Skip to main content
Erschienen in: Orphanet Journal of Rare Diseases 1/2017

Open Access 01.12.2017 | Review

Maximum inspiratory pressure as a clinically meaningful trial endpoint for neuromuscular diseases: a comprehensive review of the literature

verfasst von: Benedikt Schoser, Edward Fong, Tarekegn Geberhiwot, Derralynn Hughes, John T. Kissel, Shyam C. Madathil, David Orlikowski, Michael I. Polkey, Mark Roberts, Harm A. W. M. Tiddens, Peter Young

Erschienen in: Orphanet Journal of Rare Diseases | Ausgabe 1/2017

Abstract

Respiratory muscle strength is a proven predictor of long-term outcome of neuromuscular disease (NMD), including amyotrophic lateral sclerosis, Duchenne muscular dystrophy, and spinal muscular atrophy. Maximal inspiratory pressure (MIP), a sensitive measure of respiratory muscle strength, one of several useful tests of respiratory muscle strength, is gaining interest as a therapeutic clinical trial endpoint for NMD. In this comprehensive review we investigate the use of MIP as a measure of respiratory muscle strength in clinical trials of therapeutics targeting respiratory muscle, examine the correlation of MIP with survival, quality of life, and other measures of pulmonary function, and outline the role of MIP as a clinically significantly meaningful outcome measure. Our analysis supports the utility of MIP for the early evaluation of respiratory muscle strength, especially of the diaphragm, in patients with NMD and as a surrogate endpoint in clinical trials of therapies for NMD.
Abkürzungen
6MWT
6-minute walk test
ALS
Amyotrophic lateral sclerosis
COPD
Chronic obstructive pulmonary disease
ESS
Epworth sleepiness scale
FEV1
Forced expiratory volume in 1 second
FRC
Functional residual capacity
FVC
Forced vital capacity
MEP
Maximal expiratory pressure
MFI
Multidimensional fatigue inventory
MIP
Maximal inspiratory pressure
NMD
Neuromuscular disease
QoL
Quality of life
RCT
Randomized controlled trial
SF-36
36-Item short form
SNIP
Sniff nasal inspiratory pressure
TLC
Total lung capacity
VC
Vital capacity

Background

Weakness of the respiratory muscles is especially common among patients who have an acute or chronic neuromuscular disease (NMD), including amyotrophic lateral sclerosis (ALS), Guillain-Barré syndrome, spinal muscular atrophy, myotonic dystrophy type 1, Duchenne and other muscular dystrophies, and Pompe disease [13]. In patients with a NMD, irrespective of age, pathophysiological mechanisms that lead to the development of respiratory failure frequently include progressive weakness in the inspiratory muscles, predominantly the diaphragm, as indicated by respiratory patterns with low tidal volumes [4, 5]. However, the etiology of respiratory dysfunction can vary somewhat between different conditions [610].
NMD may impact different facets of respiratory muscle function (inspiratory, expiratory extrathoracic airways) to different extents. While expiratory muscle weakness is associated with ineffective cough, inspiratory muscle weakness causes dyspnea and/or nocturnal alveolar hypoventilation [4]. Dyspnea, which results in an increased sense of effort, is a subjective sensation of breathing discomfort, likened to being smothered or suffocated. Nocturnal hypoventilation disrupts normal sleep architecture, initially in REM sleep leading to excessive daytime fatigue and morning headaches due to hypercapnia [11, 12]. Other symptoms of nocturnal hypoventilation, include insomnia, hypersomnolence, or impaired cognition. In patients with NMD, hypoventilation during REM sleep may be an early marker of the functional impact of diaphragm weakness [12]. Lastly, expiratory muscle weakness leads to ineffective clearance of airway secretions, and depending on the severity of the muscle weakness, these patients are thus at a higher risk for aspiration (more so since such patients have concomitant swallowing difficulties), bronchitis, and pneumonia [3, 11].
The symptoms of respiratory muscle weakness can infringe significantly on daily activities (eg, walking and eating) and quality of life (QoL) of patients with NMD. Respiratory muscle weakness ultimately leads to respiratory failure and early mortality [13, 14] or the need for noninvasive or invasive mechanical ventilation to prolong survival [3, 13, 1518]. Indeed, respiratory failure secondary to muscle weakness is a common cause of premature death in NMD [19]. It is, therefore, critical that patients with NMD are regularly monitored (including for measures of respiratory muscle strength and function, cough, and swallowing [20, 21]) and subsequently managed accordingly.
Maximal inspiratory pressure (MIP) and maximal expiratory pressure (MEP) are direct measures of respiratory muscle strength and may be more sensitive in detecting early respiratory muscle dysfunction compared with spirometry, but MIP and MEP are not usually performed on all patients referred for PFTs [22]. MIP and MEP are noninvasive, straightforward tests in which individuals are asked to perform a forceful inspiration after an expiration to residual volume level (in the case of MIP) or expiration after a full inspiration to total lung capacity (TLC; in the case of MEP) with an open glottis against an occluded mouthpiece [5, 22, 23]; the tests are generally practical in individuals older than 6 or 7 years of age. They are indicated if muscle weakness could be contributing to abnormal spirometry test results, such as a low vital capacity (VC) [22]. MIP is a measure of global inspiratory muscle strength and therefore has a close relationship with diaphragmatic strength, since the diaphragm is the major inspiratory muscle; MEP is generated through the abdominal and intercostal muscles [2326].
Some debate exists about a “normal” value of MIP and MEP, and different cut-off points for percentages of predicted values have been recommended, not taking into consideration any adjustments other than gender; a recent study [27] has recommended using more ‘cautious’ reference equations. A 2009-update [22] of the statement on respiratory muscle testing by the American Thoracic Society and the European Respiratory Society [23] on the assessment of MRPs, provides more exact estimates for normal values of MIP and MEP and recommends the use of a flanged mouthpiece for the measurements. Furthermore, low MIPs can sometimes be difficult to interpret in patients with advanced illness because exertion of maximal effort is a challenge for these patients.
Together, MIP and MEP measurements can accurately assess respiratory muscle weakness, and MIP may even predict diaphragm weakness before a significant change in spirometry endpoints (eg, forced vital capacity [FVC]) [28]. However, despite the potential advantages of MIP and MEP, respiratory muscle function should be evaluated with the complete array of widely available lung volume and pressure measurements, rather than relying upon individual measurements used in isolation [29].
Given that respiratory muscle dysfunction—especially that of the diaphragm—is common to NMDs, then measurement of respiratory muscle strength through MIP may provide an additional meaningful endpoint in trials of therapeutics targeting respiratory muscle in patients with NMDs. With this in mind, we examined the use of MIP as a clinical endpoint in trials of therapeutics and investigated the relationship of MIP values with other parameters associated with respiratory muscle dysfunction, including survival and QoL metrics.

