goal. Glands can also secrete additional
mutations in the C-terminus (β, R563Q) and the TM2 segment ASL. However, submucosal glands are most commonly present
(γ, N530S) were also reported to be associated with Liddle’s in large airways (i.e. the first 16–17 generations) and are less
syndrome [71,72]. Although these mutations are not associated common/absent in successive generations [78,79]. In contrast,
with CF-like disease, mutations in individual ENaC subunits Na+ absorption has been detected from the nose and trachea,
would likely influence the impact of posttranslational proteoly- to the alveolar surfaces, which highlights its importance in
tic cleavage of this subunit. lung fluid homeostasis [80,81].
In CF airways, the lack of functional CFTR and the subse-
quent increase in ENaC activity result in Cl− hyposecretion and
1.4. SPLUNC1 and ENaC
Na+ hyperabsorption, respectively, which combine to dehy-
SPLUNC1 is a ~25-kDa secreted protein that is highly abun- drate airway surfaces (Figure 3). The importance of ENaC in
dant in the airways and plays an important role in innate the in vivo pathogenesis of CF lung disease was first demon-
defense [73]. Using its N-terminal ‘S18’-region, SPLUNC1 strated by Knowles, Boucher, and colleagues using electrophy-
binds to and regulates ENaC [74]. Indeed, stable knockdown siological techniques. Indeed, they demonstrated that
of SPLUNC1 increases the amiloride-sensitive transepithelial the amiloride-sensitive voltage was upregulated in CF air-
voltage and causes CF-like ASL dehydration in normal ways [82,83]. More recently, it has been hypothesized that
human bronchial epithelial cultures (HBECs), indicating that these observations were an artifact of the nasal potential
SPLUNC1 is required to limit ENaC activity and prevent ASL difference electrophysiological technique [84]. That is, when
dehydration [75]. SPLUNC1 binds extracellularly to ENaC and measured under open circuit conditions, amiloride could
leads to internalization of the channel. More recently, cause apical membrane hyperpolarization that generates an
SPLUNC1’s mechanism of action has become clear: αENaC- electrical gradient for Cl− secretion via basally active CFTR in
GFP and βENaC-mCherry, where the subunits were labeled normal but not CF tissues, which could have accounted for the
on their intracellular termini, along with unlabeled γENaC difference in amiloride sensitivity between normal and CF
were co-expressed in HEK293T cells. Using these constructs, subjects. However, the presence of CFTR leads to diminished
we observed that extracellular SPLUNC1 binding altered intra- ENaC activity in cell lines, as measured by the patch clamp
cellular ENaC FRET, thus demonstrating allostery [76]. We also technique [53], which would not be subject to this potential
EXPERT OPINION ON THERAPEUTIC TARGETS 691

Figure 3. Ion transport in the cystic fibrosis airway. In normal airways (A), the ASL is hydrated by a combination of Cl− secretion and Na+ absorption and in CF
airways (B), reduced Cl− secretion and Na+ hyperabsorption causes ASL dehydration that favor mucostasis.

artifact since the cells are voltage clamped to control the inhibitory functions would be beneficial for rehydrating CF
electrical gradient and the cells are studied with intracellular lungs [86]. Intracellular SPLUNC1 may be upregulated in CF
and extracellular solutions that inhibit anion channels. airways [91]. However, data suggest that SPLUNC1 is degraded
Moreover, the differences in amiloride-sensitivity were also by neutrophil elastase over time [92,93]. Similarly, we detected
observed in primary normal versus CF HBECs under thin film reduced coverage of SPLUNC1 using proteomic approaches in
conditions (i.e. with native ASL), even when the cultures were CF versus normal sputum, suggesting that SPLUNC1 may be
pretreated with bumetanide to block Cl− secretion, which also part of the normal lung’s ENaC regulatory mechanism that is
removed this potential artifact [85,86]. Furthermore, studies of absent in CF airways [74].
ASL volume homeostasis on primary airway cultures revealed Whether or not CFTR directly interacts with and/or nega-
ASL volume hyperabsorption in CF compared to normal air- tively regulates ENaC is still under debate. However, regard-
ways that was ENaC-led [26,85,87,88]. Importantly, the ASL less of the regulatory role that CFTR may exert on ENaC,
hyperabsorption was coupled with a decrease in mucus trans- other extrinsic factors can also enhance ENaC activity in CF
port rates – thus linking defective ENaC with the CF clinical airways. For example, both neutrophil elastase and cathe-
manifestation of mucus dehydration/mucus stasis. psin B can cleave and activate ENaC independently of func-
We have previously demonstrated that CF HBECs failed to tional CFTR [42,94]. Furthermore, proteases released by
regulate ENaC over time [85]. Similar results were found by Gram-negative bacteria also cleave and activate ENaC.
researchers at the University of Pittsburgh [89] and this group Indeed, alkaline protease and metalloprotease from
termed the phrase ‘ASL volume expansion,’ i.e. if you flood Pseudomonas species resulted in cleavage and activation
HBECs and wash away native ASL and its soluble/endogenous of ENaC in human bronchial epithelial cells [95,96].
ENaC inhibitors such as SPLUNC1, then ENaC activity increases. Finally, a number of mutations that lead to increased ENaC
However, addition of the protease inhibitor aprotinin reduced activity have been found in patients with atypical CF which are
CF ENaC activity, suggesting that a failure of regulation had summarized below: Sheridan et al. identified 20 patients out of
occurred in CF airway epithelia and that ENaC was ‘fixable’ by a cohort of 185 who had elevated sweat chloride concentra-
restoring key components in the extracellular regulatory path- tions and pulmonary disease without CFTR mutations. It was
way [85,90]. Subsequently, we identified dis-regulation of revealed that two patients contained heterozygous mutations
SPLUNC1 as being the cause of this increased ENaC activity. in SCNN1B: one patient exhibited a missense mutation (P26L)
Indeed, our data indicated that recombinant SPLUNC1 was while the other patient had a splice mutation and two mis-
capable of inhibiting ENaC in normal but not CF HBECs [86]. sense mutations (G294S and E539K). Both P26L and E539K
After solving SPLUNC1’s crystal structure, we identified two mutants demonstrated decreased activity while G294S muta-
pH-sensitive salt bridges (R at position 151-D 193 and K 138-D tion increased activity [97]. In a multicenter European study,
112) on the main body of SPLUNC1 that prevented SPLUNC1 30 ENaC mutations were found in 76 patients with atypical CF.
from binding to ENaC in CF HBECs at even moderately acidic In two of the patients sampled, the CF-like disease could be
pH (<7.0) [86]. Ablation of any of these four residues rendered explained by mutations in SCNN1A (F61L and V114I) that lead
SPLUNC1 capable of binding to ENaC and regulating ASL to an alteration in function [98]. Furthermore, an additional
height in CF HBECs, indicating that restoring SPLUNC1’s mutation (W293R) in SCNN1A demonstrated a fourfold
692 P. J. MOORE AND R. TARRAN

