Skip to main content

Treating sleep-disordered breathing of idiopathic pulmonary fibrosis patients with CPAP and nocturnal oxygen treatment. A pilot study

Sleep-disordered breathing treatment in IPF

Abstract

Introduction

Sleep-disordered breathing (SDB) is a major comorbidity in idiopathic pulmonary fibrosis (IPF) and is associated with a poor outcome. There is a lack of knowledge regarding the impact of SDB treatment on IPF. We assessed at one year: (1) the effect of CPAP and/or nocturnal oxygen therapy on IPF regarding lung function, blood mediators, and quality of life; (2) adherence to SDB treatment and SDB changes.

Methodology

This is a prospective study of consecutive newly diagnosed IPF patients initiating anti-fibrotic treatment. Lung function, polysomnography, blood tests and quality of life questionnaires were performed at inclusion and after one year. Patients were classified as obstructive sleep apnoea (OSA), central sleep apnoea (CSA), and sleep-sustained hypoxemia (SSH). SDB therapy (CPAP and/or nocturnal oxygen therapy) was initiated if needed.

Results

Fifty patients were enrolled (36% had OSA, 22% CSA, and 12% SSH). CPAP was started in 54% of patients and nocturnal oxygen therapy in 16%. At one-year, polysomnography found improved parameters, though 17% of patients had to add nocturnal oxygen therapy or CPAP, while 33% presented SDB onset at this second polysomnography. CPAP compliance at one year was 6.74 h/night (SD 0.74). After one year, matrix metalloproteinase-1 decreased in OSA and CSA (p = 0.029; p = 0.027), C-reactive protein in OSA (p = 0.045), and surfactant protein D in CSA group (p = 0.074). There was no significant change in lung function.

Conclusions

Treatment of SBD with CPAP and NOT can be well tolerated with a high compliance. IPF patients may exhibit SDB progression and require periodic re-assessment. Further studies to evaluate the impact of SDB treatment on lung function and serological mediators are needed.

Introduction

Idiopathic pulmonary fibrosis (IPF) is a fatal progressive fibrotic interstitial lung disease (ILD) [1,2,3]. Antifibrotic drugs slow the decline of lung function [4] and improve prognosis, which remains poor [5]. Sleep-disordered breathing (SDB) is a highly prevalent IPF comorbidity, ranging from 45 to 90% depending on the methodology used [6,7,8,9,10,11]. SDB involves different entities, and while obstructive sleep apnoea (OSA) and nocturnal hypoxemia have been associated with poor outcomes in IPF [6, 12, 13], the impact of central sleep apnoea (CSA) remains poorly understood [14, 15].

OSA and intermittent hypoxemia have been associated with ILD and an increment in the blood mediators of pulmonary interstitial remodelling and alveolar epithelial damage [14, 16,17,18]. The progression of pulmonary fibrosis has been associated with the development of SDB through a mechanism that is still under debate [14, 19,20,21].

The treatment of SDB in IPF patients seems to improve quality of life and prognosis despite the scarce number of studies [11, 22, 23]. Some methodological limitations include the absence of a generalized use of antifibrotic drug therapy [4, 5] and self-selection bias owing to a high refusal rate and poor compliance with continuous positive airway pressure (CPAP) [24]. There is limited data regarding the effect of using nocturnal oxygen therapy (NOT) in ILD patients and, to the best of our knowledge, there is no data regarding the effect of treating CSA in patients with ILD [25,26,27].

Therefore, this study of IPF patients aimed to assess: (1) the impact of personalized SDB treatment by CPAP and/or nocturnal oxygen therapy on lung function, blood mediators and quality of life; (2) tolerance of and adherence to SBD treatment and sleep-disordered breathing changes after one year of treatment and follow-up.

Methodology

This is a prospective pilot study of consecutive newly diagnosed IPF patients who were systematically studied for SBD and treated with CPAP and/or NOT depending on the type of SDB. Inclusion criteria was a confident IPF diagnosis by a multidisciplinary committee [2, 3]. Exclusion criteria included other life-threatening or unstable diseases.

