Ensifentrine
Clinical Background
Chronic obstructive pulmonary disease (COPD) is a composite of progressive lung diseases including emphysema and chronic bronchitis. Approximately 12% of adults over the age of 65 have diagnosed COPD.1 Cigarette smoking is the leading cause of COPD. It may also be caused by chronic marijuana use,2 vaping, and exposure to air pollutants from traffic, second-hand smoke, or indoor fuels used for heating and cooking.3,4 Approximately 25-30% of people with COPD have never smoked.5 Individuals may be predisposed to developing COPD due to biological factors, such as systemic inflammation, or genetic conditions, such as alpha-1 antitrypsin deficiency.6,7 A family history of COPD is an independent risk factor for the development of COPD, severity of disease, and frequency of exacerbations.4,8 People who experience more frequent acute exacerbations experience a faster progression of their COPD and worse health status compared with those with few acute exacerbations.9 Severe COPD is a disability that limits individuals from participating in activities of daily life.10,11 People with COPD are at higher risk of developing other chronic health conditions, including respiratory tract infections, lung cancer, heart disease, musculoskeletal frailty, and depression.6,12 Patients with other comorbidities (e.g., pulmonary hypertension, cardiovascular disease, lung cancer) typically have a poorer prognosis.6
Chronic Obstructive Pulmonary Disease is characterized by inflammation and mucus plugging of the airways and structural changes of the alveoli, resulting in poor gas exchange and airflow obstruction.6 Pulmonary function testing is the mainstay of diagnosis and longitudinal monitoring for disease progression. Obstruction to airflow limits the volume of air that can be exhaled in one second (FEV1) and results in hyperinflation of the lungs and a decrease in forced vital capacity (FVC). A diagnosis is made when the ratio of the forced expiratory volume in one second compared with the forced vital capacity (FEV1/FVC) is less than 0.7.13 Diffusion of carbon dioxide from the circulation into the alveoli is impaired due to damage of the alveolar basement membranes and obstruction to exhalation, resulting in chronically elevated levels of carbon dioxide. Disease severity is determined by the reduction in FEV1, as established by the Global Initiative for Chronic Obstructive Lung Disease (GOLD) classification system:
- GOLD 1 (mild disease): FEV1 ≥80 percent predicted
- GOLD 2 (moderate disease): FEV1 between 50 and 80 percent predicted
- GOLD 3 (severe disease): FEV1 between 30 and 50 percent predicted
- GOLD 4 (very severe disease): FEV1 <30 percent predicted
The Global Initiative for Chronic Obstructive Lung Disease also provides severity and symptom-specific therapy recommendations. For patients with milder, less frequent symptoms (<1 moderate exacerbations/year), initial management includes maintenance bronchodilator therapy with either a long-acting muscarinic antagonist (LAMA), a long-acting beta-agonist (LABA), or a combined LAMA+LABA bronchodilator medication. A short-acting beta-agonist (SABA) is used for acute exacerbations. In patients with more severe disease, first-line therapy includes LABA+LAMA dual bronchodilator therapy and a SABA for acute dyspnea. An inhaled corticosteroid (ICS) treatment is added to the maintenance regimen in patients with high levels of eosinophils. The decision to initiate ICS therapy must be weighed against the increased risk of developing pneumonia.6 Patients who continue to experience frequent exacerbations while on first-line maintenance therapy may benefit from the addition of methylxanthine (Phosphodiesterase (PDE) 3 inhibitor), roflumilast (Phosphodiesterase (PDE) 4 inhibitor), azithromycin, or dupilumab (a human interleukin-4 and interleukin-13 monoclonal antibody). Patients who continue to experience significant dyspnea while on maintenance bronchodilator monotherapy may require escalation to LABA-LAMA therapy; those on dual long-acting bronchodilator therapy may benefit from a change in their inhaler device, participation in a pulmonary rehabilitation program, or the addition of ensifentrine (a dual PDE3 PDE4 inhibitor).13
Product Description
Ensifentrine (Ohtuvayre; Verona Pharma) is a dual inhibitor of PDE3 and PDE4 that results in bronchodilation, decreased inflammation, and improved ciliary function via activation of the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR). The medication is inhaled via a standard jet nebulizer twice daily. In June 2024, the US Food and Drug Administration approved ensifentrine for maintenance treatment of COPD in adults.14 This was the first ever market approval worldwide.
Evidence Analysis: Ensifentrine
Clinical Questions
The formulation of specific questions for the assessment recognizes that the effect of an intervention depends substantially on how it is delivered, to whom it is applied, the alternatives with which it is being compared, and the setting where it is used. To appraise the net health outcomes of ensifentrine, the DME MACs sought to address the following two key questions (KQ):
- KQ1: Is the evidence sufficient to conclude that ensifentrine combined with standard maintenance therapy will result in a net health benefit for Medicare beneficiaries with COPD?