Methods

We conducted literature searches in multiple databases (EMBASE, Scopus, PubMed, ProQuest, and Google Scholar; articles published before August 2015) and included clinical studies that used MIP as either a primary or secondary clinical endpoint outcome measure and reported a relationship between MIP and survival, QoL, pulmonary, and/or nonpulmonary functional endpoints. BioMarin Pharmaceutical Inc. provided funding for this analysis and for medical writing and editorial support during manuscript development. BioMarin Pharmaceutical Inc. was not involved in the collection, analysis, or interpretation of data. All authors had full access to study data and were solely responsible for the decision to submit for publication.
Search terms included the following key words: (“maximal inspiratory pressure” or “nasal inspiratory pressure” or “negative inspiratory force” or “MIP” or “PImax”) AND (“outcome” or “endpoint” or “efficacy” or “treatment effectiveness” or “sleep-disordered breathing” or “nocturnal hypoventilation” or “mortality” or “survival” or “death”). We initially screened literature records on the basis of title and abstract and excluded records not meeting the inclusion criteria (eg, no MIP results reported, disease and/or natural history focused).
To identify open, ongoing clinical trials that are using MIP as an endpoint (primary or secondary), we searched the ClinicalTrials.gov database (www.​clinicaltrials.​gov). The search was conducted on July 12, 2015; search terms included maximal inspiratory pressure, PImax, and negative inspiratory force.