Polysomnography (PSG), pulmonary function test, 6-minute walking test (6MWT), blood test, and questionnaires were performed at baseline and after one year. SDBs were classified as previously described [7]: (A) OSA when the apnoea-hypopnea index (AHI) was ≥ 15/h and obstructive events were predominant (≥ 50%); (B) CSA when AHI ≥ 15/h and there were predominant central events; (C) sleep sustained hypoxemia (SSH) when total sleep time under SpO2 88% (TST88) > 5 min and AHI < 15/h; (D) No-SDB when AHI < 15/h and TST88 ≤ 5 min. PSG at one year was performed under treatment if the patients was receiving it or basally if no treatment had been started.

Blood mediators included C-reactive protein (CRP), lactate dehydrogenase (LDH), N-terminal pro-B-type natriuretic peptide, interleukin 6 (IL-6), matrix metalloproteinase-1 (MMP-1), MMP-7, MMP-9, surfactant protein D (SP-D), tenascin-c large, Krebs von den Lungen 6 (KL-6), advanced glycation end-products (AGEs), and receptor of AGEs (RAGEs). Sleep quality and sleep habits questionnaires, the Epworth Sleepiness Scale (ESS), the Functional Outcomes of Sleep Questionnaire short version, EuroQol 5D-5 L, and the King’s Brief Interstitial Lung Disease questionnaire were analysed. Comorbidities were assessed through the Beck Depression Inventory-II, the 7-item Generalized Anxiety Disorder Scale and the Gastroesophageal Reflux Disease Questionnaire. All patients received anti-fibrotic medication (nintedanib or pirfenidone) after the diagnosis and at least two weeks before the first blood sample was obtained.

The SDB treatment decision was based on the particular SDB (Fig. 1). Patients with moderate or severe OSA were treated with CPAP after a home autoCPAP titration [28, 29]. For those with moderate or severe CSA the treatment was individualized: they underwent manual CPAP titration and started CPAP if events decreased ≥ 50% [30]; if events did not decrease > 50%, servoventilation or oxygen therapy were considered and decided in each individualized case depending on IAH severity and the degree of nocturnal hypoxemia. Patients with nocturnal hypoxia using the criterion TST88 ≥ 5 min, started nocturnal oxygen therapy (NOT) [31, 32]. After one month of SDB treatment, patients with TST88 ≥ 5 min were re-assessed by nocturnal pulse-oximetry. Clinical follow-up following the standard of care assessment for IPF treatment and the SDB treatment by specialist nurses was performed at one, three, six and 12 months.

Fig. 1
figure 1

Proposal for the treatment of sleep-disordered breathing in patients with idiopathic pulmonary fibrosis. OSA: obstructive sleep apnoea; CSA: central sleep apnoea; AHI: apnoea-hypopnea index; TST88: total sleep time under SpO2 88%; CPAP: continuous positive airway pressure; O2: supplemental oxygen

Statistics

Continuous variables were presented with mean and standard deviation, except for those with asymmetry or lack of normality, which were described with median and interquartile range. Categorical variables were reported as the number of cases and the percentage of the total. Paired t-tests were used for normally distributed variables, the Wilcoxon signed-rank test for non-normally distributed variables and the McNemar test for categorical variables. Using logistic regression models, the crude effect and the effect adjusted by forced vital capacity (FVC), diffusing capacity for carbon monoxide (DLCO) and age were analysed. Statistical analyses were performed with R software version 4.2.1 or higher and STATA 17.

Results

Study population

Fifty patients were enrolled (Table 1) and 72% were male. Mean age was 72.6 years and only 28% of cases were obese (BMI > 30 kg/m2). The sleep study identified 70% of patients as having some type of SDB encompassing 36% OSA, 22% CSA and 12% SSH. The baseline characteristics have been previously reported in more detail [14]. Importantly, CSA cases were detected by coding non-obstructive hypopneas and these patients presented a predominance of hypopneas, compared to apnoeas, and did not present periodic breathing. CPAP was started in all OSA patients and 82% of CSA patients (nine patients), while NOT was initiated in SSH patients and 18% of CSA patients (two patients) (Table 2). Two patients died during follow-up (one OSA and one CSA).