- KQ2: Is the evidence sufficient to conclude that ensifentrine used as a stand-alone maintenance treatment will result in a net health benefit for Medicare beneficiaries with COPD comparable or superior to the available standard of care?
Literature Search
In its industry guidance for COPD drug development, the FDA recommends a minimum of three months follow-up to determine improvements in airflow obstruction, a minimum of six months to evaluate symptom relief, and a minimum of one year to assess exacerbation prevention.15 Consequently, we restricted our selection criteria to include RCTs with an endpoint measuring lung function improvement over at least three months or other outcomes with a minimum duration of six months. For harm outcomes, we included RCTs with a 4-week minimum duration.
The following additional PICO elements guided the literature search and study screening:
|
Population
|
Adults with COPD
|
|
Intervention
|
Ensifentrine (Ohtuvayre, Verona Pharma), 3 mg nebulized, twice a day
|
|
Comparator
|
Placebo or a proven therapy with a known effect
|
|
Outcomes
|
Lung function, exacerbations, respiratory symptoms, quality of life (QoL), treatment emergent adverse events
|
|
Study design
|
Efficacy: ≥ 12-week duration randomized-controlled trials (RCTs) or systematic reviews/meta-analyses of such trials
Harm: any RCT ≥ 4-week duration
|
Exclusion criteria comprised non-English articles and non-peer-reviewed literature, including conference abstracts.
A sensitive search of peer-reviewed publications indexed in PubMed was conducted on September 20, 2024. We used a combination of text and Medical Subject Headings (MeSH) terms such as "pulmonary disease, chronic obstructive," "RPL554," “ensifentrine” or “ohtuvayre.” The search returned 38 records. Based on title and abstract screening, 23 records were excluded, most of which were post-hoc analyses of the ENHANCE trials published as conference abstracts. Fifteen potentially relevant records were retrieved for full-text screening. Of these records, 10 were definitively excluded and 6 articles corresponding to 4 unique studies met the inclusion criteria. These studies included two phase II RCTs with 4-week follow-up16-19 and two phase III RCTs with ≥ 24-week follow-ups.20-24
Results
Phase III Trials
Summary
FDA approval of ensifentrine was based on evidence from two replicate phase III double-blind, placebo-controlled trials, which together enrolled 1,553 patients with moderate to severe COPD (ENHANCE-1, N = 760 and ENHANCE-2, N = 789).20,22,24 The trials were conducted in the US, Canada, South Korea, and 14 European countries. In both trials, participants were randomized in a 5:3 ratio to receive either 3 mg of ensifentrine or a placebo twice a day through a standard jet nebulizer. The primary endpoint, average FEV1 area under the curve (AUC)0–12 h, was selected to assess bronchodilatory effects over 12 hours. This measurement uses the trapezoidal method to calculate the AUC at predefined time points: predose and 30 min and 1, 2, 4, 6, 8, and 12 h postdose. Key secondary outcomes included respiratory symptoms and QoL measured over 24 weeks. Adverse events, including exacerbations, were recorded for all participants up to Week 24 and for a subset of participants in ENHANCE-1 up to Week 48.
Participants were current or former smokers between 40 and 75 years of age with a FEV1/FVC ratio of less than 0.70 and FEV1 between 40% and 80% of the predicted normal. Patients receiving dual or triple maintenance therapy were excluded. Across the two trials, 39% of participants were on no maintenance therapy, 61% on LABA or LAMA with or without ICS, and 18.5% on ICS alone. Most participants were white (92.5%).
Both trials met their primary endpoint of improving lung function as measured by the average FEV1 increase at 12 weeks (ENHANCE-1: 87 mL; 95% CI, 55-119; ENHANCE-2: 94 mL, 95% CI, 65-124). Importantly, ensifentrine was associated with 36% (ENHANCE-1) and 43% (ENHANCE-2) reductions in the annualized rates of moderate or severe COPD exacerbations (ENHANCE-1: rate ratio [RR] 0.64; 95% CI, 0.40-1.00; ENHANCE-2: RR, 0.57; 95% CI 0.38-0.87) as well as significant delays in time to first exacerbation (24 week endpoint: ENHANCE-1, hazard ratio [HR], 0.62; 95% CI, 0.39-0.97; ENHANCE-2, HR, 0.58; 95% CI, 0.38-0.87). Similar results were reported for the 48-week ENHANCE-1 extension. However, exacerbation reduction in ENHANCE-1 was marginally significant at both 24- and 48-week endpoints (P = .05 and P = .052, respectively), which suggests some degree of heterogeneity in participants’ responses to treatment.
Compared with placebo, the Evaluating Respiratory Symptoms Scale (E-RS)25 scores were reduced further in favor of ensifentrine at Week 24 in both trials. However, the between group difference was statistically significant in ENHANCE-2 only and did not reach the MCID (≥ 2-point improvement) in either of the two trials.