Results

MIP as an endpoint in clinical trials

We identified 8 publications in which MIP was used as a primary or secondary endpoint in a randomized controlled trial (RCT) of a pharmacologic therapy in a broad spectrum of conditions (Table 1) [26, 3036]. One of these studies was in patients with Duchenne muscular dystrophy [36]. Across the studies, the MIP endpoint was reported to be a sensitive and specific clinical measure for evaluation of the pharmacologic interventions, all of which directly targeted the respiratory musculature.
Table 1
Completed RCTs using MIP as a clinical endpoint
Reference
MIP Endpoint
Treatment Group
N
MIP Result
Golparvar M et al., 2005 [30]
Primary
Progesterone administration in adult trauma patients during partial support ventilation
40
MIP significantly increased (P < 0.05) 3 h after administration
Gontijo-Amaral C et al., 2012 [26]
Primary
Oral magnesium supplementation in adolescent cystic fibrosis patients
44
Significant increase in MIP between intervention and placebo period (P < 0.001)
Mackersie RC et al., 1991 [31]
Primary
Continuous epidural or continual IV infusions of fentanyl in patients with multiple rib fractures
32
Significant increase in MIP in epidural and fentanyl epidural groups compared with pre-analgesia (P < 0.05)
Sosis M et al., 1987 [32]
Primary
Atracurium treatment in patients requiring intubation
39
Significant decrease in MIP in patients receiving atracurium compared with placebo (P < 0.05)
Andreas S et al., 2006 [35]
Primary
Irbesartan treatment in COPD patients
60
No significant difference in MIP after 4 months of treatment (P = 0.16). Reporting a large standard deviation of MIP.
Skorodin MS et al., 1995 [33]
Primary
Magnesium sulphate administration in COPD patients
72
No significant difference in MIP after 20 and 45 min of treatment (P = NS)
Weisberg J et al., 2002 [34]
Primary
Megestrol acetate administration in COPD patients
128
No significant difference in MIP (P = NS). Reporting a large standard deviation of MIP.
Buyse GM et al., 2013 [36]
Secondary
Idebenone treatment in Duchenne muscular dystrophy patients
21
MIP improved in idebenone group but deteriorated in placebo group (P = NS)
COPD chronic obstructive pulmonary disease, IV intravenous, MIP maximum inspiratory pressure, NS not significant
A search of the clinical trials database revealed 31 open trials where MIP was cited as either a primary or secondary endpoint (Table 2). The trials reflected a range of clinical conditions; approximately 1 in 3 were in individuals with a NMD (including ALS, Duchenne muscular dystrophy, myasthenia gravis, Pompe disease, and X-linked myotubular myopathy). This suggests that MIP is gaining momentum as clinical endpoint for monitoring respiratory muscle function in these patients.
Table 2
Ongoinga clinical trials with MIP as an endpoint
NCT number
Study Title
Study Design
Primary or Secondary Endpoint
Estimated Study Completion Date
Estimated Enrollment
Neuromuscular diseases
 Amyotrophic lateral sclerosis
  NCT02478450
Study to Investigate the Safety of the Transplantation (by Injection) of Human Glial Restricted Progenitor Cells (hGRPs; Q-Cells®) Into Subjects With Amyotrophic Lateral Sclerosis (ALS)
Nonrandomized, open-label, parallel group study
Secondary
December 2017
12
Duchenne muscular dystrophy
 NCT02310906
Phase I/II Study of SRP-4053 in DMD Patients
Randomized, placebo-controlled, double-blind, parallel group study
Secondary
December 2016
48
 NCT01999075
Stacking Exercises Aid the Decline in FVC and Sick Time (STEADFAST)
Randomized, single-blind, parallel group study
Secondary
August 2016
110
 NCT02255552
Confirmatory Study of Eteplirsen in DMD Patients (PROMOVI)
Nonrandomized, open-label, parallel group study
Secondary
May 2016
160
Myasthenia gravis
 NCT01047761
Exercise for Stable Myasthenia Gravis
Nonrandomized, open-label, single group study
Secondary
December 2015
30
Parkinson’s disease
 NCT02202057
Respiratory Load Magnitude Estimation in PD
Prospective, case control study
Secondary
August 2015
80
Pompe disease
 NCT02357225
A Pilot Study of Pyridostigmine in Pompe Disease
Nonrandomized, open-label, single group study
Primary
June 2017
16
 NCT02354651
Response to Diaphragmatic Pacing in Subjects With Pompe Disease
Prospective, observational study
Secondary
February 2017
12
 NCT01924845
BMN 701 Phase 3 in rhGAA Exposed Subjects With Late Onset Pompe Disease (INSPIRE Study)
Nonrandomized, open-label, single group study
Primary
June 2020
50
Pulmonary hypertension
 NCT02288442
Whole Muscle Exercise Training (WHOLEi + 12) in Pulmonary Hypertension
Randomized, single-blind, parallel group study
Secondary
September 2016
20
Sarcopenia
 NCT02120586
Preventive Physiotherapy Intervention in Elderly People With Sarcopenia
Randomized, single-blind, parallel group study
Primary
July 2015
70
X-linked myotubular myopathy
 NCT02453152
Respiratory Muscle Function in Untreated X-Linked Myotubular Myopathy (XLMTM)
Prospective, observational study
Secondary
December 2016
12
Other disorders (non-NMDs)
 Back pain
  NCT02429752
Low Back Pain and Breathing Pattern Dysfunction (LBP & BPD)
Open-label, single group study
Secondary
June 2016
75
Breast disease
 NCT02491762
The Effect of Breast Reconstruction Surgery Using Tissue Expanders on Respiratory Functions
Nonrandomized, open-label, parallel group study
Primary
August 2017
45
 NCT02165696
Compression Bandaging and Manual Lymph Drainage in Women With Lymphedema (LYMPHATIC)
Randomized, single-blind, parallel group study
Secondary
December 2015
44
COPD
 NCT01655199
Sensitivity of the Step Test to Detect Improvement in Dyspnea Following Bronchodilation in Patients With Chronic Obstructive Pulmonary Disease (CODEx)
Double-blind, single group study
Secondary
July 2014
40
 NCT01903772
Effects of Inspiratory Muscle Training in Chronic Obstructive Pulmonary Disease (COPD) (IMTCO)
Randomized, double-blind, parallel group study
Secondary
December 2015
72
 NCT01956565
Feasibility of Inspiratory Muscle Training in People With COPD Who Decline Pulmonary Rehabilitation
Nonrandomized, open-label, single group study
Secondary
November 2015
20
 NCT02392715
Inspiratory Muscle Training Combined With General Exercise Training in COPD (IMTGET)
Randomized, double-blind, parallel group study
Secondary
October 2016
80
 NCT02007772
Effectiveness of TNI vs. BiPAP in Chronic Global Insufficiency in COPD Patients (TIBICO)
Randomized, open-label, crossover study
Secondary
August 2015
85
 NCT01582958
The Effect of OMT on Patients With COPD: Correlating Pulmonary Function Tests With Biochemical Alterations
Randomized, single-blind, parallel group study
Primary
August 2015
60
 NCT01037387
Effect of Noninvasive Ventilation on Physical Activity and Inflammation in COPD Patients
Randomized, open-label, parallel group study
Secondary
June 2016
50
COPD or asthma
 NCT02233114
Do Yogic Exercises (12 weeks) Increase Respiratory Function in Patients with Obstructive Lung Diseases?
Randomized, single-blind, parallel group study
Secondary
December 2017
40
Chronic respiratory insufficiency
 NCT01458314
Non Invasive Mechanical Ventilation in Chronic Respiratory Insufficiency Patients During Rehabilitation
Randomized, open-label, parallel group study
Secondary
June 2015
50
Chronic respiratory failure
 NCT00994552
Comparison of Pressure Support and Pressure Control Ventilation in Chronic Respiratory Failure
Randomized, single-blind crossover study
Secondary
April 2010
20
Congenital heart disease
 NCT02438293
The Impact of Rhinovirus Infections in Paediatric Cardiac Surgery’ (RISK)
Prospective, observational study
Secondary
June 2016
250
Intraventricular hemorrhage
 NCT02231411
Neonatal Resuscitation With Intact Cord (NRIC)
Randomized, single-blind, parallel group study
Secondary
August 2016
150
Lung cancer
 NCT02493114
Functional Status in Patients Undergoing Curative Treatment for Lung Cancer
Prospective, observational study
Secondary
November 2017
80
Mechanically ventilated patients
 NCT02003053
A Randomized, Controlled Trial of Inspiratory Muscle Training (IMT) in the ICU and CCU
Randomized, single-blind, parallel group study
Primary
September 2015
40
Obstructive sleep apnea
 NCT02259660
Airway Muscle Training for Obstructive Sleep Apnea (OSA)
Randomized, double-blind, parallel group study
Secondary
September 2016
60
Stroke
 NCT02400138
Home-based Respiratory Training After Stroke
Randomized, double-blind, parallel group study
Primary
November 2016
20
aClinical trials in progress as of October 31st, 2015
BiPAP biphasic positive airway pressure, CCU critical care unit, COPD chronic obstructive pulmonary disease, DMD Duchenne muscular dystrophy, FVC forced vital capacity, ICU intensive care unit, MIP maximum inspiratory pressure, OMT osteopathic manipulative treatment, PD Parkinson’s disease, TNI therapy with nasal insufflation