Table 1 Characteristics of the study population at baseline
Table 2 Sleep-disordered breathing treatment

Changes in lung function, blood mediators and QoL

Data are summarized in Table 3 according to SBD classification. At baseline, the mean FVC was lower in the SSH group (below 80%). After one year, lung function did not significantly change. Regarding the higher serum concentration of MMP-1 at baseline (Fig. 2) in the OSA and CSA groups, a significant decrease was observed after one year (p = 0.029 and p = 0.027, respectively). Furthermore, patients with OSA also showed a significant decrease of CRP in blood samples (p = 0.045) and the CSA group showed a trend to reduction of SP-D (p = 0.07). No significant differences were found when comparing the 1-year changes in lung function and blood mediators with the non-SBD group. The questionnaires (Supplemental Table 1) showed a significant improvement in ESS in OSA patients, with no other relevant changes.

Table 3 SDB groups. All data are presented as Median [Q1;Q3]
Fig. 2
figure 2

Blood biological mediators at baseline and at one year by type of SBD. Outsider values were excluded. OSA: obstructive sleep apnoea; CSA: central sleep apnoea; SSH: sleep-sustained hypoxemia; No- SDB: No sleep-disordered breathing; MMP-1: matrix metalloproteinase 1; SP-D: surfactant protein D ; CRP: C-reactive protein

Polysomnographic changes

The PSG study under SDB treatment at 1-year compared to the baseline showed a significant decrease in AHI and TST90 in the OSA and CSA groups, and a non-significant increase in mean oxygen saturation in the SHH group. The No-SDB group showed a significant decrease only in lowest oxygen saturation in basal PSG at one year. Concerning sleep phases, only the OSA group had a significant decrease in N1 (% TST).

The PSG study under SBD treatment at one year also revealed the need to add NOT to CPAP in two out of 16 OSA group patients and three out of nine patients in the CSA group (Table 2). CPAP was added to one patient in the SSH group initially treated with NOT. In the non-SDB group, the sleep study was repeated without treatment and it disclosed the existence of SDB in five out of 15 patients, requiring starting CPAP in three and NOT in two patients. The changes at one year in FVC, DLCO and weight in this group were not predictive of SDB progression in a logistic model.

SDB treatment compliance, side effects and patient satisfaction

CPAP compliance at one year was 6.74 h/night (SD 0.74) and all patients had a compliance higher than 5 h/night. Mean CPAP pressure was 7 mmHg [IQR 6;9] (OSA group: 7 mmHg [IQR 6;8] and CSA group: 8 mmHg [IQR 6;9]). The main reported adverse effect (Supplemental Table 2) was dry mouth, but no treatment discontinuation was associated with side effects. Regarding the Patient-Reported Experience Measures (PREMs) (Supplemental Table 3), satisfaction with the sleep study and SDB management was nearly 100%.

Discussion

This pilot study highlights the fact that the personalized management and treatment of SDBs with CPAP and NOT in patients with IPF is feasible, resulting in high CPAP compliance, few side effects and a high level of satisfaction. Most patients with SDBs do not present typical symptoms and a SDB may appear at any time during the follow-up.

There is very limited evidence regarding the treatment of SBD in IPF patients, evidence for OSA is scarce [33]; it was anecdotal for nocturnal hypoxia in ILD [25,26,27], and absent for CSA. Therefore, our proposal was a proof of concept treating each condition individually according to the recommendations of the respective guidelines, and applying a personalized approach in order to achieve the optimal correction of each sleep disorder. Therefore, OSA was treated with CPAP with good results. As the treatment of CSA is not established for patients with FPI [30], as a general rule, we treated it with CPAP if manual CPAP titration decreased events ≥ 50%. This objective was not achieved in only two patients who had low basal IAH (16.0/h and 21.1/h) and a TST88 ≥ 5 min and who received nocturnal oxygen, achieving a good correction in control nocturnal oximetry. For this reason, no patient was submitted to serventilation. Patients with SSH were treated with NOT [32, 34]. This approach, and the cut-off points used to initiate each treatment, should be validated in future multicentre studies and larger real-world settings.

There were no significant differences in lung function after one year. Other studies treating patients with IPF and OSA by CPAP did not measure pulmonary function, but an improvement in survival was reported [11, 22]. However, a longer follow-up of larger a larger cohort and a control group would be required to better assess the potential impact of SDB treatment on IPF outcomes.