The St. George’s Respiratory Questionnaire (SGRQ),26 an airway disease-specific QoL tool, was used in both trials to assess perceived well-being and overall health. Compared to placebo, scores were reduced further in favor of ensifentrine at Week 24 in both trials. However, the difference was statistically significant in ENHANCE-1 only. In both trials, the differences between the ensifentrine and placebo groups were below the 4-point improvement MCID.
In a subsequent publication,21,24 the investigators reported on the effect of ensifentrine on dyspnea related to daily activities over 24 weeks using pooled data from the ENHANCE trials. Transition Dyspnea Index (TDI),27 an interview-based measurement of breathlessness, was used to measure changes in dyspnea severity from baseline. Responses were evaluated post-hoc by subgroups, including sex, age group, history of chronic bronchitis, exacerbation history, smoking status, presence of background medication, baseline eosinophil levels, and morning trough FEV1 improvement. At Week 24, significantly more patients in the ensifentrine group were TDI responders compared with the placebo group (OR, 1.82; 95%CI, 1.43-2.32; P < .001). This outcome was consistent across all analyzed subgroups (all p < .02) except for the LABA + ICS subgroup.
In a second pooled analysis, the investigators explored the efficacy and safety of ensifentrine in patients receiving LAMA (N = 285) or LABA + ICS (N = 272) specifically. In both groups, ensifentrine was associated with statistically significant lung function improvements, reduced exacerbation rates, and reduced dyspnea.28 These results should be interpreted with caution due to the post-hoc nature of the analyses.
Treatment-emergent adverse event (TEAE) rates, including severe TEAEs’ were similar in the ensifentrine and placebo groups in both ENHANCE trials over 24 and 48 weeks. Notable examples of TEAE’s leading to withdrawal from the trial include COPD exacerbations, COVID-19 infection, and non-COVID-19 pneumonia. Gastrointestinal adverse events observed with oral PDE4 inhibitors such as roflumilast29 were low and comparable in both groups.
Risk of Bias/Limitations
Robust methodologies underpinned both ENHANCE-1 and ENHANCE-2. For the objective endpoints (FEV1 and exacerbation rates), the results were judged to be at low risk of bias for each domain of the Cochrane risk-of-bias tool, version 2,30 except for the Missing Outcome Data domain in ENHANCE-2. This trial had withdrawal rates exceeding 20%, largely due to Coronavirus Disease-19 (COVID).26 The authors reported that a greater proportion of participants with severe COPD withdrew from the placebo group than from the ensifentrine group. Thus, there may have been an imbalance in disease severity between the two groups, resulting in a bias in the exacerbation rate ratio against the active intervention.
Both trials excluded patients on dual (LABA/LAMA) or triple (LABA/LAMA/ICS) maintenance therapy. Therefore, it remains unclear whether ensifentrine improves outcomes in such patients.
Additional RCTs
Two additional RCTs with a 4-week duration met our inclusion criteria for harm evaluation. Both were phase II multicenter double-blind multidose studies of comparable sizes (82 participants randomized to ensifentrine 3 mg in both). In Singh 2020 (NCT03443414),17,18 participants were instructed not to take any long-acting bronchodilators during the trial following a 7-14-day washout run-in period. In Ferguson 2021 (NCT03937479),16 all participants were treated with tiotropium (LAMA), and no other long-acting bronchodilators were allowed. Statistically significant improvements in lung function (average FEV1 and peak FEV1) were reported in both trials. According to FDA recommendations for the conduct of COPD trials, however, a 4-week study period is an insufficient duration for efficacy evaluation.15 As in the phase III trials, treatment-emergent adverse event (TEAE) rates, including severe TEAEs, were similar in both the ensifentrine and placebo groups.
Systematic Reviews, Meta-Analyses, and Health Technology Assessments
The Institute for Clinical and Economic Review (ICER) recently released a comprehensive evidence report on ensifentrine.31 Since the designs and populations of ENHANCE-1 and ENHANCE-2 were similar, meta-analyses were used to pool outcome data from the two trials. The ensifentrine pooled placebo-adjusted estimate for the increase in average FEV1, 92.3 mL, was statistically significant (95% CI, 66.2-118.4; P <.001; I2=0%). Compared to placebo, ensifentrine was associated with a statistically significant 40% decrease in the annualized rate of moderate-to-severe exacerbations (rate ratio [RR], 0.60; 95% CI, 0.41-0.79; P <.001; I2=0%). On week 24, ensifentrine was associated with a 40% placebo-adjusted delay in time to first exacerbation (HR, 0.60; 95% CI, 0.41-0.78; P <.0001; I2=0%). Compared to placebo the reduction in E-RS scores in the ensifentrine group was statistically significant but short of the MCID (-0.69; 95% CI: -1.38 to -0.01; P =.047; I2=0%). Regarding QoL, the SGRQ pooled estimate was not statistically significant and did not exceed the MCID (MD: 1.51; 95% CI, -3.13 to 0.12; P =.069; I2=22%).