increase in amiloride-sensitive currents compared to wild type Thus, whilst amiloride is an effective diuretic, it has now
ENaC when expressed in X. laevis oocytes [98]. Similarly, a been superseded by so called ‘loop diuretics’ such as thia-
mutation in the β-ENaC subunit, V348M, was identified zide that target the Na+/K+/2 Cl− cotransporter (NKCC2) in
which increased ENaC PO by destabilizing the closed channel the loop of Henle to block both Na+ and K+ reabsorption
state [99]. Another study of atypical CF phenotypes detected which does not cause hyperkalemia [113].
three missense variants in SCNN1A (R204W, A357T, C641F) and Because of its affinity for ENaC, amiloride was tested as an
one missense in SCNN1B (R543Q). The authors of this study inhaled CF therapeutic that could reduce Na+ absorption in the
suggested that these variants that appear at a higher inci- airways to rehydrate mucus and restore mucus clearance [87,114].
dence in patients with atypical CF may be responsible for When added 2–4 times per day, amiloride had a moderate effect
the observed CF-like symptoms [100]. More recently, a muta- on MCC and lung function in CF patients [111,115,116]. The lung
tion in the δ subunit reduced ENaC activity indicating δENaC expresses pumps and transporters that can remove xenobiotics
as a potential genetic CF disease modifier [101]. SPLUNC1 has from the lung and concentrate them in the bloodstream, so that
also been shown to be a potential gene modifier and CF they can be metabolized by the liver and/or excreted by the
patients who have a polymorphism on the G allele of kidneys [117,118]. Amiloride is a substrate for some of these
rs1078761 had decreased SPLUNC1 expression and increased transporters and was found to have a half-life in humans of
disease severity [102]. Taken together, these data suggest that ~40 min, and a half-life in the ASL of human airway cultures of
increased ENaC activity and/or a failure of SPLUNC1 to regu- ~10 min [116,119]. This short half-life likely accounted for its failure
late ENaC might contribute to the development of CF to achieve significant clinical efficacy in the lung. Indeed, the
lung disease. increase in mucus clearance lasted ~40 min [111]. Amiloride was
also studied in βENaC overexpressing mice: Mall and colleagues
reported that intranasal administration of amiloride to these mice,
1.6. Mouse models of CF-like lung disease 3 times daily from the first day of life resulted in the reduction of
Mice have fewer generations of airways than humans (13 versus pulmonary mortality and significantly reduced in mucus accumu-
21, respectively) and CFTR−/− mice do not have a lower airways lation/obstruction, goblet cell hyperplasia, and pulmonary inflam-
disease phenotype [78]. However, there is a dehydration phe- mation [120]. These data indicated that if the poor
notype in the nasal tissues of these mice [103,104]. pharmacodynamics can be overcome, then ENaC inhibition via
Interestingly, murine nasal epithelia are the only part of the inhalation could be a viable therapy for the treatment of CF [120].
mouse airways that show elevated Na+ transport [105]. Since CF Amiloride derivatives have been developed in an attempt
mice lacked any obvious lower airway pathology, Mall et al. to improve the efficacy and potency seen with amiloride.
overexpressed β-ENaC in a transgenic mouse, which caused Parion Sciences have developed several amiloride-derivatives.
spontaneous lung disease [106]. Furthermore, this lung disease Parion-552-002 (P-552-02) is an ENaC antagonist that
approximated the development of the CF lung, including was more efficacious and more selective for ENaC than amilor-
mucus plugging, neutrophilia, and infection, leading to ide [119]. However, P-552-02 studies were halted after Phase II
increased mortality rates [106]. Knockout of the ubiquitin ligase clinical trials when hyperkalemia was observed. Parion
NEDD4.2 also caused an increase in ENaC surface densities and Sciences P-680 (Gilead Sciences, GS-9411) was a subsequent
spontaneous lung disease that was similar to that seen in the amiloride derivative that was 100-fold more potent than
βENaC mouse [67]. The development of the β-ENaC overexpres- amiloride that also had a longer duration of action. P-680/
sing and NEDD4.2 knockout transgenic mice clearly demon- GS-9411 was retained in the ASL for up to 8 h and also
strate a link between unregulated ENaC activity and the increased ASL volume in vitro. However, Phase I clinical trials
development of pulmonary disease. Importantly, given the failed, after administration of GS-9411 to healthy subjects also
aforementioned lack of phenotype of the CF mice, mice with induced hyperkalemia [112]. Parion sciences subsequently
altered ENaC activity have served as an important test bed for developed a long-acting ENaC antagonist called P-1037.
new therapies aimed at rehydrating CF lungs. Preclinical studies demonstrated promising results when P-
1037 was used in combination with hypertonic saline and
P-1037 produced a sustained increase in ASL height over 8 h
2. ENaC inhibitors in vitro, resulting in an eight- and fourfold increase in
volume compared to isotonic saline and hypertonic saline,
2.1. Small molecule/pore blocking ENaC antagonists
respectively [121]. In collaboration with Vertex
Amiloride is a first-generation ENaC antagonist that was Pharmaceuticals, P-1037 (now VX-371) entered Phase II clinical
developed in the 1960s as a potassium-sparing compound trials. However, following a 28-day trail, VX-371 and hypertonic
by Cragoe and colleagues at Merck Pharmaceuticals [107]. saline caused a nonsignificant increase in FEV1 of 0.1% when
ENaC is highly expressed in the distal convoluted tubule of added along with Orkambi (VX-770/VX-809) in ΔF508 homo-
the kidney and while this portion of the nephron is only zygous patients (https://www.businesswire.com/news/home/
responsible for handling 10% of reabsorbed Na+, it plays an 20171025006319/en/). This lack of effect may have been due
important role in fine-tuning Na+ and fluid levels in the to a short duration of action and/or poor bioavailability in vivo.
body [108,109]. Amiloride was initially used to prevent AstraZeneca developed ‘compound A’ which is a small
Na+ absorption in the kidney and to induce natriuresis/ molecule ENaC antagonist that may have been amiloride-
diuresis[110]. However, inhibition of ENaC leads to potas- derived [122]. Whilst this compound was more potent than
sium-sparing excretion of Na+ and hyperkalemia [111,112]. amiloride, was capable of increasing ASL height, and could
EXPERT OPINION ON THERAPEUTIC TARGETS 693

restore mucus clearance, it also showed hyperkalemia in ani- were pulmonary exacerbations after camostat treatment and
mal models and was not taken into the clinic [122]. A second one subject had appendicitis following placebo treatment. In
ENaC inhibitor, AZD5634, was subsequently developed. This addition, six subjects had adverse events which were thought
compound was retained in the ASL and had a high affinity for to be drug related including epistaxis, nasal disruption, and
ENaC [123]. In primary ΔF508 homozygous HBECs, AZD5634 hematuria. Nasal sensitivity and rhinorrhea were also reported.
elicited greater changes in ASL height and mucociliary clear- ENaC is highly promiscuous and is cleaved by multiple pro-
ance than benzamil, a second generation amiloride-derivative. teases. Thus, a possible contribution to the failure of protease
AZD5634 completed Phase I clinical trials and it was generally inhibitors to induce clinically meaningful changes in lung
well tolerated with no obvious hyperkalemia [124]. To date, a function in the clinic is that they tend to be relatively specific
Phase II study in CF patients using MCC as the readout is and inhibit only one protease or one class of protease per
ongoing. Novartis developed NVP-QBE170, a dimeric-amiloride drug. In contrast, multiple proteases are upregulated in the CF
derivative that has shown some promising results in lung and for example if neutrophil elastase is inhibited, cathe-
sheep [125]. For example, NVP-QBE170 was able to enhance psin B or another protease can still independently cleave and
mucus clearance in a dose dependent manner without indu- activate ENaC.
cing hyperkalemia [125]. Similarly, Novartis and Boehringer
Ingelheim have also developed the novel ENaC antagonists,
2.3. Peptides and other approaches
QBW 276, and BI 443651, respectively. Both have entered
clinical testing and showed good safety profiles following Whilst the idea of using small molecules and protease inhibi-
Phase I clinical trials. However, Phase II/efficacy results are tors to inhibit ENaC has been around for a while, more
still pending. recently alternate approaches to regulate ENaC are now
being evaluated. For example, the ability to specifically inhibit
the individual ENaC subunits is possible through the use of
2.2. Protease inhibitors
antisense oligonucleotides (ASO). ASO are strands of nucleic
In healthy airways, a balance exists between the number of acid-based complementary messenger RNA (mRNA). There are
proteases and their inhibitors. Whilst these proteases have two classes of ASO: (1) the RNase-H-dependent oligonucleo-
many innate defense functions, they also affect ENaC activity tides which induce degradation of mRNA and (2) the steric-
and hence can influence ASL hydration [42]. In CF airways, an blocker oligonucleotides which physically inhibit the splicing
imbalance between protease/protease inhibitor levels results translational machinery. Most of the currently available ASOs
may cause increased proteolysis of α- and γ-ENaC subunits function via the RNase-H-dependent mechanism [133].
that may contribute to ASL/mucus dehydration. Levels of furin, Generally, uptake of the ASO through active transport
cathepsin B, neutrophil elastase, and prostasin are all depends on the structure and concentration of the oligonu-
increased in CF airways [42,126–128]. Thus, inhibition of CF cleotide [134,135]. Recently, an ASO complimentary to SCNN1A
proteases represents an alternate approach to limit ENaC was administrated directly into a Nedd4L knockout mouse
hyperactivity in the CF lung that is currently being evaluated. model, which resulted in a 70% mRNA target knockdown
A novel protease inhibitor, QUB-TL1, was developed by the and an overall improvement in biomarkers of disease [136].
Martin group at Queen’s University in Belfast. This compound ASOs are currently in preclinical development with Ionis
reduced mucosal protease activity on CF HBECs [129]. QUB- Pharmaceuticals. However, consistent issues with gene-based
TL1-depdendent inhibition of protastin, furin, and P. aerugi- approaches to treating CF lung disease are poor delivery and
nosa exotoxin A resulted in reduced ENaC-mediated Na+ local inflammation [137,138]. Thus, whether or not ASOs will
absorption in CF HBECs that lead to decreased amiloride- be able to achieve therapeutic doses in order to decrease
sensitive currents and increased ASL volume/mucociliary clear- ENaC levels without inducing inflammation in CF patients
ance [129]. Whilst QUB-TL1 is a broad spectrum protease remains to be determined.
inhibitor that looks promising in preclinical development, it The discovery of an ENaC-inhibitory domain on SPLUNC1’s
has relatively low potency and this program may benefit from N-terminus has led to the development of novel ENaC antago-
a second generation compound with improved efficacy. Other nists that are peptides rather than small molecules. Despite
protease inhibitors have also been tested in CF patients. For having an ENaC inhibitory domain, SPLUNC1 fails to regulate
example, AZD9668 was tested and had no effect on inflam- ENaC in CF airway cultures [86]. An explanation for this was
mation or lung function in CF patients [130]. Similarly, found in SPLUNC1’s crystal structure. Indeed, SPLUNC1 possess
recombinant α1 antitrypsin also failed to affect lung inflam- pH-sensitive salt bridges that prevent SPLUNC1 from binding
mation [131]. Camostat, an inhibitor of protastin, induced a to and regulating ENaC in the mildly acidic CF ASL environ-
potent and prolonged attenuation of ENaC in the guinea pig ment [86]. In contrast, unlike full-length SPLUNC1, SPLUNC1-
tracheal potential difference (PD) model and enhanced mucus derived peptides that correspond to the N-terminal ENaC
clearance for 5 h after nebulization into sheep [125]. Using the inhibitory domain lack the salt bridges and can regulate ASL
nasal potential difference assay, it was reported that the hydration equally well in normal and CF HBECs [139]. However,
amiloride-sensitive PD was significantly reduced in CF patients perceived issues with inhaled peptides for the treatment of CF
treated with camostat compared to placebo, indicating that a lung disease include the possibility that they will stick to mucus
reduction in Na+ transport in CF airways can be achieved and fail to reach their target and/or that they will degrade by
though protease inhibition [132]. Three subjects experienced lung proteases. We have previously demonstrated that the
serious adverse events during the clinical trial; two of which SPLUNC1-derived S18 peptide can be mixed with neutrophil
694 P. J. MOORE AND R. TARRAN