Increased levels of oxidative, inflammatory and profibrotic serological mediators were observed in IPF patients with SDB in previous studies [10, 14, 16,17,18]. Our previous study characterized different SDBs in IPF patients and their relationship with lung function and blood mediators [14]. We hypothesized that different SDBs may be associated with distinct metabolic pathways. Apnoea groups presented higher levels of MMP-1 at baseline, which decreased significantly after one year of SDB treatment. MMP-1 is a profibrotic metalloproteinase related to hypoxia-inducible factor 1 (HIF-1) [35], which is increased in intermittent hypoxia, but not in sustained hypoxia [36]. Furthermore, OSA and CSA present certain differences. As for non-ILD OSA patients [37], CRP was reduced after one year in the OSA group. On the other hand, SP-D was reduced to a figure close to statistical significance (p = 0.07) in CSA patients. SP-D is a blood mediator of epithelial alveolar tissue [38] associated with an increased death risk rate in IPF patients [39]. SP-D may be up and down-regulated by HIF-1 in acute and persistent hypoxia, respectively [40], and higher levels of hypopnea index were associated with a decrease in SP-D blood levels [41]. However, it is important to note that our study has an exploratory character and these changes cannot be extended to the treatment of SDB because many confounding factors, such as the initiation of antifibrotic treatment, may lead to erroneous conclusions. Future prospective randomized studies with longer follow-ups should evaluate these findings.

On the other hand, there is mounting evidence that non-physiological mechanical lung strain can produce an activation of pro-inflammatory and pro-fibrotic pathways that results in the progression of pulmonary fibrosis [42, 43]. Therefore, the long-term safety of CPAP treatment depending on the required positive end expiratory pressure (PEEP) levels should be considered in the future. Our data showed no significant changes in lung function. In addition, apnoea and hypopnea events in IPF patients have been also suggested as a pro-fibrotic mechanism [21, 44, 45]. CPAP treatment could reduce repetitive lung strain and hypoxia-related damage. The accurate estimation of low CPAP pressure required may also have been a key factor in preventing mechanical damage and reducing the harmful effect of SDBs [43].

The high CPAP acceptance and compliance shown by the patients in our study contrasts with previous studies with IPF patients. In two similar studies, 8% and 16% of IPF candidate patients withdrew from CPAP treatment within the first month [11, 23]. In line with our data, a recent study did not report any CPAP refusal or discontinuation [22]. Regarding CPAP compliance, 33% and 62% reported poor adherence (≥ 4 h/night) [11, 22]. Conversely, none of the patients in our series, had an adherence of less than five hours per night. The reasons for this good SDB treatment compliance are unknown, as they were not the focus of this study. However, a combination of comprehensive information on SDB, personalized SDB treatment and a close follow-up with an assessment of side effects may have had an influence.

One-third of patients without baseline SDB had some SDB at one year of control and had to start treatment, while 17% of patients treated for SBD had to increase their SDB treatment only one year later. Surprisingly, that was not associated with lung function decline or weight change. If this finding is confirmed in further series, sleep studies should be considered not only at diagnosis [8,9,10, 46, 47], but also during follow-up.

There were no significant differences in quality of life at one year. The COVID-19 outbreak may have negatively impacted quality of life results, which may explain the differences with regard to previous studies [11, 22, 23]. However, the satisfaction score and PREMs of sleep screening, treatment and follow-up were high, suggesting that the process does not cause great inconvenience to patients.

Strengths and limitations

Limited sample size is the main limitation of this exploratory pilot study and constrained the statistical analyses, particularly when examining distinct SDB types such as the SSH group. Furthermore, although the enrolment of all patients after initiating antifibrotic drugs better enabled the assessment of SDB outcomes, its effect on respiratory function and serum mediators could not be completely attributed to SDB treatment and the results should be considered with caution before being properly validated. All patients were treated with antifibrotics, whereas previous studies did not report or reported only partially on the use of antifibrotic drug treatment [11, 22, 23]. The holistic diagnosis and management of SDB is another remarkable strength of the study, since it does not preclude different SDBs such as CSA [22], or treating nocturnal hypoxia with NOT and not only OSA by CPAP [11, 22, 23]. Adherence may impact SDB outcomes, but due to the high level of compliance among the included patients, we were not able to analyse outcomes depending on the level of adherence. Finally, these results must be interpreted with caution, not only due to the limited number of patients, but also other potential confounding factors such as the absence of a control group with SDB but without treatment.