Relationship between MIP and survival in patients with NMD

As noted, respiratory failure is a common cause of premature death in patients with NMD [19]. Consequently, patients with progressive disease require frequent monitoring of their pulmonary function. Sensitive, noninvasive predictive measures are needed to quantify the risk of mortality due to respiratory failure in these patients; predictive measures could also quantify the potential mortality risk benefit of a therapeutic intervention.
Studies have investigated the correlation between MIP and survival in various conditions (Table 3) [3751]. In regard to NMD, the majority of data comes from patients with ALS. All ALS published studies we identified consistently found a correlation between MIP and survival [3741]. A cohort study in 95 patients with ALS found a significant association between MIP and 1-year survival (P < 0.05) [37]. The study found that, whereas a normal (>80% predicted) supine FVC predicted a > 80% chance of 1-year tracheostomy-free survival, a normal MIP or MEP predicted a > 90% chance of survival. In a second ALS study, reduced MIP predicted poor 2-years survival. Extensively controlled for nonpulmonary factors known to predict survival in ALS, Kaplan-Meier and receiver operating characteristic curve analysis showed that 2-years survival was more probable in patients with initially normal MIP values (P = 0.0001) compared with patients who had initially reduced MIP (<70 cm H2O; P < 0.05) [38]. In a third study of ALS patients (N = 21), MIP (−60 cm H2O or less) was 100% sensitive as a “threshold” for predicting 18-months survival, whereas FVC (<80% of predicted) was not as sensitive for predicting survival (<80% sensitive) [39]. In a fourth study of 53 patients with ALS, comparison of baseline data in patients who were dead or alive at 18 months showed that survivors had a higher mean MIP (38 ± 24% predicted) than nonsurvivors (20 ± 18% predicted; P < 0.01). The absence of cough spikes (defined as peak flow rate transients during voluntary cough) had no significant influence on survival [40]. Finally, clinical results from a 5-years prospective, comparative trial of patients with ALS using noninvasive ventilation found that determinants of respiratory function (including MIP [P = 0.0001]) were an independent predictor of 5-years survival, emphasizing the potential utility of MIP as a prognostic indicator in patients with ALS [41].
Table 3
Summary of studies investigating the correlation between MIP and survival
Reference
Therapy Area/Population
Study Type
N
MIP and Survival
Correlation between MIP and survival
 Schmidt EP et al., 2006 [37]
ALS
Cohort study
95
MIP was an important predictor of 1-year survival (P < 0.05) after controlling for nonpulmonary factors known to predict survival in ALS
 Baumann F et al., 2010 [38]
ALS
Cohort study
80
MIP was significantly associated with survival (P < 0.05); survival time was increased in patients with normal MIP
 Gay PC et al., 1991 [39]
ALS
Prospective study
21
MIP was 100% sensitive for predicting 18-months survival (r = 0.57; P < 0.007)
 Chaudri MB et al., 2002 [40]
MND (ALS)
Single-center cohort study
53
Survivors had a higher percentage of predicted MIP than nonsurvivors (37.83 ± 24.32% vs 20.13 ± 18.43%; P < 0.01)
 Lopes Almeida JP, et al., 2012 [41]
ALS
Prospective, comparative study
60
There was a significant correlation between MIP and 5-years survival (P = 0.02) in patients with ALS using NIV
 Benzo R et al., 2013 [42]
COPD
Analysis of clinical data from NETT
1218
Decrease in MIP > 11 cm H2O was a predictor of 1-year mortality (OR, 2.19; P = 0.0217)
 Gray-Donald K et al., 1996 [43]
COPD
3 to 5-years follow-up of a double-blind RCT
348
Low MIP was a significant independent predictor of respiratory (HR, 0.64; 95% CI, 0.44–0.95) and all-cause mortality (HR, 0.67; 95% CI, 0.47–0.95)
 Schols AMWJ et al., 1998 [44]
COPD
RCT
203
Improvement in MIP during rehabilitation decreased the risk of death
 Hodgev VA et al., 2006 [45]
COPD
Prospective cohort study
63
A Cox proportional hazards analysis showed that MIP was a significant predictor of mortality (r = 0.91; 95% CI, 0.85–0.97; P = 0.005)
 Meyer FJ et al., 2001 [47]
Heart failure
Prospective study
244
In a univariate Cox regression analysis, MIP was found to be a significant prognostic indicator of survival (P = 0.001)
 Frankenstein L et al., 2009 [48]
Heart failure
Prospective, observational study
686
MIP was identified as a significant predictor of survival by univariate analysis; survivors had a significantly higher MIP and percentage of predicted MIP than nonsurvivors
 Ionescu AA et al., 1998 [49]
Cystic fibrosis
Single-center study
49
Mean % predicted MIP (SD) for survivors was 85.5% (28.4) compared with 64.1% (23.9) for nonsurvivors
 Marroni CA et al., 2014 [50]
Liver cirrhosis
Prospective cohort study
86
Sixty-two percent of patients with MIP < −70 cm H2O survived compared with 93% of patients with MIP > −70 cm H2O (P = 0.0001)
 Budweiser S et al., 2007 [46]
Chronic hypercapnic respiratory failure
Cohort study
464
MIP was a significant predictor of long-term survival; according to stepwise multivariate Cox regression analysis, P0.1/MIP was identified as an independent predictor of survival (P < 0.05)
 van der Palen J et al., 2004 [51]
Elderly with a mean age of 72.5 years
Cohort study
3839
Subjects in the lowest quintile of MIP had a 1.5-fold increased risk of cardiovascular-related death (HR, 1.54; 95% CI, 1.09–2.15) after adjustment for nonpulmonary function covariates
No correlation between MIP and survival
 Nizet TAC et al., 2005 [58]
Chronic hypercapnic COPD
Prospective, single-center study
47
No significant association
 Frankenstein L et al., 2008 [54]
CHF
Prospective, single-arm study
158
No significant association
 Habedank D et al., 2013 [55]
CHF
Single-center study
249
No significant association
 Hui D et al., 2014 [56]
Advanced cancer
Prospective, single-center study
222
No significant association
 White AC et al., 2005 [59]
Hematopoietic stem cell transplantation
Prospective observational study
56
No significant association
 Jackson M et al., 1994 [57]
Patients with a thoracoplasty for tuberculosis
Single-center study
32
No significant association
ALS amyotrophic lateral sclerosis, CHF congestive heart failure, CI confidence interval, COPD chronic obstructive pulmonary disease, HR hazard ratio, MIP maximum inspiratory pressure, MND motor neurone disease, NETT National Emphysema Treatment Trial, NIV noninvasive ventilation, OR odds ratio, P 0.1 mouth occlusion pressure, RCT randomized controlled trial, SD standard deviation
In a further study recently published by one of the current authors [52], multiple outcome measures were obtained in 78 patients with ALS who were then followed until death. Low values for MIP were highly specific predictors of time to death or initiation of NIV; conversely, while VC was also a specific predictor, the cut points suggested by ROC analysis were >80% of normal at all time points except for 3 months mortality prediction (when it was 78%), suggesting that a normal VC was of limited practical value. However, a small (N = 18) study of MIP and survival in patients with Duchenne muscular dystrophy [53] did not find a predictive association. It should be noted that participants had extremely low values of MIP and VC at the start of study and that the analysis did not include the use of noninvasive ventilation.
Some studies in non-neuromuscular diseases—chronic obstructive pulmonary disease (COPD) [4245], cystic fibrosis [49], liver cirrhosis [50], hypercapnic respiratory failure [46], and congestive heart failure [47, 48, 51]—suggest that MIP may correlate with survival, while others report no correlation [5459] (Table 3), indicating that further investigation is needed. We also caution that in non-neuromuscular disease states, a reduced MIP may simply reflect generalized cachexia, which is a recognized marker of a poor prognosis and hyperinflation [60] in pulmonary disease.