elastase and/or activated neutrophil supernatant that contains hyperkalemia. Thus, SPX-101 shows promise for improving
neutrophil elastase and cathepsins and it still functions to inhibit lung function by inhibiting ENaC since it is active in CF
ENaC [74], suggesting that the stability issue can be overcome. patients and has good pharmacodynamics and a favorable
Indeed, due to the long duration of action induced by the safety profile. We speculate that CF subjects with higher base-
removal of ENaC subunits from the plasma membrane after line FEV1 may have more mucus obstruction and less bronch-
exposure to SPLUNC1-derived peptides, it is likely that the effects iectasis and are thus more amenable to mucus rehydration
persist over time even after washout. Using biophysical techni- therapies. Importantly, these data suggest that SPX-101 may
ques, we recently demonstrated that the S18 ENaC inhibitory be more beneficial in these milder CF patients. Furthermore, it
peptide does not bind to mucins and that fluorescently labeled was demonstrated that SPX-101 also induces ASL rehydration
S18 was able to easily diffuse through dehydrated mucus that in COPD-derived/tobacco smoke-exposed HBECs, suggesting
had accumulated on CF HBECs [139]. This is consistent with that it SPX-101 may also be efficacious in COPD patients who
previous observations that positively charged residues bind to/ also exhibit defective CFTR function and mucus dehydration
interact with mucins [140] and S18 (GGLPVPLDQTLPLNVNPA) [144,145].
does not contain positive K, R, or H residues. Additionally, using
this peptide, we were able to reprise the experiments of Zhou
et al. and significantly increase survival of newly born βENaC
2.4. ENaC inhibition and other therapies
mice. Interestingly, while amiloride caused a ~10% loss of body
weight/day, S18 did not have these effects [139]. Further inves- CF sputum contains large amounts of extracellular DNA that is
tigations revealed that S18 did not affect renal function when released by infiltrating inflammatory cells such as neutrophils.
injected intraperitoneally to mice and did not affect renal func- Nebulization of recombinant human deoxyribonuclease
tion when added intravenously to rats. Furthermore, when (rhDNase) is a commonly used method to mobilize CF sputum
100 mM of S18 was added intranasally to mice, only ~1 nM [146]. Dornase alfa (Pulmozyme; Genentech) is a purified
peptide was detected in serum, suggesting that SPLUNC1- rhDNase that cleaves extracellular DNA, thereby reducing the
derived peptides are poorly absorbed across pulmonary epithelia viscosity of the mucus layer [146,147]. Daily treatment with
and have very favorable pharmacodynamics [139]. rhDNase reduces the number of pulmonary exacerbations,
SPX-101 is a novel, size-optimized SPLUNC1-derived pep- improves lung function, and is well tolerated [148,149]. Use of
tide/ENaC inhibitor that is in clinical development for the Dornase alfa would not affect ASL hydration or ENaC activity
treatment of CF lung disease. SPX-101 is more potent than directly but would likely be complementary to an inhaled ENaC
SPLUNC1/S18 and bound to ENaC to promote subunit inter- antagonist, since the latter would likely facilitate mucus hydra-
nalization in both normal and CF HBECs, leading to a signifi- tion/clearance. Clinical trials have demonstrated that rhDNase
cant reduction in the amiloride-sensitive current and an significantly improves lung function [147,150]. However, varia-
increase in ASL height/mucus clearance [141]. A once daily tions in clinical response to rhDNase have been observed in CF
installation of SPX-101 increased survival and reduced airway patients [151,152] which have been attributed to treatment
inflammation in βENaC mice [141]. The Abraham lab in Miami regimens and optimal timing [153,154].
recently developed a pharmacological CF ovine mucus clear- Increasing ASL hydration by influencing the osmotic gradi-
ance model. Here, inhalation of the CFTR antagonist INH172 ent is another attractive therapeutic target. Mannitol is a
slows mucus clearance rates by ~50%. Using this model, SPX- naturally occurring sugar alcohol which has been inhaled by
101 increased tracheal mucus velocity for extended periods, CF patients [155]. Similarly, hypertonic saline has also been
consistent with the long duration of action seen in vitro and in used as an osmolyte [156]. When inhaled, these compounds
the βENaC mice [141]. These findings, coupled with its unique create an osmotic gradient that induces water movement into
mechanism of action, indicate that SPX-101 is an attractive the airway lumen, thus rehydrating the ASL and enhancing
therapeutic peptide to reduce ENaC activity in the CF lung. A mucus clearance [157]. Two randomized multicenter, double-
major advantage of using intrinsically disordered peptides like blind, controlled, parallel-group Phase III studies were carried
SPX-101 is that due to their inherent flexibility, they achieve out to assess the safety and efficacy of inhaled mannitol in CF
good contact with their target protein, thus maximizing their patients aged 6 years over a period of 6 months. Bilton et al.
binding efficiency. In preclinical toxicology, SPX-101 was admi- and Aitken et al. observed a statistically significant improve-
nistered by inhalation to two species (rats and dog), was safe, ment in FEV1 within 6 weeks of treatment with mannitol was
and did not alter serum electrolyte levels [142]. Similarly, SPX- maintained throughout the study [158,159]. Inhaled mannitol
101 did not cause hyperkalemia in humans following 14-day has since been approved for use in adults with CF in the
administration in a Phase I study [143]. To date, SPX-101 has European Union and in children over 6 years of age in
entered Phase II clinical trials in CF patients (NCT03229252). Australia. Phase III trials of Bronchitol (mannitol, Pharmaxis
Cohort 1 of this trial has now been completed and these data Ltd.) in adults in the United States has reached its primary
were recently presented by Professor Isabelle Fajac at the end point and FDA submission is expected in 2018. The usage
European CF Conference. Indeed, in CF patients with a percent of rhDNase and mannitol have been well documented in
predicted FEV1 (ppFEV1) of 40–80%, SPX-101 increased ppFEV1 enhancing the CF patients' quality of life. ENaC antagonists
by 5.2% in a mutation-agnostic fashion relative to placebo. have been shown to be synergistic with hypertonic saline in
However, in patients with an FEV1 greater than 55%, SPX-101 vitro [160]. However, amiloride-pretreatment before hyper-
significantly increased ppFEV1 by 8.3% without causing tonic saline resulted in a reduction in FEV1 [156], and whether
EXPERT OPINION ON THERAPEUTIC TARGETS 695

osmolytes and ENaC agonists will work synergistically remains the ASL, ENaC needs to be inhibited, which, due to the lack
to be determined and may depend on the ENaC antagonist of Na+ absorption, causes apical membrane hyperpolariza-
used and the order of administration. tion [170]. This increased negative potential causes an electrical
driving force to induce Cl− secretion against the unfavorable
concentration gradient. This is why experimentally, amiloride is
2.5. ENaC inhibition therapy: with or without CFTR
added in Ussing chambers before activating CFTR, even in the
modulators?
presence of Vertex-type CFTR modulators [162,163]. In the clinic,
Whilst some epithelia can only secrete or only absorb fluid, therefore, ENaC inhibition will increase the electrical driving force
airway epithelia are capable of both modalities. This is espe- for Cl− secretion, even for wild-type or fully corrected CFTR.
cially relevant to airways hydration since net ASL/fluid secre- Whilst CFTR potentiators and modulators would likely be
tion is the sum of Cl− secretion minus opposing Na+ synergistic with ENaC antagonists, approximately 7% of CF
absorption. That is, any given Cl− secretory event will be larger patients have Type I mutations where CFTR protein is not
if absorption is blocked. For example, we have previously made [171] (i.e. W1282X), which will never be amenable to
applied phasic shear stress to normal and CF HBECs in order corrector/potentiator-type therapies. In contrast, ENaC inhibi-
to mimic tidal breathing, which causes an increase in ATP tion is mutation agnostic and should be equally effective even
release, subsequent adenosine formation, and activation of in the complete absence of CFTR protein (i.e. for Type I muta-
Cl− secretion via purinergic receptors [161]. Here, we found tions). Thus, whilst read-through therapies for Type I muta-
that phasic shear stress elicited similar amounts of bumeta- tions are being developed [172], ENaC antagonism may be the
nide-sensitive transepithelial voltage, indicative of Cl− secre- earliest chance at normalizing ASL volume homeostasis in
tion, in both normal and CF HBECs. However, whilst the these patients.
amiloride-sensitive voltage (a marker of ENaC activity) was
spontaneously abolished (a reduction of >90%) in normal
HBECs, it was only reduced by ~50% in CF HBECs. In parallel, 3. Conclusion
we observed an increase in ASL height to ~15 μm in normal
Therapeutics for the treatment of CF lung disease have been
HBECs, but only to ~7 μm in CF HBECs, suggesting that the
focused on symptomatic management of the existing pulmon-
persistent ENaC activity was hampering CF ASL secretion [161].
ary disease. However, in recent years, there has been a shift
These observations highlight the importance of homeostasis
with to an emphasis on pharmacotherapy for ENaC inhibition
and indicate how balancing multiple facets of airway ion
and CFTR correction that can both contribute to ASL rehydra-
transport can favor Cl− secretion.
tion that may prevent lung disease from occurring. The
Recent attention has justifiably been focused on the pharma-
advances in ENaC therapeutics, especially with new and emer-
cological rescue of mutant CFTR using novel potentiators [162]
ging therapeutics such as SPX-101, make it clear that the
that increase CFTR PO and correctors that increase CFTR traffick-
evolving CF pharmacotherapy is an exciting field that warrants
ing to the plasma membrane [163]. While ENaC antagonists
further investigation. ENaC antagonists can be either inhaled
show efficacy when administered alone, it is likely that an ENaC
or oral. However, whilst oral ENaC antagonists exist, inhalation
antagonist in combination with CFTR modulators would be ben-
is the preferred route in order to maximize exposure and limit
eficial/synergistic in increasing CF ASL hydration. When VX-770
extrapulmonary effects including K+ sparing natriuresis and
(ivacaftor, Kalydeco) is added to CFTRG551D-expressing cultures,
hyperkalemia [107]. Thus, a major hurdle to developing ENaC
there is a 10–20% correction of CFTRG551D function [164]. While
antagonists is to constrain the compound to the lung and
this is a significant improvement, it is still 80–90% less than wild-
prevent any renal effects. A schematic summary of the main
type CFTR function, and how much CFTR function is needed to
classes of ENaC inhibitors is depicted in Figure 4
‘fix’ the CF lung is still under debate. Crucially, CFTRG551D, which
traffics to the plasma membrane but fails to open, with VX-770, is
currently the best case scenario for approved CFTR modulators;
4. Expert opinion
yet these patients fail to eradicate bacteria from their lungs over
time [165], although we acknowledge that newer, more potent Due to the complex and severe nature of CF lung disease, it is
CFTR modulators are under development which may improve on increasingly evident that the use of novel pharmacological
this. For CFTRΔF508, which has (1) reduced expression, (2) trouble ENaC inhibitors to attenuate Na+ absorption is becoming an
escaping the endoplasmic reticulum’s quality control mechan- attractive therapeutic approach to treat CF lung disease that
isms, (3) has a reduction in Po, and (4) has a reduced residency may be used alone or in combination with other therapies.
time in the plasma membrane, restoration of function may be Early ENaC antagonists such as amiloride suffered from poor
more problematic [166–169]. Thus, multiple therapies will likely duration of action and off target effects. Although the spec-
be needed to prolong patient survival and improve their quality trum of ENaC inhibitors has expanded during the last
of life and ENaC inhibition in the face of CFTR modulation is 5–10 years with many showing increased potency over amilor-
predicted to be synergistic. This hypothesis is born out experi- ide, many have failed to progress past Phase II clinical trials,
mentally: the intracellular Cl− concentration is almost threefold most commonly due to hyperkalemia. Thus, the two greatest
lower than that in the ASL [26]. Thus, there is no chemical hurdles to ENaC antagonists becoming viable therapies are (1)
gradient for Cl− secretion. This is important, because ion chan- duration of action and (2) preventing hyperkalemia. While
nels such as CFTR are passive and only move Cl− according to the protease inhibitors are unlikely to cause hyperkalemia, they
electrochemical gradient. So, in order for Cl− to be secreted into may suffer from not being broad-spectrum enough for the
696 P. J. MOORE AND R. TARRAN