Conclusions

Our study suggests that a personalized selection of SDB treatment including CPAP and/or NOT involving a comprehensive follow-up of the therapy may be well-accepted and achieve high compliance. SDB may appear or increase SDB treatment requirements only one year after their study. Therefore, not only sleep studies in IPF patients should be considered, but also periodic re-assessment. Future randomized and longer follow-up studies are needed to evaluate the impact of SDB treatment on both lung function and biological mediators.

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Abbreviations

IPF:

Idiopathic pulmonary fibrosis

ILD:

Interstitial lung diseases

SDB:

Sleep-disordered breathing

OSA:

Obstructive sleep apnoea

CSA:

Central sleep apnoea

CPAP:

Continuous positive airway pressure

NOT:

Nocturnal oxygen therapy

6MWT:

6-minute walking tests

PSG:

Polysomnography

SSH:

Sleep-sustained hypoxemia

AHI:

Apnoea-hypopnea index

TST:

Total sleep time

TST90/88:

Total sleep time under SpO2 90% /88%

CRP:

C-reactive protein

LDH:

Lactate dehydrogenase

NT-BNP:

N-terminal pro-B-type natriuretic peptide

IL-6:

Interleukin 6

MMP:

Matrix metalloproteinase

SP-D:

Surfactant protein D

FNIII-C:

Tenascin-c large

KL-6:

Krebs von den Lungen 6

AGEs:

Advanced glycation end-products

RAGEs:

Receptor of advanced glycation end-products

ESS:

Epworth sleepiness scale

FOSQ-10:

Functional outcomes of sleep questionnaire short version

EQ-5D-5L:

EuroQol 5D-5 L

K-BILD:

King’s brief interstitial lung disease questionnaire

BDI-II:

Beck depression inventory-II

GAD-7:

7-item generalized anxiety disorder scale

GerdQ:

Gastroesophageal reflux disease questionnaire

FVC:

Forced vital capacity

DLCO:

Diffusing capacity of the lungs for carbon monoxide

BMI:

Body mass index

GERD:

Gastroesophageal reflux disease

SD:

Standard deviation

PREMs:

Patient-Reported Experience Measures

IQR:

Interquartile range

PEEP:

Positive end expiratory pressure

References

  1. Khor YH, Ng Y, Barnes H, Goh NSL, McDonald CF, Holland AE. Prognosis of idiopathic pulmonary fibrosis without anti-fibrotic therapy: a systematic review. Eur Respir Rev. 2020;29(157).

  2. Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, et al. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management. Am J Respir Crit Care Med. 2011;183(6):788–824.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Raghu G, Remy-Jardin M, Myers JL, Richeldi L, Ryerson CJ, Lederer DJ, et al. Diagnosis of idiopathic pulmonary fibrosis. An Official ATS/ERS/JRS/ALAT Clinical Practice Guideline. Am J Respir Crit Care Med. 2018;198(5):e44–68.

    Article  PubMed  Google Scholar 

  4. Raghu G, Remy-Jardin M, Richeldi L, Thomson CC, Inoue Y, Johkoh T, et al. Idiopathic pulmonary fibrosis (an update) and progressive pulmonary fibrosis in adults: an Official ATS/ERS/JRS/ALAT Clinical Practice Guideline. Am J Respir Crit Care Med. 2022;205(9):e18–47.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Platenburg M, van Moorsel CHM, Wiertz IA, van der Vis JJ, Vorselaars ADM, Veltkamp M et al. Improved Survival of IPF patients Treated With Antifibrotic Drugs Compared With Untreated Patients. Lung. 2023.

  6. Kolilekas L, Manali E, Vlami KA, Lyberopoulos P, Triantafillidou C, Kagouridis K, et al. Sleep oxygen desaturation predicts survival in idiopathic pulmonary fibrosis. J Clin Sleep Med. 2013;9(6):593–601.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Lee RN, Kelly E, Nolan G, Eigenheer S, Boylan D, Murphy D, et al. Disordered breathing during sleep and exercise in idiopathic pulmonary fibrosis and the role of biomarkers. QJM. 2015;108(4):315–23.

    Article  CAS  PubMed  Google Scholar 

  8. Bosi M, Milioli G, Fanfulla F, Tomassetti S, Ryu JH, Parrino L, et al. OSA and prolonged Oxygen Desaturation during Sleep are strong predictors of poor outcome in IPF. Lung. 2017;195(5):643–51.