ICER noted the uncertainty about the magnitude of benefits in patients with dual or triple maintenance therapy since they were excluded from the trial. Based on the results in patients on no or single therapy and the comparable incidence of serious TEAEs in the ensifentrine and placebo groups, the ICER investigators gave a B+ net health benefit rating, concluding that there is “high certainty that ensifentrine added to maintenance therapy results in at least a small net health benefit and may result in substantial net health benefit compared with maintenance therapy alone.”
Two systematic reviews with meta-analyses, both including the two ENHANCE trials and the two earlier phase II trials, were published shortly after the ICER review.32,33 Their analyses echoed those of ICER, concluding that ensifentrine improves lung function and patients' health status. One of the reviews utilized the GRADE approach, assigning “moderate certainty” evidence to most critical outcomes.32
Clinical Guidelines
The Global Initiative for Chronic Obstructive Lung Disease incorporated ensifentrine in its updated 2025 guidelines.13 Specifically, the organization assigned a “level A” to the evidence regarding lung function and dyspnea, while evidence related to health status was assigned a “level B.”
For patients with persistent breathlessness or exercise limitation on bronchodilator monotherapy, the organization recommends the use of two long-acting bronchodilators. If the addition of a second long-acting bronchodilator does not improve symptoms, the organization suggests either:
- Switching inhaler device or molecule
- Implementing or escalating non-pharmacological treatments
- Considering adding ensifentrine if available
For patients who need to be treated for both dyspnea and exacerbations, GOLD recommends adding roflumilast, azithromycin or dupilumab but not ensifentrine.
On February 2, 2024, we searched the following organization sites for guidelines published or updated after the FDA approval of ensifentrine. No guidelines matching these criteria were found on the following society sites:
- American Thoracic Society (ATS)
- American College of Chest Physicians (CHEST)
- American Academy of Family Physicians (AAFP)
Hypertonic Saline
Clinical Background
Muco-obstructive lung diseases are characterized by the presence of large amounts of viscous mucous in the airways. Alterations in mucus composition, hypersecretion, and/or deficits in ion-fluid transport result in thick, highly concentrated mucous leading to airway obstruction and inflammation, chronic cough, and persistent bacterial infections.34 While all diagnoses within this category share the clinical pattern of copious, thick airway mucous and airway obstruction, the prevalence of these diseases vary with regard to the number and demographics of the people affected.
Cystic fibrosis (CF) is the second-most common autosomal recessive genetic disease in the United States.35 It is caused by a mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. The CFTR gene regulates the secretion and absorption of chloride and bicarbonate in epithelial tissues; mutation in the CFTR gene results in dysregulation of anion secretion and hyperviscous mucous.36 Cystic fibrosis affects approximately 40,000 people in the United States.37 Patients from the Southern and Western United States experience higher mortality and hospitalization rates than CF patients in other parts of the country.35 There are sex-related differences in both diagnosis and outcomes. Females are diagnosed later in life than males with the disease, and experience a lower quality of life while living with CF. The median life-expectancy is approximately 40 years.36 Life expectancy for people living with CF has increased over the past several decades. Although the median age of death in 2023 was 37 years, people with CF born between 2019 and 2023 are predicted to have a mean survival age of 61 years due to new advances in treatment with the introduction of CFTR modulator tri-therapy called elexacaftor-tezacaftor-ivacaftor (ETI).38,39 Approximately 50% of patients taking ETI no longer expectorate sputum, negating the need for daily airway clearance therapies.35,36 Notably, adherence to respiratory therapy and chest physiotherapy is lower in older CF patients and those with advanced disease.40 Adherence to treatment with daily hypertonic saline is reported to be approximately 36%; by contrast, adherence to daily inhaled rhDNAse therapy is reported to be 62% due to the ease of therapy.41,42
Bronchiectasis is the result of an inability to clear the products of an immune response (neutrophils and other inflammatory mediators) to an airway infection. The most common etiologies of bronchiectasis are immunodeficiency, autoimmune disease, and focal airway obstruction due to tumor or foreign body. Environmental or occupational exposure to mold and other respiratory pollutants are factors associated with bronchiectasis and poorer disease management.43,44 The thick, viscous sputum of bronchiectasis differs from that of CF due to a high concentration of DNA, inflammatory mediators, and mucin. A diagnosis of bronchiectasis is made based upon clinical symptoms of daily cough with tenacious sputum, frequent exacerbations, and the presence of bronchial dilation on radiographic imaging. Although bronchiectasis may be diagnosed in patients of any age, the prevalence increases with age; a 2018 Medicare survey reported an annual prevalence of 701 per 100,000 beneficiaries over the age of 65 years.45 The prevalence and incidence have been increasing over time.46 Non-cystic fibrosis bronchiectasis (NCFB) is more common in adults over 75 years and in females.45
Primary ciliary dyskinesia (PCD) is caused by a congenital defect in the airway cilia that results in an inability to effectively clear mucous. Over 50 different genetic mutations have been identified that cause PCD. Common clinical manifestations of PCD include recurrent respiratory infections, rhinosinusitis, hearing loss,37 infertility, and differences in organ laterality. The prevalence of PCD is approximated at 1:7500 individuals across all races and ethnicities, with an average age at diagnosis of 5 years. Fewer than 200,000 people in the United States are estimated to have PCD48; however, the disease is likely underrecognized49 due to lack of access to diagnostic tools and a heterogeneity of clinical phenotypes. A recent genetic study (Hannah et al 2022) found the frequency of PCD varies by ancestry (1 in 9906 in people of African descent vs. 1 in 10,388 in European vs. 1 in 14,606 in people of East Asian descent vs. 1 in 16,309 in Latinos).49
Product Description
Hypertonic saline is a crystalloid solution of sodium chloride dissolved in water at a higher concentration of sodium than is normally present in blood. The high sodium concentration draws water into the airways and thins mucus, making it easier to expectorate and clear from the airways. It may also reduce airway inflammation in patients with muco-obstructive diseases.