Figure 4. ENaC inhibition as a strategy for ASL rehydration. (1) Pharmacological inhibitors such as amiloride directly inhibit Na+ absorption by occupying the channel
pore. (2) Novel peptides such as SPX-101 reduce the number of available ENaC subunits at the plasma membrane. (3) Protease inhibitors prevent channel activating
proteases from cleaving ENaC, maintaining ENaC in an inactive form; thus, preventing Na+ absorption. (4) Antisense oligonucleotides inhibit mRNA translation and
prevent ENaC trafficking to the surface of the cell.

highly proteolytic CF lung environment. Given the previous Declaration of interest


failures in getting foreign DNA/RNA into the lung, ASOs may R. Tarran is a founder of and has equity in Spyryx Biosciences, USA. The
suffer from achieving sufficient ENaC knockdown. authors have no other relevant affiliations or financial involvement with
In this review, we have highlighted the failures of small any organization or entity with a financial interest in or financial conflict
molecule antagonists, as well as potential successes of novel with the subject matter or materials discussed in the manuscript. This
approaches that may be used in alone or in combination with includes employment, consultancies, honoraria, stock ownership or
options, expert testimony, grants or patents received or pending, or
current CFTR modulators to alleviate CF symptoms. ENaC royalties.
inhibitors such as P-1037/VX-371 demonstrated high affinity
for ENaC. However, CF clinical trials with this compound were
not positive. It is important to highlight, therefore, that even
Reviewer of interest
though a small molecule may be highly potent in vitro and in
animal models, the complex nature of the CF lung may reduce One reviewer is affiliated with Enterprise Therapeutics, Sussex, UK. Other
peer reviewers on this manuscript have no relevant financial or other
the efficacy and may lead to unfavorable pharmacodynamics.
relationships to disclose
Currently, there are several potential new drugs that are in
the development pipeline, some of which are in clinical trials.
The novel peptide SPX-101 offers a key advantage over ‘tradi-
tional’ small molecule antagonists including negligible sys- References
temic absorption and a long duration of action. Generating Papers of special note have been highlighted as either of interest (•) or of
novel peptides for therapeutic intervention presents a unique considerable interest (••) to readers.
and exciting opportunity but is not without challenges, includ- 1. Internet. Foundation NACF. 2016 North American CFF Patient
Registry Annual Data Report [cited 2018 Mar 14]. Available from:
ing the need for stability in the face of a highly proteolytic
https://www.cff.org/Research/Researcher-Resources/Patient-
environment. Given the number of ENaC antagonists currently Registry/2016-Patient-Registry-Annual-Data-Report.pdf.
being developed that employ multiple modes of action, it is 2. Riordan J, Rommens J, Kerem B, et al. Identification of the cystic
hopeful that one of these will translate into the clinic in the fibrosis gene: cloning and characterization of complementary DNA.
near future. Science. 1989;245(4922):1066–1073.
3. Vankeerberghen A, Cuppens H, Cassiman J-J. The cystic fibrosis
transmembrane conductance regulator: an intriguing protein with
Funding pleiotropic functions. J Cyst Fibros. 2002;1(1):13–29.
4. Gibson RL, Burns JL, Ramsey BW. Pathophysiology and
This paper was funded by the CF Foundation, USA, and Emily’s Entourage, Management of Pulmonary Infections in Cystic Fibrosis. Am J
USA. Respir Crit Care Med. 2003;168(8):918–951.
EXPERT OPINION ON THERAPEUTIC TARGETS 697