    Article  PubMed  Google Scholar 

  9. Tudorache V, Traila D, Marc M, Oancea C, Manolescu D, Tudorache E, et al. Impact of moderate to severe obstructive sleep apnea on the cognition in idiopathic pulmonary fibrosis. PLoS ONE. 2019;14(2):e0211455.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gille T, Didier M, Boubaya M, Moya L, Sutton A, Carton Z et al. Obstructive sleep apnoea and related comorbidities in incident idiopathic pulmonary fibrosis. Eur Respir J. 2017;49(6).

  11. Mermigkis C, Bouloukaki I, Antoniou K, Papadogiannis G, Giannarakis I, Varouchakis G, et al. Obstructive sleep apnea should be treated in patients with idiopathic pulmonary fibrosis. Sleep Breath. 2015;19(1):385–91.

    Article  PubMed  Google Scholar 

  12. Corte TJ, Wort SJ, Talbot S, Macdonald PM, Hansel DM, Polkey M, et al. Elevated nocturnal desaturation index predicts mortality in interstitial lung disease. Sarcoidosis Vasc Diffuse Lung Dis. 2012;29(1):41–50.

    CAS  PubMed  Google Scholar 

  13. Troy LK, Young IH, Lau EMT, Wong KKH, Yee BJ, Torzillo PJ, et al. Nocturnal hypoxaemia is associated with adverse outcomes in interstitial lung disease. Respirology. 2019;24(10):996–1004.

    Article  PubMed  Google Scholar 

  14. Bordas-Martinez J, Salord N, Vicens-Zygmunt V, Perez S, Prado E, Calvo M et al. Characterization of Sleep-Disordered Breathing in Idiopathic Pulmonary Fibrosis. Arch Bronconeumol. 2023.

  15. Hagmeyer L, Herkenrath SD, Treml M, Pietzke-Calcagnile A, Anduleit N, Randerath W. Sleep-related breathing disorders in idiopathic pulmonary fibrosis are frequent and may be associated with pulmonary vascular involvement. Sleep Breath. 2022.

  16. Lederer DJ, Jelic S, Basner RC, Ishizaka A, Bhattacharya J. Circulating KL-6, a biomarker of lung injury, in obstructive sleep apnoea. Eur Respir J. 2009;33(4):793–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Aihara K, Oga T, Harada Y, Chihara Y, Handa T, Tanizawa K, et al. Comparison of biomarkers of subclinical lung injury in obstructive sleep apnea. Respir Med. 2011;105(6):939–45.

    Article  PubMed  Google Scholar 

  18. Kim JS, Podolanczuk AJ, Borker P, Kawut SM, Raghu G, Kaufman JD, et al. Obstructive sleep apnea and subclinical interstitial lung disease in the multi-ethnic study of atherosclerosis (MESA). Ann Am Thorac Soc. 2017;14(12):1786–95.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Bouloukaki I, Fanaridis M, Testelmans D, Pataka A, Schiza S. Overlaps between obstructive sleep apnoea and other respiratory diseases, including COPD, asthma and interstitial lung disease. Breathe (Sheff). 2022;18(3):220073.

    Article  PubMed  Google Scholar 

  20. Locke BW, Lee JJ, Sundar KM. OSA and Chronic Respiratory Disease: mechanisms and epidemiology. Int J Environ Res Public Health. 2022;19(9).

  21. Mermigkis C, Bouloukaki I, Schiza SE. Sleep as a new target for improving outcomes in idiopathic pulmonary fibrosis. Chest. 2017;152(6):1327–38.

    Article  PubMed  Google Scholar 

  22. Papadogiannis G, Bouloukaki I, Mermigkis C, Michelakis S, Ermidou C, Mauroudi E, et al. Patients with idiopathic pulmonary fibrosis with and without obstructive sleep apnea: differences in clinical characteristics, clinical outcomes, and the effect of PAP treatment. J Clin Sleep Med. 2021;17(3):533–44.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Mermigkis C, Bouloukaki I, Antoniou KM, Mermigkis D, Psathakis K, Giannarakis I, et al. CPAP therapy in patients with idiopathic pulmonary fibrosis and obstructive sleep apnea: does it offer a better quality of life and sleep? Sleep Breath. 2013;17(4):1137–43.