Inhaled hypertonic saline is available in concentrations of 3% - 7% and is administered via a nebulizer device, with a mouthpiece preferred over a mask. The usual dose is 4ml once or twice per day.50 It is often combined with other inhaled mucous thinning medications and chest physiotherapy in a once or twice daily regimen.
Nebulizers are covered under the durable medical equipment (DME) benefit.51
Food & Drug Administration Designation
The FDA regulates hypertonic saline vials for inhalation under 21 CFR 868.5630 (Nebulizers).
- Design: Sterile, preservative-free Sodium Chloride Inhalation Solutions supplied in single use vials.
- Indication for use: Sodium Chloride Inhalation Solution, USP is used in conjunction with a nebulizer. The contents of these vials are for the induction of sputum production where sputum production is indicated (For ≥3% concentrations only)
- Patient Population: Any patient population where sputum production is indicated.52
Hypertonic saline solutions fall under the regulatory category of "approved" drugs based on long-standing medical use before the modern FDA drug approval process was established. Current products on the market are approved via the 510(k) pathway via predicate nebulized hypertonic saline products. Three products with varied sodium chloride concentrations (3.5%, 6%, 7%, and 10%) received clearance and served as predicates for the clearance of subsequent products through the 510(k) pathway.53
|
510(k) NUMBER
|
Cited as predicate
|
PRODUCT
|
COMPANY
|
APPROVAL DATE
|
|
K232523
|
|
Sodium Chloride Inhalation Solution USP 3% & 7%
|
Mankind Pharma Limited
|
2024-04-10
|
|
K210126
|
|
Sodium Chloride Inhalation Solutions 3% 3.5% 7% and 10%
|
The Ritedose Corporation
|
2021-12-03
|
|
K120051
|
|
Sodium Chloride Inhalation Solutions 3% 7% AND 10% USP-4ML
|
Nephron Pharmaceuticals Corp.
|
2012-04-20
|
|
K101424
|
Yes
|
Inhaled saline solutions - 3%, 3.5%, 6%, 7%, and 10%
|
PharmaCaribe
|
2010-08-07
|
|
K070498
|
Yes
|
Inhaled saline solutions - 3.5%, 6% and 7%
|
Pari Innovative
|
2007-04-26
|
|
K972778
|
Yes
|
Sodium chloride 3% and 10% used with a nebulizer
|
Dey-Laboratories
|
1997-10-08
|
|
K884359
|
|
Unidose 3% Sodium Chloride Solution for inhalation
|
Automatic Liquid Packaging Inc.
|
1988-12-20
|
Summary of Evidence: Hypertonic Saline
Cystic Fibrosis
Primary studies
Nebulized hypertonic saline is commonly utilized by both adults and children with cystic fibrosis (CF) across numerous countries worldwide. Widespread use has been reported in many European countries, where it is typically covered by health insurance.54 Extensive utilization has also been reported in the US. In a survey conducted among US adults and parents of children with CF, 88.5% of respondents indicated they or their children used hypertonic saline daily.55
Three landmark randomized controlled trials (RCTs) are commonly cited in support of the use of HS in patients with CF.56-58 With the addition of an infant trial that did not reach its primary and secondary endpoints,59 these studies are the largest double-blind RCTs of HS in CF currently available.
Since no inhaled agent is completely inert, conducting a true placebo-controlled study of nebulized HS is not feasible.59 Nebulized isotonic saline (IS) has been established as an appropriate control in clinical trials of HS.60,61 It has been utilized as a substitute for a placebo in most blinded RCTs of HS, regardless of the indication.