5. Wilschanski M, Novak I. The Cystic Fibrosis of Exocrine Pancreas. 26. Tarran R, Grubb BR, Gatzy JT, et al. The Relative Roles of Passive
Cold Spring Harb Perspect Med. 2013;3(5):a009746. PubMed PMID: Surface Forces and Active Ion Transport in the Modulation of
PMC3633181. Airway Surface Liquid Volume and Composition. J Gen Physiol.
6. VanDevanter DR, Kahle JS, O’Sullivan AK, et al. Cystic fibrosis in 2001;118(2):223.
young children: a review of disease manifestation, progression, and 27. Garty H, Palmer LG. Epithelial sodium channels: function, structure,
response to early treatment. J Cyst Fibros. 2016;15(2):147–157. and regulation. Physiol Rev. 1997;77(2):359–396. PubMed PMID:
7. Nichols D, Chmiel J, Berger M. Chronic Inflammation in the Cystic 9114818.
Fibrosis Lung: alterations in Inter- and Intracellular Signaling. Clin 28. Butterworth MB. Regulation of the epithelial sodium channel
Rev Allergy Immunol. 2008;34(2):146–162. (ENaC) by membrane trafficking. Biochim Biophys Acta. 2010 Mar
8. Puchelle E, Bajolet O, Abély M. Airway mucus in cystic fibrosis. 27;1802(12):1166–1177. PubMed PMID: PMC2921481.
Paediatr Respir Rev. 2002;3(2):115–119. 29. Hamm LL, Feng Z, Hering-Smith KS. Regulation of sodium transport by
9. Salsgiver EL, Fink AK, Knapp EA, et al. Changing Epidemiology of ENaC in the kidney. Curr Opin Nephrol Hypertens. 2010;19(1):98–105.
the Respiratory Bacteriology of Patients With Cystic Fibrosis. Chest. PubMed PMID: PMC2895494.
2016 Jan 12;149(2):390–400. PubMed PMID: PMC5831653. 30. Stockand JD, Staruschenko A, Pochynyuk O, et al. Insight toward
10. Muhlebach MS, Zorn BT, Esther CR, et al. Initial acquisition and epithelial Na+ channel mechanism revealed by the acid-sensing
succession of the cystic fibrosis lung microbiome is associated with ion channel 1 structure. IUBMB Life. 2008;60(9):620–628.
disease progression in infants and preschool children. PLoS Pathog. 31. Sheng S, McNulty KA, Harvey JM, et al. Second Transmembrane
2018;14(1):e1006798. Domains of ENaC Subunits Contribute to Ion Permeation and
11. Kidd TJ, Ramsay KA, Vidmar S, et al. Pseudomonas aeruginosa Selectivity. J Biol Chem. 2001;276(47):44091–44098.
genotypes acquired by children with cystic fibrosis by age 5- 32. Kellenberger S, Gautschi I, Schild L. An External Site Controls Closing
years. J Cyst Fibros. 2015;14(3):361–369. of the Epithelial Na+ Channel ENaC. J Physiol. 2004;543(2):413–424.
12. Landry RM, An D, Hupp JT, et al. Mucin–pseudomonas aeruginosa 33. Kerem E, Bistritzer T, Hanukoglu A, et al. Pulmonary epithelial
interactions promote biofilm formation and antibiotic resistance. sodium-channel dysfunction and excess airway liquid in pseudo-
Mol Microbiol. 2006;59(1):142–151. hypoaldosteronism. N Engl J Med. 1999 Jul 15;341(3):156–162.
13. Mathee K, Ciofu O, Sternberg C, et al. Mucoid conversion of PubMed PMID: 10403853.
Pseudomonas aeruginos by hydrogen peroxide: a mechanism for 34. Staub O, Gautschi I, Ishikawa T, et al. Regulation of stability and
virulence activation in the cystic fibrosis lung. Microbiology. function of the epithelial Na+ channel (ENaC) by ubiquitination.
1999;145(6):1349–1357. Embo J. 1997 Nov 3;16(21):6325–6336. PubMed PMID: 9351815.
14. Cornelis P, Dingemans J. Pseudomonas aeruginosa adapts its iron 35. Kashlan OB, Kleyman TR. ENaC structure and function in the wake
uptake strategies in function of the type of infections. Front Cell of a resolved structure of a family member. Am J Physiology-Renal
Infect Microbiology.. 2013;3:75. Physiol. 2011;301(4):F684–F696.
15. Pezzulo AA, Tang XX, Hoegger MJ, et al. Reduced Airway Surface pH 36. Snyder PM, Bucher DB, Olson DR. Gating Induces a
Impairs Bacterial Killing in the Porcine Cystic Fibrosis Lung. Nature. Conformational Change in the Outer Vestibule of Enac. J Gen
2012 Jul 04;487(7405):109–113. PubMed PMID: PMC3390761. Physiol. 2000;116(6):781.
16. Worlitzsch D, Tarran R, Ulrich M, et al. Effects of reduced mucus 37. Harris M, Garcia-Caballero A, Stutts MJ, et al. Preferential Assembly
oxygen concentration in airway Pseudomonas infections of cystic of Epithelial Sodium Channel (ENaC) Subunits in Xenopus Oocytes:
fibrosis patients. J Clin Invest. 2002 Feb 01;109(3):317–325. . ROLE OF FURIN-MEDIATED ENDOGENOUS PROTEOLYSIS. J Biol
17. Nakamura H, Yoshimura K, McElvaney NG, et al. Neutrophil elastase Chem. 2008;283(12):7455–7463.
in respiratory epithelial lining fluid of individuals with cystic fibrosis 38. Ji H-L, Su X-F, Kedar S, et al. δ-Subunit Confers Novel Biophysical
induces interleukin-8 gene expression in a human bronchial Features to αβγ-Human Epithelial Sodium Channel (ENaC) via a
epithelial cell line. J Clin Investig. 1992;89(5):1478–1484. PubMed Physical Interaction. J Biol Chem. 2006;281(12):8233–8241.
PMID: PMC443018. 39. Bangel-Ruland N, Sobczak K, Christmann T, et al. Characterization
18. Black HR, Yankaskas JR, Johnson LG, et al. Interleukin-8 Production of the Epithelial Sodium Channel δ-Subunit in Human Nasal
by Cystic Fibrosis Nasal Epithelial Cells after Tumor Necrosis Factor- Epithelium. Am J Respir Cell Mol Biol. 2010;42(4):498–505.
α and Respiratory Syncytial Virus Stimulation. Am J Respir Cell Mol PubMed PMID: 19520916.
Biol. 1998;19(2):210–215. PubMed PMID: 9698592. 40. Hughey RP, Bruns JB, Kinlough CL, et al. Epithelial Sodium Channels
19. Bonfield TL, Panuska JR, Konstan MW, et al. Inflammatory cytokines in Are Activated by Furin-dependent Proteolysis. J Biol Chem.
cystic fibrosis lungs. Am J Respir Crit Care Med. 1995;152(6):2111–2118. 2004;279(18):18111–18114.
20. Mitola S, Sorbello V, Ponte E, et al. Tumor Necrosis Factor-α in 41. Vuagniaux G, Vallet V, Jaeger NF, et al. Activation of the Amiloride-
Airway Secretions from Cystic Fibrosis Patients Upregulate Sensitive Epithelial Sodium Channel by the Serine Protease mCAP1
Endothelial Adhesion Molecules and Induce Airway Epithelial Cell Expressed in a Mouse Cortical Collecting Duct Cell Line. J Am Soc
Apoptosis: implications for Cystic Fibrosis Lung Disease. Int J Nephrol. 2000;11(5):828–834.
Immunopathol Pharmacol. 2008;21(4):851–865. 42. Caldwell RA, Boucher RC, Stutts MJ. Neutrophil elastase activates
21. Norman D, Elborn JS, Cordon SM, et al. Plasma tumour necrosis near-silent epithelial Na+ channels and increases airway epithelial
factor alpha in cystic fibrosis. Thorax. 1991;46(2):91–95. PubMed Na+ transport. Am J Physiology-Lung Cell Mol Physiol. 2005;288(5):
PMID: PMC462953. L813–L819. PubMed PMID: 15640288.
22. Greally P, Hussein MJ, Cook AJ, et al. Sputum tumour necrosis 43. Donaldson SH, Hirsh A, Li DC, et al. Regulation of the Epithelial
factor-alpha and leukotriene concentrations in cystic fibrosis. Arch Sodium Channel by Serine Proteases in Human Airways. J Biol
Dis Child. 1993;68(3):389–392. PubMed PMID: PMC1793872. Chem. 2002;277(10):8338–8345.
23. Rowe SM, Miller S, Ej S. Cystic Fibrosis. New England J Med. 44. Antalis TM, Buzza MS, Hodge KM, et al. The Cutting Edge: mem-
2005;352(19):1992–2001. PubMed PMID: 15888700. brane Anchored Serine Protease Activities in the Pericellular
24. Weiner DJ, Bucki R, Janmey PA. The Antimicrobial Activity of the Microenvironment. Biochem J. 2010;428(3):325–346. PubMed
Cathelicidin LL37 Is Inhibited by F-actin Bundles and Restored by PMID: PMC3680374.
Gelsolin. Am J Respir Cell Mol Biol. 2003;28(6):738–745. PubMed 45. García-Caballero A, Dang Y, He H, et al. ENaC Proteolytic Regulation
PMID: 12600826. by Channel-activating Protease 2. J Gen Physiol. 2008;132(5):521.
25. É F, Humphreys H, Greene CM, et al. Potential of Host Defense 46. Bruns JB, Carattino MD, Sheng S, et al. Epithelial Na+ Channels
Peptide Prodrugs as Neutrophil Elastase-Dependent Anti- Are Fully Activated by Furin- and Prostasin-dependent Release
Infective Agents for Cystic Fibrosis. Antimicrob Agents of an Inhibitory Peptide from the γ-Subunit. J Biol Chem.
Chemother. 2014;58(2):978–985. 2007;282(9):6153–6160.
698 P. J. MOORE AND R. TARRAN