Relationship between MIP and QoL in patients with NMD

As NMD pathology progresses and patients develop respiratory dysfunction, QoL (with respect to a patient’s physical, emotional, social functioning, mental health, bodily pain, endurance, and general health perceptions) and sleep can be severely impacted [18, 61, 62]. The relationship between changes in MIP and QoL in NMD was addressed in 2 studies from our literature search: one in ALS [18] and one in patients with post-poliomyelitis syndrome [63].
Bourke et al. evaluated the impact of noninvasive intervention on QoL in 22 patients with ALS using the 36-Item Short Form (SF-36) and the National Center for Health Statistics General Well-Being Schedule and concluded that respiratory muscle weakness had an impact on QoL [18]. Overall, the researchers found that patients with ALS with significantly lower QoL displayed lower MIP values. Lower MIP values corresponded with lower SF-36 scores in all domains except the pain and physical components [18]. Instruments specifically assessing respiratory and sleep-related problems (eg, the Epworth Sleepiness Scale [ESS]) were most sensitive to changes in MIP [18]. In this regard, ESS scores were highest (indicating sleep disruption) in patients with MIP values below 50% and were lowest in patients with MIP values above 50% (10.3 and 4.8%, respectively; P = 0.01) [18].
Similar to the findings of Bourke et al., a cross-sectional study of 52 patients with post-poliomyelitis syndrome observed a significant correlation between MIP and both the fatigue severity scale and the Multidimensional Fatigue Inventory (MFI) (r = −0.31 and r = −0.41, respectively; P < 0.05). The researchers also found that a 10-unit decline for MIP (% predicted) corresponded to patients scoring a 0.3-unit increase on the General Fatigue dimension of the MFI scale [63]. This dimension of the MFI scale ranges from 4 to 20, with higher scores indicating more severe fatigue.

Relationship between MIP and spirometry in patients with NMD

In patients with NMD whose lungs are restricted from fully expanding, spirometry is widely used to assess respiratory muscle function. Patients performing spirometry are asked to take a maximal inspiration and perform a FVC maneuver. A drop of more than 20% of FVC going from the upright to the supine position is a useful diagnostic of diaphragmatic weakness [64]. FVC, however, has a curvilinear relationship with respiratory muscle strength, and substantial weakness may be present while FVC is still within the normal range [65].
Lung volume measurements are sometimes performed in patients with NMD, including TLC and functional residual capacity (FRC; defined as the amount of air in the lungs following normal expiration). In these patients, when inspiratory muscles are weak, then a maximum effort may be insufficient to fully expand the lungs, and the TLC and FRC will be reduced [22, 24]. Similarly, if abdominal muscle strength is impaired, residual volume may also be elevated [22].
When carbon monoxide gas transfer is measured, the classic picture of respiratory muscle weakness is a low diffusion capacity (DLCO) and an elevated transfer coefficient (KCO). However, a study in patients with NMDs showed that the rise in KCO was often less than expected; in patients with combined inspiratory and expiratory muscle weakness, a reduced value was observed [66]. The results demonstrated the limitations of using KCO in the diagnosis of respiratory muscle weakness. Gas exchange anomalies can also be multifactorial in their origin, for example, mechanical problems and airway obstructions can affect results [67].
While the above discussed spirometry tests have utility, they can be considered insensitive measures of respiratory muscle function since a significant reduction in lung volume may not be observed until severe impairment of respiratory muscles has occurred [67]. Also, other factors such as airway obstruction due to asthma or lower airway obstruction may affect the reliability of some spirometry results [5, 6870]. Consistent with these limitations, when measured within 30 days of the need for tracheostomy in a clinical trial of therapeutics for ALS [71], VC was ≥ 60% predicted in 14% of ALS patients (n = 50).
MIP has physiological relationships with spirometry endpoints [65]. Specifically, in patients with a NMD, several studies have shown a correlation between MIP and FVC. In a study of patients with Duchenne muscular dystrophy, a significant 1-year decrease in MIP was associated with decreases in FVC, FEV1 (defined as forced expiratory volume in 1 sec), and peak expiratory flow rate (P < 0.05 for all measures) [72]. Similarly, Schmidt et al. found a significant association between MIP and upright FVC in a 1-year cohort study of 95 patients with ALS [37].
Also, the LOTS study investigated the effect of alglucosidase alfa treatment in patients with late-onset Pompe disease [73]. Patients who entered an open-label extension phase of this trial showed additional improvement in MIP but a slight decline in FVC from week 78 through week 104; a statistical correlation however was not reported [74]. Finally, a RCT conducted by Cheah et al. to assess the effects of a 12-weeks inspiratory muscle training program in patients with ALS found that improvements in MIP reflected improvements in FVC and TLC [75]. In 3 RCTs where MIP improvements were observed following pharmacologic treatment in patients with non-neuromuscular diseases, these values also correlated with improvements in other pulmonary measures, including FVC (as well as airway resistance and MEP) [26, 30, 31].
Additional non-RCT studies supporting a correlation between MIP and spirometry in patients with NMDs have also been performed in patients with Pompe disease, Guillain-Barré syndrome, and myasthenia gravis [70, 74, 76, 77]. In a prospective cohort study in patients with Pompe disease, MIP and MEP were both strongly correlated with VC (r = 0.75 and r = 0.79, respectively) [76]. Follow-up data (median 1.6 years) showed that VC (upright or supine) deteriorated by 0.9–1.2% points per year, respectively, with deteriorations in MIP and MEP of 3.2% (P = 0.018) and 3.8% (P < 0.01) per year, respectively [76]. In a study of patients with respiratory muscle weakness and one study of multiple NMDs, MIP was found to significantly correlate with FRC (r = 0.62; P < 0.001) and VC (r = 0.88; P < 0.001) [77]. A direct investigation of the sensitivity of MIP versus VC in patients with either Guillain-Barré syndrome (n = 40) or myasthenia gravis (n = 44) found a linear relationship between the 2 measurements [70].
Taken together, these studies demonstrate that in patients with NMD there is a correlation between MIP and FVC, FEV1, body plethysmography, and diffusion techniques. In addition, MIP could have utility as a clinical endpoint in therapeutic trials for the treatment of neuromuscular diseases. A recent study in ALS patients with progressive respiratory dysfunction provided indirect evidence that reductions in MIP occur prior to reductions in FVC [28]. In patients with ALS, clinicians detected the progression of respiratory dysfunction 6.5 months earlier when monitoring MIP compared with FVC alone. This indicates that MIP may be a more sensitive measure of respiratory disease progression, supporting its potential utility as a clinical trial endpoint.