In 2006, Elkins et al published the findings of a multicenter double-blind, parallel-group trial that investigated the long-term effects of HS in a mixed group of children (aged over 6 years) and adults with CF.57 One hundred sixty-four participants were randomized 1:1 to twice-daily administration of either HS (7%) or IS (0.9%) over 48-weeks. The mean age was approximatively 18±9 years in both groups and the mean percent of predicted forced expiratory volume (ppFEV1)62 at baseline was 73 ± 21% in the HS group and 76 ± 21% in the control group. The 48-week rate of change from baseline in lung function (primary outcome measured as a composite of spirometric parameters) did not differ significantly between groups. However, the mean number of exacerbations requiring intravenous antibiotics per patient over 48 weeks was significantly reduced in the HS group compared to the control group (RR 0.56; 95%CI, 0.14-0.86; P=0.02). Similarly, the mean number of days participants experienced exacerbations decreased by an additional 11 days in the HS group compared to the control one (95% CI, -3 to -19 days; P=0.02). Moreover, participants in the HS group experienced significantly fewer days of missed school or work (7, IQR 0 to 21 days vs 24, IQR 12 to 48, P<0.001). Treatment-related adverse events (TRAEs) were significantly more frequent in the HS group (15.5% vs .2%; P=0.001). The most common TRAEs included cough, chest tightness, and pharyngitis.
In 2016, Dentice et al published the results a double-blind RCT evaluating the efficacy of HS in adults experiencing exacerbations.58 One hundred thirty-two adults hospitalized with CF exacerbations were randomized to inhale either 7% HS or 0.12% hypotonic saline three times a day. The primary outcome, length of stay, was 12 days in the treatment group and 13 days in the control group. The mean one-day difference was below the minimal 2 day expected and did not reach statistical significance (95%CI, 0-2, p=0.07). However, patients in the HS group were significantly more likely to regain their pre-exacerbation ppFEV1 levels (75% vs 57%, p<0.05), and experienced significantly greater reduction in congestion and dyspnea severity (0-100 scale: mean difference [MD],10; 95% CI 3-18 and MD, 8; 95%CI 1-16, respectively).
In 2019, Ratjen et al published the findings of a multicenter double-blind, parallel-group trial involving 150 children with CF aged 3 to 6 years.56 Participants were randomized 1:1 to twice-daily administration of either 7% HS or 0.9% IS over 48-weeks. The primary outcome measure was the change in lung clearance index (LCI2.5) from baseline to week 48. The LCI2.5 measures the number of breaths required to reduce the exhaled nitrogen concentration to one fortieth of the starting concentration during a maximum breath washout test. At 48 weeks, HS was associated with a small statistically significant improvement (i.e., decrease) in LCI2.5 compared with IS (MD, –0.63 LCI2.5 units, 95%CI, –1.10 to –0.15, p=0.01). There was no difference between groups in the frequency of adverse events. Reported serious adverse events (SAEs) included cough (3% with HS vs 4% with IS), gastrostomy tube placement or rupture (3% vs 1%), upper gastrointestinal disorders (1% vs 3%), distal intestinal obstruction syndrome (1% vs 1%), and decreased pulmonary function (0 vs 1%). None of these SAEs were judged to be treatment-related.
A parallel study with an identical design conducted by the same team investigated whether HS reduces structural lung disease by comparing chest computed tomography (CT) scans from baseline to 48 weeks.63 Chest CTs were scored using the Perth-Rotterdam Annotated Grid Morphometric Analysis for Cystic Fibrosis (PRAGMA-CF), which measures the percentage of total lung volume occupied by abnormal airways.64 Baseline and 48 week chest CTs were available for 49 participants in the HS group and 55 in the IS group. Mean PRAGMA-CF % disease at 48 weeks was significantly lower in the HS group compared to the IS group (MD, 0.67, 95% CI 0.26–1.08; p=0.009).
The largest double-blind trial involving infants with CF was published in 2012.59 It enrolled 321 infants and young children aged 4 to 60 months across 30 centers. The trial failed to reach its primary endpoint–a decrease in exacerbations–or any secondary endpoints (height, weight, respiratory rate, oxygen saturation, cough, or respiratory symptom scores). Participants had been randomized to 7% HS or 0.9% IS inhaled twice daily for 48 weeks. There were no significant differences between groups in the proportion of participants experiencing SAEs. The most common SAE in both groups was cough or increased cough, occurring in 8% of participants in the HS group and 10% in the IS group.
The Cystic Fibrosis Foundation (CFF) recently sponsored a large multicenter study examining whether the treatment burden of patients with CF on ETI could be safely reduced by discontinuing either HS or dornase alfa (DA).65 The study, named SIMPLIFY, encompassed two non-inferiority open label RCTs, each independently evaluating the effect of discontinuing versus continuing HS (Trial A) or DA (Trial B). The six-week study enrolled collectively more than 800 participants aged 12 and older between 2020 and 2022. The overall average ppFEV1 was 96.9%. At six weeks, discontinuing either HS or DA did not result in clinically meaningful changes in ppFEV1 and LCI2.5 among participants with relatively good lung function. However, within the small group of adults with ppFEV1, 60 to 70% more respiratory adverse events were reported in those randomized to stop any of the two inhaled medications. The authors concluded that greater caution should be applied when considering changes to daily therapy in individuals with more advanced CF pulmonary disease.