47. Vallet V, Chraibi A, Gaeggeler H-P, et al. An epithelial serine pro- 66. Knight KK, Olson DR, Zhou R, et al. Liddle’s syndrome mutations
tease activates the amiloride-sensitive sodium channel. Nature. increase Na+ transport through dual effects on epithelial Na+
1997 Oct 09;389:607. channel surface expression and proteolytic cleavage. Proc Natl
48. Vuagniaux G, Vallet V, Jaeger NF, et al. Synergistic Activation of Acad Sci U S A. 2006;103(8):2805.
ENaC by Three Membrane-bound Channel-activating Serine 67. Kimura T, Kawabe H, Jiang C, et al. Deletion of the ubiquitin ligase
Proteases (mCAP1, mCAP2, and mCAP3) and Serum- and Nedd4L in lung epithelia causes cystic fibrosis-like disease. Proc
Glucocorticoid-regulated Kinase (Sgk1) in Xenopus Oocytes. J Gen Natl Acad Sci U S A. 2011 Feb 07;108(8):3216–3221. PubMed PMID:
Physiol. 2002;120(2):191. PMC3044364.
49. Andreasen D, Vuagniaux G, Fowler-Jaeger N, et al. Activation of 68. Staub O, Dho S, Henry P, et al. WW domains of Nedd4 bind to the
Epithelial Sodium Channels by Mouse Channel Activating proline-rich PY motifs in the epithelial Na+ channel deleted in
Proteases (mCAP) Expressed in Xenopus Oocytes Requires Liddle’s syndrome. EMBO J. 1996;15(10):2371–2380. PubMed
Catalytic Activity of mCAP3 and mCAP2 but not mCAP1. J Am PMID: PMC450167.
Soc Nephrol. 2006;17(4):968–976. 69. Butterworth MB, Edinger RS, Ovaa H, et al. The Deubiquitinating
50. Chraïbi A, Vallet V, Firsov D, et al. Protease Modulation of the Enzyme UCH-L3 Regulates the Apical Membrane Recycling of the
Activity of the Epithelial Sodium Channel Expressed in Xenopus Epithelial Sodium Channel. J Biol Chem. 2007;282(52):37885–37893.
Oocytes. J Gen Physiol. 1998;111(1):127. 70. Schild L, Lu Y, Gautschi I, et al. Identification of a PY motif in the
51. Harris M, Firsov D, Vuagniaux G, et al. A Novel Neutrophil Elastase epithelial Na channel subunits as a target sequence for mutations
Inhibitor Prevents Elastase Activation and Surface Cleavage of the causing channel activation found in Liddle syndrome. EMBO J.
Epithelial Sodium Channel Expressed in Xenopus laevis Oocytes. J 1996;15(10):2381–2387. PubMed PMID: PMC450168.
Biol Chem. 2007;282(1):58–64. 71. Bl R, Ep O, Ja K, et al. A new mutation, R563Q, of the beta subunit
52. Snyder PM, Olson DR, Kabra R, et al. cAMP and Serum and of the epithelial sodium channel associated with low-renin, low-
Glucocorticoid-inducible Kinase (SGK) Regulate the Epithelial Na+ aldosterone hypertension. J Hypertens. 2003;21:5.
Channel through Convergent Phosphorylation of Nedd4-2. J Biol 72. Hiltunen TP, Hannila-Handelberg T, Petäjäniemi N, et al. Liddle’s
Chem. 2004;279(44):45753–45758. syndrome associated with a point mutation in the extracellular
53. Stutts MJ, Canessa CM, Olsen JC, et al. CFTR as a cAMP-dependent domain of the epithelial sodium channel γ subunit. J Hypertens.
regulator of sodium channels. Science. 1995;269(5225):847. 2002;20:12.
54. Stutts MJ, Rossier BC, Boucher RC. Cystic Fibrosis Transmembrane 73. Di YP. Functional roles of SPLUNC1 in the innate immune
Conductance Regulator Inverts Protein Kinase A-mediated response against Gram-negative bacteria. Biochem Soc Trans.
Regulation of Epithelial Sodium Channel Single Channel Kinetics. 2011;39(4):1051.
J Biol Chem. 1997;272(22):14037–14040. 74. Hobbs CA, Blanchard MG, Alijevic O, et al. Identification of the
55. Kunzelmann K, Mall M, Briel M, et al. The cystic fibrosis transmem- SPLUNC1 ENaC-inhibitory domain yields novel strategies to treat
brane conductance regulator attenuates the endogenous Ca2+ sodium hyperabsorption in cystic fibrosis airway epithelial cultures.
activated Cl- conductance of Xenopus oocytes. Pflugers Arch. Am J Physiol Lung Cell Mol Physiol. 2013;305(12):L990–L1001.
1997 Dec;435(1):178–181. PubMed PMID: 9359918. 75. Garcia-Caballero A, Rasmussen JE, Gaillard E, et al. SPLUNC1
56. Schreiber R, Hopf A, Mall M, et al. The first-nucleotide binding regulates airway surface liquid volume by protecting
domain of the cystic-fibrosis transmembrane conductance regula- ENaC from proteolytic cleavage. Proc Natl Acad Sci. 2009 Jul
tor is important for inhibition of the epithelial Na+ channel. Proc 7;106(27):11412–11417.
Natl Acad Sci U S A. 1999 Apr 27;96(9):5310–5315. PubMed PMID: •• Important study demonstrating ENaC activity and ASL hydra-
10220462; eng. tion is regulated by SPLUNC1.
57. Ji HL, Chalfant ML, Jovov B, et al. The cytosolic termini of the 76. Kim CS, Ahmad S, Wu T, et al. SPLUNC1 is an allosteric modulator of
beta- and gamma-ENaC subunits are involved in the functional the epithelial sodium channel. FASEB J. 2018;fj.201701126R.
interactions between cystic fibrosis transmembrane conductance DOI:10.1096/fj.201701126R.
regulator and epithelial sodium channel. J Biol Chem. 2000 Sep 77. Kerem E, Bistritzer T, Hanukoglu A, et al. Pulmonary Epithelial Sodium-
8;275(36):27947–27956. PubMed PMID: 10821834. Channel Dysfunction and Excess Airway Liquid in
58. Reddy MM, Light MJ, Quinton PM. Activation of the epithelial Na+ Pseudohypoaldosteronism. New England J Med. 1999;341(3):156–162.
channel (ENaC) requires CFTR Cl- channel function. Nature. 1999 PubMed PMID: 10403853.
11 18;402(6759):301–304. 78. Hyde DM, Hamid Q, Irvin CG. Anatomy, pathology, and physiology
59. Mo L, Wills NK. ClC-5 chloride channel alters expression of the epithelial of the tracheobronchial tree: emphasis on the distal airways. J
sodium channel (ENaC). J Membr Biol. 2004 Nov;202(1):21–37. PubMed Allergy Clin Immunol. 2009;124(6):S72–S77.
PMID: 15702377. 79. Patwa A, Shah A. Anatomy and physiology of respiratory system
60. Bachhuber T, Konig J, Voelcker T, et al. Cl- interference with the relevant to anaesthesia. Indian J Anaesth. 2015;59(9):533–541.
epithelial Na+ channel ENaC. J Biol Chem. 2005 Sep 9;280 PubMed PMID: PMC4613399.
(36):31587–31594. PubMed PMID: 16027156. 80. Liedtke CM. Electrolyte transport in the epithelium of
61. Willumsen NJ, Boucher RC. Intracellular pH and its relationship to pulmonary segments of normal and cystic fibrosis lung. FASEB J.
regulation of ion transport in normal and cystic fibrosis human 1992;6(12):3076–3084.
nasal epithelia. J Physiol. 1992;455: 247–269. PubMed PMID: 81. Knowles MR, Buntin WH, Bromberg PA, et al. Measurements of
PMC1175643. Transepithelial Electric Potential Differences in the Trachea and
62. Hall IP. Second messengers, ion channels and pharmacology of Bronchi of Human Subjects in vivo. Am Rev Respir Dis.
airway smooth muscle. Eur Respir J. 2000;15(6):1120. 1982;126(1):108–112.
63. Ma HP, Saxena S, Warnock DG. Anionic phospholipids regulate 82. Knowles M, Gatzy J, Boucher R. Increased Bioelectric Potential
native and expressed epithelial sodium channel (ENaC). J Biol Difference across Respiratory Epithelia in Cystic Fibrosis. New
Chem. 2002 Mar 8;277(10):7641–7644. . England J Med. 1981;305(25):1489–1495.
64. Yue G, Malik B, Eaton DC. Phosphatidylinositol 4,5-bisphosphate 83. Boucher RC, Stutts MJ, Knowles MR, et al. Na+ transport in cystic fibrosis
(PIP2) stimulates epithelial sodium channel activity in A6 cells. J respiratory epithelia. Abnormal basal rate and response to adenylate
Biol Chem. 2002 Apr 5;277(14):11965–11969. . cyclase activation. J Clin Invest. 1986 Nov 01;78(5):1245–1252. .
65. Mall M, Wissner A, Gonska T, et al. Inhibition of Amiloride-Sensitive 84. Itani OA, Chen J-H, Karp PH, et al. Human cystic fibrosis airway
Epithelial Na+ Absorption by Extracellular Nucleotides in Human epithelia have reduced Cl(−) conductance but not increased Na(+)
Normal and Cystic Fibrosis Airways. Am J Respir Cell Mol Biol. conductance. Proc Natl Acad Sci U S A. 2011 June 06;108
2000;23(6):755–761. (25):10260–10265. PubMed PMID: PMC3121869.
EXPERT OPINION ON THERAPEUTIC TARGETS 699