Relationship between MIP and nonpulmonary measures in patients with NMD

Walking tests are an integrated assessment of cardiac, pulmonary, circulatory, and muscular capacity, providing a measure of the functional exercise level required to undertake daily physical activities. Neuromuscular and pulmonary studies investigating the impact of MIP on ambulatory measures showed that, in some cases, improvements in MIP coincided with improvements in walking tests [25, 34, 54, 74, 75, 7885]. Two studies were identified comparing MIP with walking tests: one in Pompe disease and one in ALS. The study of patients with Pompe disease [74] receiving enzyme replacement therapy found that changes in the 6-min walk test (6MWT) were directionally consistent with changes in MIP. However, no such association was found in the study of patients with ALS [75].

Discussion

Since the development of respiratory failure is a significant predictor of early death, many clinical trials currently employ established spirometry endpoints, including FVC, to evaluate an intervention in patients with NMD. However, as these endpoints are measures of overall pulmonary function, they may also be affected by factors that are independent of respiratory muscle dysfunction. Given that respiratory muscle dysfunction is common in neuromuscular diseases, directly evaluating diaphragm muscle strength by measuring MIP could complement spirometric endpoints in studies of patients with these diseases. In this regard, our analysis identified the use of MIP as an endpoint in several RCTs of pharmacologic therapies across a spectrum of diseases [26, 3036]. These trials found MIP to be a clinically relevant outcome measure in chronic diseases when respiratory failure is secondary to respiratory muscle weakness.
However, diminished MIP does not always reliably confirm inspiratory muscle weakness. This is due to MIP measurement errors, including submaximal effort, poor transmission of intrathoracic pressure to the extrathoracic airways [22] as well as NMD patient-device interface issues or additional chest wall alterations [75]. For example, interface issues occur in patients with NMD with bulbar and/or facial weakness who have difficulty making a good lip seal. However, with proper training, MIP can be a reliable, accurate, and an early indicator of respiratory muscle weakness, which is more independent of existing lung abnormalities than FVC and VC [22, 8688]. In this respect, a number of ongoing and planned clinical trials are evaluating MIP as a primary or secondary endpoint in studies of patients with NMDs (Table 2).
In order to further validate MIP as a clinical endpoint in studies of patients with NMD, it is important to establish whether MIP is associated with clinically meaningful outcomes, such as time to ventilator support or even overall survival. In this regard, our analysis found that MIP was correlated with survival in patients with some NMDs (particularly ALS) and that MIP made an important contribution to predictive multivariate modeling analyses [3741]. Studies in patients with ALS consistently found that higher MIP values were associated with increased survival. One study found MIP to be 100% sensitive as a threshold for predicting survival in patients with ALS at 18-months follow-up [39]. Additionally, clinical studies that extended up to 5 years found a positive association between MIP and survival in a number of therapy areas [41, 43].
Our analysis also suggests that MIP may be reflective of challenges faced by patients with NMD in their daily life. Two studies conducted in patients with NMD found that MIP correlated with improvements in QoL scores, including domains relating to sleep and fatigue [18, 63]. Another study in patients with Pompe disease indicated that MIP was significantly correlated with the 6MWT [74, 85]. These findings suggest that MIP may have useful long-term clinical relevance in patients with NMDs. Furthermore, in the majority of NMD studies examined, there was a strong correlation between MIP with other pulmonary measures, including with FVC, FEV1, VC, and TLC [37, 72, 7476]. In addition, there is some evidence to indicate that, when respiratory muscle function starts to deteriorate, MIP measurements may decrease earlier than other pulmonary measures, suggesting that MIP may be a more sensitive method for monitoring patients [28]. In ALS patients, for example, reduced inspiratory muscle strength (MIP) was noted after cessation of inspiratory muscle training despite other pulmonary measures remaining unchanged [75].
In addition to MIP, the SNIP test can be used to measure respiratory muscle strength in the clinic. Like MIP, SNIP also reflects esophageal pressure in most patients, but a minority of patients will obtain substantially larger values on one test than the other [89]. SNIP is measured by plugging one nostril while the other is free; this test is useful when there is facial weakness or dental malocclusion, which may make the MIP test difficult [5]. It is a reproducible and accurate measure of inspiratory muscle strength [90] and can be reliably performed and used even in infants and children [91]. SNIP has been used to monitor respiratory muscle strength in patients with NMD [92, 93]. However, it has limitations such as underestimating esophageal pressure swing in patients with severe nasal obstruction or airway diseases such as COPD, which impair pressure transmission [9496]. Additionally, SNIP measures a fast contraction of inspiratory muscles, whereas MIP measures a sustained isometric contraction [95]. As isometric muscle force of limb muscles is a standard measure of muscle function, it has been argued that in some NMDs, such as ALS, MIP might provide a more meaningful measurement of respiratory muscle function than SNIP [95].
Our analysis of MIP as a clinical outcome measure in patients with NMDs is limited by the small number of published studies and small sample sizes. However, the fact that there are currently 12 registered trials in patients with NMD where MIP is being used as a clinical endpoint suggests that specific measurements of ventilatory function are important outcomes in these patient cohorts. These trials will provide further evidence of the reliability and utility of MIP. Additionally, MIP may prove to be more sensitive than FVC for assessing ventilatory dysfunction since spirometric pulmonary function tests are influenced by other factors (including scoliosis and other lung diseases), which may affect the reliability of results [5, 6870].

Conclusions

In summary, our analysis supports the use of MIP as a diagnostic of respiratory muscle dysfunction in patients with chronic NMDs and its utility as an endpoint in future clinical trials which monitor the efficacy of therapeutics in neuromuscular diseases. Through continued investigation of MIP in NMD, clinicians and researchers will gain a comprehensive understanding of the role of this direct measure of respiratory muscle strength in clinical practice.

Acknowledgments

The authors wish to acknowledge Roger Hill PhD and Ryan Woodrow for medical writing and Dena McWain for editorial assistance in the preparation of the manuscript (Ashfield Healthcare Communications, Middletown, CT, USA).

Funding

BioMarin Pharmaceutical Inc. provided funding for this analysis and for medical writing and editorial support during manuscript development.

Availability of data and materials

Not applicable.