Systematic Reviews
A recent Cochrane systematic review (SR) concluded that the evidence regarding the efficacy of HS in improving lung function in the overall patient population with CF aged 12 and over is uncertain. However, the authors deemed the evidence sufficient to conclude that HS is an effective adjunct to physiotherapy during acute exacerbations of lung disease in adults.66
Similarly, Zhang et al found no statistical difference in ppFEV1 between HS and non-HS groups in a recent meta-analysis that included 12 studies involving participants with CF of all ages. However, the meta-analysis of the outcome data from the two studies reporting on mucociliary clearance (MCC in 90 minutes) showed a significant improvement in favor of HS (MD, 10.5; 95% CI 6.5-14.4). Additionally, the meta-analysis of the data from the five studies reporting on quality of life (QoL) also showed a significant improvement in the HS group compared to the IS group (standardized mean difference [SMD], 0.44, 95% CI 0.02-0.87).67
Practice Guidelines
Cystic Fibrosis Foundation
Infants
For infants with CF under 2 years of age, 7 percent hypertonic saline may be used in symptomatic infants. Certainty: Low, Benefit: Moderate, Consensus recommendation.68 Last reviewed July 2021.
Preschool Children
Hypertonic saline and dornase alfa may be selectively offered based on individual circumstances. Grade: C, Certainty: Moderate, Benefit: Moderate.41 Last reviewed July 2021.
Children Aged Six Years and Older, and Adults
For individuals with CF, 6 years of age and older, the CF Foundation recommends the chronic use of inhaled hypertonic saline to improve lung function and quality of life and reduce exacerbations. Moderate Certainty of Net Benefit, estimate of net benefit Moderate, Recommendation “B” (the committee recommends clinicians routinely provide the therapy).70,71 Last reviewed 2021.
CFF Otolaryngology Care Multidisciplinary Consensus Recommendations
Per CFF guidelines, individuals with CF benefit from the use of nebulized dornase alfa and nebulized hypertonic saline. Based on these recommendations, individuals with CF who undergo endoscopic sinus surgery should have access to perioperative airway clearance therapy, nebulized dornase alfa, and nebulized hypertonic saline perioperatively. These therapies could be beneficial to avoid exacerbation in the perioperative period when pain medication and sedation is necessary.72 Published in 2022.
National Institute for Health and Care Excellence (NICE, UK)
Cystic fibrosis: diagnosis and management NICE guideline [NG78]. Last reviewed, 2024.
NICE recommends HS as a second choice if there is an inadequate response to dornase alfa.73
- 1.6.18. Offer rhDNase (dornase alfa) as the first choice of mucoactive agent.
- 1.6.19. If clinical evaluation or lung function testing indicates an inadequate response to rhDNase, consider both rhDNase and hypertonic sodium chloride or hypertonic sodium chloride alone. Last reviewed, 2024.
Non-Cystic Fibrosis Bronchiectasis
Primary Studies
A recent registry study revealed HS is commonly used by US patients with non-cystic fibrosis bronchiectasis (NCFB). The study evaluated the utilization of HS and positive expiratory pressure (PEP) devices in this population by analyzing patient data from the United States Bronchiectasis and Nontuberculous Mycobacterial (NTM) Lung Disease Research Registry (BRR). From an initial pool of 4670 patients diagnosed with NCFB, the records of 1113 patients using other forms of chest therapy and 1362 patients with conflicting data were excluded. Among the remaining 2195 patients, one-third used HS daily, with or without concomitant PEP.74
The largest RCT in adults with NCFB, which included 40 participants, was published in 2012. No significant differences were found in the number of exacerbations, hospital admissions, lung function tests, and QoL between the 6% HS and 0.9% IS groups after 12 months of use. One participant in the HS group experienced chest tightness while inhaling, which improved after treating an underlying acute exacerbation.75
In a cross-over RCT involving 28 adult patients, published around the same period, ppFEV1 and FVC improved significantly more in the 7% HS group compared to the 0.9% IS group after three months of daily use (change from baseline: 15.1% vs 1.8% p<0.01 and 11.2% vs 0.7%, p<0.01, respectively). Quality of life, as measured by the St. George's Respiratory Questionnaire (SGRQ), also improved significantly more with HS compared to IS (change from baseline: 6.0 points vs 1.2-point, p<0.05).76
A triple-arm, double-blind, randomized crossover trial published in 2018 compared three solutions: 0.1% hyaluronic acid added to 7% HS (HS +HA), 7% HS alone (HS), and 0.9% isotonic saline (IS). Twenty- three adult participants received each solution in single sessions separated by a 7-day washout period. Sessions one and two included 30 minutes of airway clearance techniques. The primary outcome measure, sputum weight, was recorded at each session and at 24-hour follow-ups. Quality of life, assessed using the Leicester Cough Questionnaire (LCQ), and lung function were measured both before and after each treatment. Sputum collected over the 24-hour follow-ups tended to be lighter in weight in the HS and HA+HS groups compared with the IS group (HS vs IS: -1.7g, 95%CI -4.2g to 0.0g; HA+HS vs IS -1.1g, 95%CI -3.6g to 0.7g). However, the differences were not statistically significant. The most common adverse events reported by participants were coughing and throat irritation. These symptoms were especially prevalent after inhaling HS, and to a lesser extent, after inhaling HA+HS. Three small sputum samples containing blood were observed from two participants: one during the use of the IS solution, and the other during the use of both the HS and IS solutions.77 As with all the other RCTs of HS involving patients with NCFB, these favorable or unfavorable results should be interpreted carefully because of the study’s small size.