85. Tarran R, Trout L, Donaldson SH, et al. Soluble Mediators, Not Cilia, Nat Med. 2004Apr;10(5):487–493. http://www.nature.com/nm/jour
Determine Airway Surface Liquid Volume in Normal and Cystic nal/v10/n5/suppinfo/nm1028_S1.html.
Fibrosis Superficial Airway Epithelia. J Gen Physiol. 2006;127(5):591. 107. Cragoe EJ, Woltersdorf OW, Bicking JB, et al. Pyrazine Diuretics. II.
86. Garland AL, Walton WG, Coakley RD, et al. Molecular basis for pH- N-Amidino-3-amino-5-substituted 6-Halopyrazinecarboxamides. J
dependent mucosal dehydration in cystic fibrosis airways. Proc Natl Med Chem. 1967;10(1):66–75.
Acad Sci. 2013;110(40):15973–15978. 108. Schmitt R, Ellison DH, Farman N, et al. Developmental expression of
87. Matsui H, Grubb BR, Tarran R, et al. Evidence for Periciliary Liquid Layer sodium entry pathways in rat nephron. Am J Physiology-Renal
Depletion, Not Abnormal Ion Composition, in the Pathogenesis of Physiol. 1999;276(3):F367–F381.
Cystic Fibrosis Airways Disease. Cell. 1998;95(7):1005–1015. 109. Loffing J, Zecevic M, Féraille E, et al. Aldosterone induces rapid
88. Tarran R, Button B, Picher M, et al. Normal and Cystic Fibrosis apical translocation of ENaC in early portion of renal collecting
Airway Surface Liquid Homeostasis: the effects of phasic shear system: possible role of SGK. Am J Physiology-Renal Physiol.
stress and viral infections. J Biol Chem. 2005;280(42):35751–35759. 2001;280(4):F675–F682.
89. Myerburg MM, Butterworth MB, McKenna EE, et al. Airway Surface 110. Rengo F, Trimarco B, Bonaduce D, et al. Potassium sparing effect of
Liquid Volume Regulates ENaC by Altering the Serine Protease- amiloride in patients receiving diuretics: a quantitative study. Acta
Protease Inhibitor Balance: a mechanism for sodium hyperabsorp- Cardiol. 1979;34(4):259–267. PubMed PMID: 315689; eng.
tion in cystic fibrosis. J Biol Chem. 2006;281(38):27942–27949. • Development of amiloride for treatment of hypokalemia.
90. Myerburg MM, Harvey PR, Heidrich EM, et al. Acute Regulation of 111. Em A, King M, Helfesrieder R, et al. Acute and Long-term Amiloride
the Epithelial Sodium Channel in Airway Epithelia by Proteases and Inhalation in Cystic Fibrosis Lung Disease: A Rational Approach to
Trafficking. Am J Respir Cell Mol Biol. 2010;43(6):712–719. Cystic Fibrosis Therapy. Am Rev Respir Dis. 1990;141(3):605–612.
91. Bingle L, Barnes FA, Cross SS, et al. Differential epithelial expression 112. Tg Dk O, Hodsman P, Ansede JH, et al. Acute Hyperkalemia
of the putative innate immune molecule SPLUNC1 in Cystic Associated with Inhalation of a Potent ENaC Antagonist: phase 1
Fibrosis. Respir Res. 2007;8(1):79. Trial of GS-9411. J Aerosol Med Pulm Drug Deliv. 2014;27:3.
92. Ahmad S, Tyrrell J, Walton WG, et al. Short Palate, Lung, and Nasal 113. Wilcox CS. New Insights into Diuretic Use in Patients with Chronic
Epithelial Clone 1 Has Antimicrobial and Antibiofilm Activities Renal Disease. J Am Soc Nephrol. 2002;13(3):798–805.
against the Burkholderia cepacia Complex. Antimicrob Agents 114. Matsui H, Randell SH, Peretti SW, et al. Coordinated clearance of
Chemother. 2016;60(10):6003–6012. periciliary liquid and mucus from airway surfaces. J Clin Invest.
93. Jiang D, Wenzel SE, Wu Q, et al. Human Neutrophil Elastase 1998 Sep 15;102(6):1125–1131. .
Degrades SPLUNC1 and Impairs Airway Epithelial Defense against 115. Köhler D, App E, Schmitz-Schumann M, et al. Inhalation of amilor-
Bacteria. PLOS ONE. 2013;8(5):e64689. ide improves the mucociliary and the cough clearance in patients
94. Prulière-Escabasse V, Clerici C, Vuagniaux G, et al. Effect of neutro- with cystic fibroses. Eur J Respir Dis Suppl. 1986;146:319–326.
phil elastase and its inhibitor EPI-hNE4 on transepithelial sodium PubMed PMID: 3465558; eng.
transport across normal and cystic fibrosis human nasal epithelial •• Use of amiloride increased mucociliary clearance rates in CF
cells. Respir Res. 2010;11(1):141. patients.
95. Butterworth MB, Zhang L, Heidrich EM, et al. Activation of the 116. Knowles MR, Church NL, Waltner WE, et al. A Pilot Study of
epithelial sodium channel (ENaC) by the alkaline protease from Aerosolized Amiloride for the Treatment of Lung Disease in Cystic
Pseudomonas aeruginosa. J Biol Chem. 2012. Fibrosis. New England J Med. 1990;322(17):1189–1194.
96. Butterworth MB, Zhang L, Liu X, et al. Modulation of the Epithelial 117. Scheffer GL, Pijnenborg ACLM, Smit EF, et al. Multidrug resistance
Sodium Channel (ENaC) by Bacterial Metalloproteases and Protease related molecules in human and murine lung. J Clin Pathol. 2002
Inhibitors. PLOS ONE. 2014;9(6):e100313. Dec 12;55(5):332–339. PubMed PMID: PMC1769658.
97. Sheridan MB, Fong P, Groman JD, et al. Mutations in the beta- 118. Bahadduri PM, D’Souza VM, Pinsonneault JK, et al. Functional
subunit of the epithelial Na+ channel in patients with a cystic Characterization of the Peptide Transporter PEPT2 in Primary
fibrosis-like syndrome. Hum Mol Genet. 2005;14(22):3493–3498. Cultures of Human Upper Airway Epithelium. American Journal of
98. Azad Abul K, Rauh R, Vermeulen F, et al. Mutations in the amilor- Respiratory Cell and Molecular. Biology. 2005;32(4):319–325.
ide-sensitive epithelial sodium channel in patients with cystic fibro- 119. Hirsh AJ, Zhang J, Zamurs A, et al. Pharmacological Properties of
sis-like disease. Hum Mutat. 2009;30(7):1093–1103. N-(3,5-Diamino-6-chloropyrazine-2-carbonyl)-N′-4-[4-(2,3-dihydrox-
99. Rauh R, Soell D, Haerteis S, et al. A mutation in the β-subunit of ypropoxy)phenyl]butyl-guanidine Methanesulfonate (552-02), a
ENaC identified in a patient with cystic fibrosis-like symptoms has a Novel Epithelial Sodium Channel Blocker with Potential Clinical
gain-of-function effect. Am J Physiology-Lung Cell Mol Physiol. Efficacy for Cystic Fibrosis Lung Disease. J Pharmacol Exp Ther.
2013;304(1):L43–L55. PubMed PMID: 23087020. 2008;325(1):77–88.
100. Ramos MD, Trujillano D, Olivar R, et al. Extensive sequence analysis 120. Zhou Z, Treis D, Schubert SC, et al. Preventive but Not Late
of CFTR, SCNN1A, SCNN1B, SCNN1G and SERPINA1 suggests an Amiloride Therapy Reduces Morbidity and Mortality of Lung
oligogenic basis for cystic fibrosis-like phenotypes. Clin Genet. Disease in βENaC-overexpressing Mice. Am J Respir Crit Care
2013;86(1):91–95. Med. 2008;178(12):1245–1256. PubMed PMID: 18849497.
101. Agrawal PB, Wang R, Li HL, et al. The Epithelial Sodium Channel Is 121. Thelin WD, Ansede K, Johnson J. Poster Session Abstract 201. The
a Modifier of the Long-Term Nonprogressive Phenotype ENaC inhibitor P-1037 is CFTR Independent Therapeutic Agent That
Associated with F508del CFTR Mutations. Am J Respir Cell Mol Promotes Sustained Airways Hydration and Mucociliary Transport.
Biol. 2017;57(6):711–720. PubMed PMID: 28708422. Pediatr Pulmonol. 2015;50(S41):S193–S453.
102. Saferali A, Obeidat M, J-C B, et al. Polymorphisms Associated with 122. Abm Å, Hemmerling M, Root J, et al. Linking increased airway
Expression of BPIFA1/BPIFB1 and Lung Disease Severity in Cystic hydration, ciliary beating, and mucociliary clearance through
Fibrosis. Am J Respir Cell Mol Biol. 2015;53(5):607–614. ENaC inhibition. Am J Physiology-Lung Cell Mol Physiol.
103. Snouwaert JN, Brigman KK, Latour AM, et al. An Animal Model for Cystic 2014;308(1):L22–L32.
Fibrosis Made by Gene Targeting. Science. 1992;257(5073):1083–1088. 123. Fortinberry H, Birket S, Astrand A, et al. ENaC Inhibitor AZD5634
104. Tarran R, Grubb BR, Parsons D, et al. The CF Salt Controversy: in Vivo Augments Airway Surface Liquid and Mucociliary Transport in
Observations and Therapeutic Approaches. Mol Cell. 2001;8(1):149–158. Primary Cystic Fibrosis Airway Cells. C80-A. EVERYTHING YOU
105. Grubb BR, Vick RN, Boucher RC. Hyperabsorption of Na+ and raised NEED TO KNOW ABOUT BRONCHOPULMONARY DYSPLASIA,
Ca(2+)-mediated Cl- secretion in nasal epithelia of CF mice. Am J CYSTIC FIBROSIS AND DIFFUSE PARENCHYMAL LUNG DISEASES.
Physiology-Cell Physiol. 1994;266(5):C1478–C1483. PubMed PMID: A6466–A6466.
7515571. 124. Gardiner P, Malmgren A, Ersdal E, et al. ENaC Inhibitor AZD5634
106. Mall M, Grubb BR, Harkema JR, et al. Increased airway epithelial Na First in Human Trial Reveals Promising Clinical Profile for the
+ absorption produces cystic fibrosis-like lung disease in mice []. Treatment of Cystic Fibrosis. D94. Advances in cystic fibrosis and
700 P. J. MOORE AND R. TARRAN