Authors’ contributions

All authors (BS EF TH DH JK SM DO MP MR HT PY) contributed to writing, reviewing, and analyzing the MIP review manuscript. All authors read and approved the final manuscript.

Competing interests

BS received speaker honoraria from and is member of advisory boards for Audentes Inc, BioMarin Pharmaceutical Inc., and Genzyme, a Sanofi company.
MIP’s contribution to this project was supported by the NIHR Respiratory Biomedical Research Unit at the Royal Brompton and Harefield NHS Foundation Trust and Imperial College London who partially fund his salary. The views expressed in this publication are those of the authors and not necessarily those of the NHS, The National Institute for Health Research, or the Department of Health. He discloses receiving personal and institutional support for research and consultancy from BioMarin Pharmaceutical Inc. His institution has received research support from Genzyme.
PY received speaker honoraria from and is member of advisory boards for BioMarin Pharmaceutical Inc. and Genzyme, a Sanofi company.
EF received speaker honoraria from and/or is a member of advisory boards for BioMarin Pharmaceutical Inc. and Gilead Sciences Inc.
The other authors declare that they have no competing interests.
Not applicable.
Not applicable.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.
Literatur
1.
Zurück zum Zitat Perrin C, Unterborn JN, Ambrosio CD, Hill NS. Pulmonary complications of chronic neuromuscular diseases and their management. Muscle Nerve. 2004;29:5–27.CrossRefPubMed Perrin C, Unterborn JN, Ambrosio CD, Hill NS. Pulmonary complications of chronic neuromuscular diseases and their management. Muscle Nerve. 2004;29:5–27.CrossRefPubMed
2.
Zurück zum Zitat Epstein SK. An overview of respiratory muscle function. Clin Chest Med. 1994;15:619–39.PubMed Epstein SK. An overview of respiratory muscle function. Clin Chest Med. 1994;15:619–39.PubMed
3.
Zurück zum Zitat Sansone VA, Gagnon C. 207th ENMC Workshop on chronic respiratory insufficiency in myotonic dystrophies: management and implications for research, 27–29 June 2014, Naarden, The Netherlands. Neuromuscul Disord. 2015;25:432–42.CrossRefPubMed Sansone VA, Gagnon C. 207th ENMC Workshop on chronic respiratory insufficiency in myotonic dystrophies: management and implications for research, 27–29 June 2014, Naarden, The Netherlands. Neuromuscul Disord. 2015;25:432–42.CrossRefPubMed
4.
Zurück zum Zitat Ambrosino N, Gherardi M, Carpenè N. End stage chronic obstructive pulmonary disease. Pneumonol Alergol Pol. 2009;77:173–9.PubMed Ambrosino N, Gherardi M, Carpenè N. End stage chronic obstructive pulmonary disease. Pneumonol Alergol Pol. 2009;77:173–9.PubMed
5.
Zurück zum Zitat Farrero E, Antón A, Egea CJ, et al. Guidelines for the management of respiratory complications in patients with neuromuscular disease. Arch Bronconeumol. 2013;49:306–13.CrossRefPubMed Farrero E, Antón A, Egea CJ, et al. Guidelines for the management of respiratory complications in patients with neuromuscular disease. Arch Bronconeumol. 2013;49:306–13.CrossRefPubMed
6.
Zurück zum Zitat Gajdos P, Chevret S, Toyka KV. Intravenous immunoglobulin for myasthenia gravis. Cochrane Database Syst Rev. 2012;12:CD002277.PubMed Gajdos P, Chevret S, Toyka KV. Intravenous immunoglobulin for myasthenia gravis. Cochrane Database Syst Rev. 2012;12:CD002277.PubMed
7.
Zurück zum Zitat de Carvalho M, Matias T, Coelho F, Evangelista T, Pinto A, Luis ML. Motor neuron disease presenting with respiratory failure. J Neurol Sci. 1996;139(Suppl):117–22.CrossRefPubMed de Carvalho M, Matias T, Coelho F, Evangelista T, Pinto A, Luis ML. Motor neuron disease presenting with respiratory failure. J Neurol Sci. 1996;139(Suppl):117–22.CrossRefPubMed
8.
Zurück zum Zitat Benditt JO, Boitano LJ. Pulmonary issues in patients with chronic neuromuscular disease. Am J Respir Crit Care Med. 2013;187:1046–55.CrossRefPubMed Benditt JO, Boitano LJ. Pulmonary issues in patients with chronic neuromuscular disease. Am J Respir Crit Care Med. 2013;187:1046–55.CrossRefPubMed
9.
Zurück zum Zitat Mehanna R, Jankovic J. Respiratory problems in neurologic movement disorders. Parkinsonism Relat Disord. 2010;16:628–38.CrossRefPubMed Mehanna R, Jankovic J. Respiratory problems in neurologic movement disorders. Parkinsonism Relat Disord. 2010;16:628–38.CrossRefPubMed
10.
Zurück zum Zitat Smeltzer SC, Utell MJ, Rudick RA, Herndon RM. Pulmonary function and dysfunction in multiple sclerosis. Arch Neurol. 1988;45:1245–9.CrossRefPubMed Smeltzer SC, Utell MJ, Rudick RA, Herndon RM. Pulmonary function and dysfunction in multiple sclerosis. Arch Neurol. 1988;45:1245–9.CrossRefPubMed
11.
Zurück zum Zitat Ambrosino N, Carpene N, Gherardi M. Chronic respiratory care for neuromuscular diseases in adults. Eur Respir J. 2009;34:444–51.CrossRefPubMed Ambrosino N, Carpene N, Gherardi M. Chronic respiratory care for neuromuscular diseases in adults. Eur Respir J. 2009;34:444–51.CrossRefPubMed
12.
Zurück zum Zitat Boentert M, Karabul N, Wenninger S, et al. Sleep-related symptoms and sleep-disordered breathing in adult Pompe disease. Eur J Neurol. 2015;22:369–76.CrossRefPubMed Boentert M, Karabul N, Wenninger S, et al. Sleep-related symptoms and sleep-disordered breathing in adult Pompe disease. Eur J Neurol. 2015;22:369–76.CrossRefPubMed
13.
Zurück zum Zitat Winkel LP, Hagemans ML, van Doorn PA, et al. The natural course of non-classic Pompe’s disease; a review of 225 published cases. J Neurol. 2005;252:875–84.CrossRefPubMed Winkel LP, Hagemans ML, van Doorn PA, et al. The natural course of non-classic Pompe’s disease; a review of 225 published cases. J Neurol. 2005;252:875–84.CrossRefPubMed