    Article  PubMed  Google Scholar 

  24. Mermigkis C, Mermigkis D, Varouchakis G, Schiza S. CPAP treatment in patients with idiopathic pulmonary fibrosis and obstructive sleep apnea–therapeutic difficulties and dilemmas. Sleep Breath. 2012;16(1):1–3.

    Article  PubMed  Google Scholar 

  25. Al Aghbari J, Wong K, Lau E, Taylor N, Webster S, Yee B, et al. Supplemental oxygen improves sleep disordered breathing but not quality of life in patients with interstitial lung disease. Respirol (Carlton Vic). 2016;21:147.

    Google Scholar 

  26. Vazquez JC, Perez-Padilla R. Effect of oxygen on sleep and breathing in patients with interstitial lung disease at moderate altitude. Respiration. 2001;68(6):584–9.

    Article  CAS  PubMed  Google Scholar 

  27. Adegunsoye A, Neborak JM, Zhu D, Cantrill B, Garcia N, Oldham JM, et al. CPAP Adherence, Mortality, and progression-free survival in interstitial lung Disease and OSA. Chest. 2020;158(4):1701–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Mediano O, Gonzalez Mangado N, Montserrat JM, Alonso-Alvarez ML, Almendros I, Alonso-Fernandez A, et al. International Consensus Document on Obstructive Sleep Apnea. Arch Bronconeumol. 2022;58(1):52–68.

    Article  PubMed  Google Scholar 

  29. Patil SP, Ayappa IA, Caples SM, Kimoff RJ, Patel SR, Harrod CG. Treatment of adult obstructive sleep apnea with positive Airway pressure: an American Academy of Sleep Medicine Clinical Practice Guideline. J Clin Sleep Med. 2019;15(2):335–43.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Randerath W, Verbraecken J, Andreas S, Arzt M, Bloch KE, Brack T et al. Definition, discrimination, diagnosis and treatment of central breathing disturbances during sleep. Eur Respir J. 2017;49(1).

  31. Jacobs SS, Krishnan JA, Lederer DJ, Ghazipura M, Hossain T, Tan AM, et al. Home Oxygen Therapy for adults with chronic lung disease. An official American thoracic Society Clinical Practice Guideline. Am J Respir Crit Care Med. 2020;202(10):e121–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hardinge M, Annandale J, Bourne S, Cooper B, Evans A, Freeman D, et al. British thoracic society guidelines for home oxygen use in adults. Thorax. 2015;70(Suppl 1):i1–43.

    Article  PubMed  Google Scholar 

  33. Srivali N, Thongprayoon C, Cheungpasitporn W. Assessing the impact of continuous positive airway pressure therapy on clinical outcomes in interstitial lung disease patients with coexisting obstructive sleep apnea: a systematic review. Sleep Breath. 2024.

  34. O’Driscoll BR, Howard LS, Earis J, Mak V, British Thoracic Society Emergency Oxygen, Guideline G, Group BTSEOGD. BTS guideline for oxygen use in adults in healthcare and emergency settings. Thorax. 2017;72(Suppl 1):ii1–90.

    Article  PubMed  Google Scholar 

  35. Pardo A, Cabrera S, Maldonado M, Selman M. Role of matrix metalloproteinases in the pathogenesis of idiopathic pulmonary fibrosis. Respir Res. 2016;17:23.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Minoves M, Hazane-Puch F, Moriondo G, Boutin-Paradis A, Lemarie E, Pepin JL et al. Differential Impact of intermittent vs. sustained hypoxia on HIF-1, VEGF and proliferation of HepG2 cells. Int J Mol Sci. 2023;24(8).

  37. Friscic T, Percic M, Vidovic D, Stajduhar A, Galic E. Impact of CPAP therapy on new inflammation biomarkers. J Clin Med. 2022;11:20.

    Article  Google Scholar 

  38. Carreto-Binaghi LE, Aliouat el M, Taylor ML. Surfactant proteins, SP-A and SP-D, in respiratory fungal infections: their role in the inflammatory response. Respir Res. 2016;17(1):66.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Wang K, Ju Q, Cao J, Tang W, Zhang J. Impact of serum SP-A and SP-D levels on comparison and prognosis of idiopathic pulmonary fibrosis: a systematic review and meta-analysis. Med (Baltim). 2017;96(23):e7083.