non-cystic fibrosis bronchiectasis. American Thoracic Society 144. Moore PJ, Reidel B, Ghosh A, et al. Cigarette smoke modifies and
International Conference Abstracts: American Thoracic Society. inactivates SPLUNC1, leading to airway dehydration. FASEB J. 2018;
2017. A7306–A7306. fj.201800345R. DOI:10.1096/fj.201800345R.
125. Coote K, Hc A-W, Sugar R, et al. Camostat Attenuates Airway 145. Clunes LA, Davies CM, Coakley RD, et al. Cigarette smoke exposure
Epithelial Sodium Channel Function in Vivo through the induces CFTR internalization and insolubility, leading to airway
Inhibition of a Channel-Activating Protease. J Pharmacol Exp Ther. surface liquid dehydration. FASEB J. 2012 Jul 19;26(2):533–545.
2009;329(2):764–774. PubMed PMID: PMC3290447.
126. Ornatowski W, Poschet JF, Perkett E, et al. Elevated furin levels in 146. Shak S, Capon DJ, Hellmiss R, et al. Recombinant human DNase I
human cystic fibrosis cells result in hypersusceptibility to exotoxin A– reduces the viscosity of cystic fibrosis sputum. Proc Natl Acad Sci.
induced cytotoxicity. J Clin Invest. 2007 Nov 01;117(11):3489–3497. . 1990;87(23):9188.
127. Tan CD, Hobbs C, Sameni M, et al. Cathepsin B contributes to Na+ 147. Fuchs HJ, Borowitz DS, Christiansen DH, et al. Effect of Aerosolized
hyperabsorption in cystic fibrosis airway epithelial cultures. J Recombinant Human DNase on Exacerbations of Respiratory
Physiol. 2014;592(23):5251–5268. Symptoms and on Pulmonary Function in Patients with Cystic
128. Tong Z, Illek B, Vj B, et al. Prostasin, a membrane-anchored serine Fibrosis. New England J Med. 1994 Sep 08;331(10):637–642.
peptidase, regulates sodium currents in JME/CF15 cells, a cystic 148. Quan JM, Tiddens HAWM, Sy JP, et al. A two-year randomized,
fibrosis airway epithelial cell line. Am J Physiology-Lung Cell Mol placebo-controlled trial of dornase alfa in young patients with
Physiol. 2004;287(5):L928–L935. PubMed PMID: 15246975. cystic fibrosis with mild lung function abnormalities. J Pediatr.
129. Reihill JA, Walker B, Hamilton RA, et al. Inhibition of Protease– 2001;139(6):813–820.
epithelial Sodium Channel Signaling Improves Mucociliary 149. Hodson ME, McKenzie S, Harms HK, et al. Dornase alfa in the
Function in Cystic Fibrosis Airways. Am J Respir Crit Care Med. treatment of cystic fibrosis in Europe: A report from the
2016;194(6):701–710. PubMed PMID: 27014936. Epidemiologic Registry of Cystic Fibrosis. Pediatr Pulmonol.
130. Elborn JS, Perrett J, Forsman-Semb K, et al. Efficacy, safety and 2003;36(5):427–432.
effect on biomarkers of AZD9668 in cystic fibrosis. Eur Respir J. 150. Shah PL, Scott SF, Geddes DM, et al. Two years experience with
2012 Oct;40(4):969–976. PubMed PMID: 22267768; eng. recombinant Human DNase I in the treatment of pulmonary dis-
131. Martin SL, Moffitt KL, McDowell A, et al. Association of airway ease in cystic fibrosis. Respir Med. 1995;89(7):499–502.
cathepsin B and S with inflammation in cystic fibrosis. Pediatr 151. Davies J, Trinda De M- T, Wallis C, et al. Retrospective review of the
Pulmonol. 2010 Sep;45(9):860–868. PubMed PMID: 20632407; eng. effects of rhDNase in children with cystic fibrosis. Pediatr Pulmonol.
132. Rowe SM, Reeves G, Hathorne H, et al. Reduced Sodium Transport 1998. [cited 1995 Aug 01];23(4):243–248. AID-PPUL1>3.0.CO;2-N.
With Nasal Administration of the Prostasin Inhibitor Camostat in 152. Bollert FG, Paton JY, Marshall TG, et al. Recombinant DNase in
Subjects With Cystic Fibrosis. Chest. 2013;144(1):200–207. cystic fibrosis: a protocol for targeted introduction through n-of-1
133. Dias N, Stein CA. Antisense Oligonucleotides: basic Concepts and trials. Scottish Cystic Fibrosis Group. Eur Respir J. 1999;13(1):107.
Mechanisms. Mol Cancer Ther. 2002;1(5):347. 153. Van Der Giessen Lianne J, De Jongste Johan C, Gosselink R, et al.
134. Loke SL, Stein CA, Zhang XH, et al. Characterization of oligonucleo- RhDNase before airway clearance therapy improves airway patency
tide transport into living cells. Proc Natl Acad Sci U S A. 1989;86 in children with CF. Pediatr Pulmonol. 2007;42(7):624–630.
(10):3474–3478. PubMed PMID: PMC287160. 154. Wilson Christine J, Robbins Lisel J, Murphy Jennifer M, et al. Is a longer
135. Vlassov VV, Blakireva LA, Yakubov LA. Transport of oligonucleotides time interval between recombinant human deoxyribonuclease (dor-
across natural and model membranes. Biochimica Et Biophysica nase alfa) and chest physiotherapy better?: A multi-center, rando-
Acta (BBA) - Reviews on Biomembranes. 1994;1197(2):95–108. mized crossover trial. Pediatr Pulmonol. 2007;42(12):1110–1116.
136. Crosby JR, Zhao C, Jiang C, et al. Inhaled ENaC antisense oligonu- 155. De Boeck K, Haarman E, Hull J, et al. Inhaled dry powder mannitol
cleotide ameliorates cystic fibrosis-like lung disease in mice. J Cyst in children with cystic fibrosis: A randomised efficacy and safety
Fibros. 2017;16(6):671–680. trial. J Cyst Fibros. 2017;16(3):380–387.
137. Grubb BR, Pickles RJ, Ye H, et al. Inefficient gene transfer by 156. Donaldson SH, Bennett WD, Zeman KL, et al. Mucus Clearance and
adenovirus vector to cystic fibrosis airway epithelia of mice and Lung Function in Cystic Fibrosis with Hypertonic Saline. New
humans. Nature. 1994;371:802. . England J Med. 2006;354(3):241–250. PubMed PMID: 16421365.
138. Yang Y, Su Q, Wilson JM. Role of viral antigens in destructive 157. Daviskas E, Anderson SD, Brannan JD, et al. Inhalation of dry-
cellular immune responses to adenovirus vector-transduced cells powder mannitol increases mucociliary clearance. Eur Respir J.
in mouse lungs. J Virol. 1996;70(10):7209–7212. 1997;10(11):2449.
139. Terryah ST, Fellner RC, Ahmad S, et al. Evaluation of a SPLUNC1- 158. Bilton D, Robinson P, Cooper P, et al. Inhaled dry powder mannitol in
Derived Peptide for the Treatment of Cystic Fibrosis Lung Disease. cystic fibrosis: an efficacy and safety study. Eur Respir J. 2011;38(5):1071.
Am J Physiol Lung Cell Mol Physiol. 2017. DOI:10.1152/ • Pooled analysis of two Phase III trials demonstrated mannitol
ajplung.00546.2016. significantly improved FEV1 and observed a decrease in pul-
140. Kesimer M, Sheehan JK. Analyzing the functions of large glycocon- monary exacerbations.
jugates through the dissipative properties of their absorbed layers 159. Aitken ML, Bellon G, De Boeck K, et al. Long-Term Inhaled Dry
using the gel-forming mucin MUC5B as an example. Glycobiology. Powder Mannitol in Cystic Fibrosis. Am J Respir Crit Care Med.
2008;18(6):463–472. 2012;185(6):645–652.
141. Scott DW, Walker MP, Sesma J, et al. SPX-101 is a Novel ENaC- 160. Goralski JL, Wu D, Thelin WR, et al. The in vitro effect of nebulised
targeted Therapeutic for Cystic Fibrosis that Restores Mucus hypertonic saline on human bronchial epithelium. Eur Respir J.
Transport. Am J Respir Crit Care Med. 2017.. 2017;51(5). DOI:10.1183/13993003.02652.
•• This study demonstrated that SPX-101 significantly improves 161. Tarran R, Button B, Picher M, et al. Normal and Cystic Fibrosis
ASL rehydration and mucociliary clearance rates Airway Surface Liquid Homeostasis: THE EFFECTS OF PHASIC
142. Walker MP, Cowlen M, Christensen D, et al. Nonclinical safety SHEAR STRESS AND VIRAL INFECTIONS. J Biol Chem. 2005 Aug
assessment of SPX-101, a novel peptide promoter of epithelial 08;280(42):35751–35759. PubMed PMID: PMC2924153.
sodium channel internalization for the treatment of cystic fibrosis. 162. Van Goor F, Hadida S, Grootenhuis PDJ, et al. Rescue of CF airway
Inhal Toxicol. 2017;29(8):356–365. epithelial cell function in vitro by a CFTR potentiator, VX-770. Proc
143. Wheeler A, Schaberg A, Scott D, et al. Safety and Pharmacokinetics of Natl Acad Sci. 2009;106(44):18825.
SPX-101 in Healthy Human Subjects. B103. CLINICAL STUDIES IN 163. Van Goor F, Hadida S, Grootenhuis PDJ, et al. Correction of the
BRONCHIECTASIS, IMMUNODEFICIENCY, AND DRUG INDUCED F508del-CFTR protein processing defect in vitro by the investi-
LUNG DISEASE. American Thoracic Society International Conference gational drug VX-809. Proc Natl Acad Sci U S A. 2011 Oct
Abstracts: American Thoracic Society. 2017. A4736–A4736. 05;108(46):18843–18848. PubMed PMID: PMC3219147.
EXPERT OPINION ON THERAPEUTIC TARGETS 701

164. Ramsey BW, Davies J, McElvaney NG, et al. A CFTR Potentiator in 168. Sheppard DN, Rich DP, Ostedgaard LS, et al. Mutations in CFTR
Patients with Cystic Fibrosis and the G551D Mutation. New associated with mild-disease-form CI- channels with altered pore
England J Med. 2011;365(18):1663–1672. PubMed PMID: properties. Nature. 1993;362:160. .
22047557. 169. Haardt M, Benharouga M, Lechardeur D, et al. C-terminal Truncations
•• Landmark study showing that VX-770 significantly improves Destabilize the Cystic Fibrosis Transmembrane Conductance
lung function in CF patients with G551D mutation. Regulator without Impairing Its Biogenesis: A NOVEL CLASS OF
165. Hisert KB, Heltshe SL, Pope C, et al. Restoring Cystic Fibrosis MUTATION. J Biol Chem. 1999;274(31):21873–21877.
Transmembrane Conductance Regulator Function Reduces Airway 170. Boucher RC. Human airway ion transport. Part one. Am J Respir Crit
Bacteria and Inflammation in People with Cystic Fibrosis and Care Med. 1994;150(1):271–281.
Chronic Lung Infections. American. J Respir Crit Care Med. 171. Watson MS, Cutting GR, Desnick RJ, et al. Cystic fibrosis population
2017;195(12):1617–1628. carrier screening: 2004 revision of American College of Medical
166. Li M, McCann JD, Liedtket CM, et al. Cyclic AMP-dependent protein Genetics mutation panel. Genet Med. 2004 Sep-Oct;6(5):387–391.
kinase opens chloride channels in normal but not cystic fibrosis PubMed PMID: PMC3110945.
airway epithelium. Nature. 1988;331:358. . 172. Xue X, Mutyam V, Tang L, et al. Synthetic Aminoglycosides
167. Cheng SH, Gregory RJ, Marshall J, et al. Defective intracellular Efficiently Suppress Cystic Fibrosis Transmembrane Conductance
transport and processing of CFTR is the molecular basis of most Regulator Nonsense Mutations and Are Enhanced by Ivacaftor. Am
cystic fibrosis. Cell. 1990;63(4):827–834. J Respir Cell Mol Biol. 2013;50(4):805–816.

View publication stats

You might also like