The only RCT evaluating HS in pediatric patients with NCFB was a crossover trial involving 52 children aged five to 15 years. Participants were randomized to receive either 200 µg of inhaled salbutamol followed by 3% HS nebulization or 200 µg of inhaled salbutamol alone for 8 weeks. After a 30-day washout period participants were crossed over to the other arm. Participants receiving 3% HS showed significantly greater improvement in ppFEV1 compared to patients receiving salbutamol alone (pre-crossover: 14.2% vs 5%, p<0.01; post-crossover: 10.8% vs 3.5%, P<0.01). The first group of HS recipients experienced significantly fewer exacerbations than the control group, though this difference was not significant post-crossover. While the study was welcomed due to the paucity of research in pediatric patients with NCFB, it has been questioned for its lack of blinding and absence of within-subject analytical comparison.78
Systematic Reviews
In 2020, Xie et al published an SR with meta-analyses assessing the effects of inhaled HS vs IS in treating adult patients with NCFB. The review included the three RCTs mentioned above plus a small pilot RCT published in 2005.79 Compared to IS, HS showed no significant impact on FV1 (SMD, 0.12; 95% CI, -0.06 to 0.30; P=0.18), FVC (SMD, 0.10; 95% CI, -0.09 to 0.28; p=0.30), sputum expectoration (SMD, -0.03; 95% CI, -2.73 to 2.68; P=0.99), or Leicester Cough Questionnaire (LCQ) scores (SMD, 0.15; 95% CI, -0.89 to 0.58; p=0.68).80
Practice Guidelines
While several US guidelines for managing bronchiectasis associated with CF are available, there are no US guidelines yet for managing NCFB although one is currently being developed.81 Other regions, including Europe, have published guidelines for NCFB management, but the recommendations are based on evidence of low quality or limited in scope.81
The European Respiratory Society guidelines for the management of children and adolescents with bronchiectasis suggests not using HS routinely in pediatric populations (conditional recommendation, very low quality of evidence) with some exceptions. The guideline states that 6–7% HS may be considered in selected patients with high daily symptoms, frequent exacerbations, difficulty in expectoration and/or poor QoL. The panel judged that HS could improve QoL and facilitate expectoration if well tolerated. Children should be old enough to tolerate the intervention. The panel also considered that short-acting β2-agonists should be used prior to inhaling HS and the first dose administered under medical supervision.82
The European Respiratory Society guidelines for the management of adults with bronchiectasis suggest offering long-term (≥3 months) mucoactive treatment, including HS but not DA, in adult patients with bronchiectasis who have difficulty in expectorating sputum and poor quality of life and where standard airway clearance techniques have failed to control symptoms (weak recommendation, low quality of evidence). The society issued a strong recommendation against offering DA to adult patients with bronchiectasis.83
Primary Ciliary Dyskinesia
Primary Studies
The only randomized trial of hypertonic saline (HS) in primary ciliary dyskinesia (PCD) we identified was a double-blind crossover trial published in 2017. Twenty-two adult participants were randomized to either 7% HS or 0.9% IS twice daily for 4 weeks. After a 4-week washout period, participants switched solutions for another 4 weeks. The primary outcome was the change in QoL as measured by the SGRQ. Secondary outcome measures included SGRQ sub scores, the QoL Questionnaire-Bronchiectasis (QOL-B), lower respiratory tract symptoms, exacerbations, spirometry results, inflammatory markers in the body and sputum, adherence, and adverse events. There were no significant differences in SGRQ total score change from baseline at 4 weeks between the HS and IS groups (−2.6, IQR -9.0, -1.5 points; −0.3, IQR, -8.1, -6.1 points, respectively). Most secondary outcomes were either not statistically significant or did not meet the minimum clinically important difference. Adverse events were more frequent with HS, though they were generally mild. These included throat irritation, cough, and chest tightness.84 These negative results should be interpreted with caution due to the small size of the study’s sample.
Practice Guidelines
PCD Foundation
Based on evidence obtained from NCFB studies, the 2016 PCD Foundation Consensus Recommendations suggest HS may be used after physicians instruct patients in proper equipment sterilization.85
European Respiratory Society
The European Respiratory Society (ERS) last consensus statement for the treatment of PCD was a pediatric guideline issued in 2006. It stated that nebulized HS may theoretically be effective in increasing mucus clearance in patients with PCD. Noting the quasi absence of clinical research in PCD, ERS is currently developing a set of core outcomes for clinical research in the condition.83