    Article  CAS  Google Scholar 

  40. Sakamoto K, Hashimoto N, Kondoh Y, Imaizumi K, Aoyama D, Kohnoh T, et al. Differential modulation of surfactant protein D under acute and persistent hypoxia in acute lung injury. Am J Physiol Lung Cell Mol Physiol. 2012;303(1):L43–53.

    Article  CAS  PubMed  Google Scholar 

  41. Liang S, Li N, Heizhati M, Yao X, Abdireim A, Wang Y, et al. What do changes in concentrations of serum surfactant proteins A and D in OSA mean? Sleep Breath. 2015;19(3):955–62.

    Article  PubMed  Google Scholar 

  42. Freeberg MAT, Perelas A, Rebman JK, Phipps RP, Thatcher TH, Sime PJ. Mechanical feed-Forward loops Contribute to Idiopathic Pulmonary Fibrosis. Am J Pathol. 2021;191(1):18–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Marchioni A, Tonelli R, Cerri S, Castaniere I, Andrisani D, Gozzi F et al. Pulmonary Stretch and Lung Mechanotransduction: implications for progression in the fibrotic lung. Int J Mol Sci. 2021;22(12).

  44. Leslie KO. Idiopathic pulmonary fibrosis may be a disease of recurrent, tractional injury to the periphery of the aging lung: a unifying hypothesis regarding etiology and pathogenesis. Arch Pathol Lab Med. 2012;136(6):591–600.

    Article  PubMed  Google Scholar 

  45. Melani AS, Croce S, Messina M, Bargagli E. Untreated obstructive sleep apnea in interstitial lung disease and impact on interstitial lung Disease outcomes. Sleep Med Clin. 2024.

  46. Pitsiou G, Bagalas V, Boutou A, Stanopoulos I, Argyropoulou-Pataka P. Should we routinely screen patients with idiopathic pulmonary fibrosis for nocturnal hypoxemia? Sleep Breath. 2013;17(2):447–8.

    Article  PubMed  Google Scholar 

  47. Lancaster LH, Mason WR, Parnell JA, Rice TW, Loyd JE, Milstone AP, et al. Obstructive sleep apnea is common in idiopathic pulmonary fibrosis. Chest. 2009;136(3):772–8.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank Fujirebio Europe N.V. for providing KL-6 reactives free of charge and David Bridgewater for the English language revision of the manuscript.

Funding

This study has been funded by: Instituto de Salud Carlos III through the grants CM20/00093 (Co-funded by European Social Fund. ESF investing in your future) and PMP22/00083 (Co-funded by European Regional Development Fund, ERDF, a way to build Europe); Spanish Society of Pneumology and Thoracic Surgery (SEPAR) grants 631/2018 and 685/2018; Emerging ILD Group of SEPAR grant 005 (Boehringer-Roche); Pneumology Foundation of Catalonia (FUCAP) grant 2019; Spanish Sleep Society (SES) grant 2019. Investigation support BRN-Fundació Ramon Pla Armengol. We thank the CERCA Programme / Generalitat de Catalunya for their institutional support.

Author information

Authors and Affiliations

Authors

Contributions

J.B.M. made substantial contributions to the conception, acquisition and interpretation of data and drafted the work; S.P., E.P., M.C., R.B. and G.B. made substantial contributions to data acquisition; N.S., V.V-Z., S.S. made substantial contributions to the design of the work and substantively revised the manuscript. J.C. made substantial contributions to the analysis; C.M. and M.M-M. made substantial contributions to the conception, the acquisition, interpretation of data and substantively revised the manuscript. All the authors have approved the submitted version.

Corresponding author

Correspondence to Carmen Monasterio.

Ethics declarations

Consent for publication

Not applicable.

Ethics approval

This study was approved by the Ethics Committee of Bellvitge University Hospital (PR413/18).

Consent to participate

All patients have signed the informed consent.

Competing interests

The authors declare no competing interests.

Conflict of interest

The authors state that they have no conflict of interests to declare.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bordas-Martinez, J., Salord, N., Vicens-Zygmunt, V. et al. Treating sleep-disordered breathing of idiopathic pulmonary fibrosis patients with CPAP and nocturnal oxygen treatment. A pilot study. Respir Res 25, 247 (2024). https://doi.org/10.1186/s12931-024-02871-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12931-024-02871-6

Keywords