14.
Zurück zum Zitat Gϋngör D, de Vries JM, Hop WC, et al. Survival and associated factors in 268 adults with Pompe disease prior to treatment with enzyme replacement therapy. Orphanet J Rare Dis. 2011;6:34.CrossRef Gϋngör D, de Vries JM, Hop WC, et al. Survival and associated factors in 268 adults with Pompe disease prior to treatment with enzyme replacement therapy. Orphanet J Rare Dis. 2011;6:34.CrossRef
15.
Zurück zum Zitat Mehta S. Neuromuscular disease causing acute respiratory failure. Respir Care. 2006;51:1016–21. discussion 21–3.PubMed Mehta S. Neuromuscular disease causing acute respiratory failure. Respir Care. 2006;51:1016–21. discussion 21–3.PubMed
16.
Zurück zum Zitat Sharshar T, Chevret S, Bourdain F, Raphael JC. Early predictors of mechanical ventilation in Guillain-Barre syndrome. Crit Care Med. 2003;31:278–83.CrossRefPubMed Sharshar T, Chevret S, Bourdain F, Raphael JC. Early predictors of mechanical ventilation in Guillain-Barre syndrome. Crit Care Med. 2003;31:278–83.CrossRefPubMed
17.
Zurück zum Zitat Durand MC, Porcher R, Orlikowski D, et al. Clinical and electrophysiological predictors of respiratory failure in Guillain-Barre syndrome: a prospective study. Lancet Neurol. 2006;5:1021–8.CrossRefPubMed Durand MC, Porcher R, Orlikowski D, et al. Clinical and electrophysiological predictors of respiratory failure in Guillain-Barre syndrome: a prospective study. Lancet Neurol. 2006;5:1021–8.CrossRefPubMed
18.
Zurück zum Zitat Bourke SC, Tomlinson M, Williams TL, Bullock RE, Shaw PJ, Gibson GJ. Effects of non-invasive ventilation on survival and quality of life in patients with amyotrophic lateral sclerosis: a randomised controlled trial. Lancet Neurol. 2006;5:140–7.CrossRefPubMed Bourke SC, Tomlinson M, Williams TL, Bullock RE, Shaw PJ, Gibson GJ. Effects of non-invasive ventilation on survival and quality of life in patients with amyotrophic lateral sclerosis: a randomised controlled trial. Lancet Neurol. 2006;5:140–7.CrossRefPubMed
19.
Zurück zum Zitat Miller RG, Jackson CE, Kasarskis EJ, et al. Practice parameter update: the care of the patient with amyotrophic lateral sclerosis: multidisciplinary care, symptom management, and cognitive/behavioral impairment (an evidence-based review): report of the quality standards subcommittee of the American academy of neurology. Neurology. 2009;73:1227–33.CrossRefPubMedPubMedCentral Miller RG, Jackson CE, Kasarskis EJ, et al. Practice parameter update: the care of the patient with amyotrophic lateral sclerosis: multidisciplinary care, symptom management, and cognitive/behavioral impairment (an evidence-based review): report of the quality standards subcommittee of the American academy of neurology. Neurology. 2009;73:1227–33.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Bach JR. Amyotrophic lateral sclerosis: prolongation of life by noninvasive respiratory AIDS. Chest. 2002;122:92–8.CrossRefPubMed Bach JR. Amyotrophic lateral sclerosis: prolongation of life by noninvasive respiratory AIDS. Chest. 2002;122:92–8.CrossRefPubMed
21.
Zurück zum Zitat Benditt JO. The neuromuscular respiratory system: physiology, pathophysiology, and a respiratory care approach to patients. Respir Care. 2006;51:829–37. discussion 37–9.PubMed Benditt JO. The neuromuscular respiratory system: physiology, pathophysiology, and a respiratory care approach to patients. Respir Care. 2006;51:829–37. discussion 37–9.PubMed
22.
Zurück zum Zitat Evans JA, Whitelaw WA. The assessment of maximal respiratory mouth pressures in adults. Respir Care. 2009;54:1348–59.PubMed Evans JA, Whitelaw WA. The assessment of maximal respiratory mouth pressures in adults. Respir Care. 2009;54:1348–59.PubMed
23.
Zurück zum Zitat American Thoracic Society/European Respiratory Society (ATS/ERS). ATS/ERS Statement on respiratory muscle testing. Am J Respir Crit Care Med. 2002;166:518–624.CrossRef American Thoracic Society/European Respiratory Society (ATS/ERS). ATS/ERS Statement on respiratory muscle testing. Am J Respir Crit Care Med. 2002;166:518–624.CrossRef
25.
Zurück zum Zitat Gomieiro LT, Nascimento A, Tanno LK, Agondi R, Kalil J, Giavina-Bianchi P. Respiratory exercise program for elderly individuals with asthma. Clinics (Sao Paulo). 2011;66:1163–9.CrossRef Gomieiro LT, Nascimento A, Tanno LK, Agondi R, Kalil J, Giavina-Bianchi P. Respiratory exercise program for elderly individuals with asthma. Clinics (Sao Paulo). 2011;66:1163–9.CrossRef
26.
Zurück zum Zitat Gontijo-Amaral C, Guimaraes EV, Camargos P. Oral magnesium supplementation in children with cystic fibrosis improves clinical and functional variables: a double-blind, randomized, placebo-controlled crossover trial. Am J Clin Nutr. 2012;96:50–6.CrossRefPubMed Gontijo-Amaral C, Guimaraes EV, Camargos P. Oral magnesium supplementation in children with cystic fibrosis improves clinical and functional variables: a double-blind, randomized, placebo-controlled crossover trial. Am J Clin Nutr. 2012;96:50–6.CrossRefPubMed
27.
Zurück zum Zitat Rodrigues A, Da Silva ML, Berton DC, et al. Maximal Inspiratory Pressure: Does the Choice of Reference Values Actually Matter? Chest 2016. doi:10.1016/j.chest.2016.11.045. Rodrigues A, Da Silva ML, Berton DC, et al. Maximal Inspiratory Pressure: Does the Choice of Reference Values Actually Matter? Chest 2016. doi:10.1016/j.chest.2016.11.045.
28.
Zurück zum Zitat Mendoza M, Gelinas DF, Moore DH, Miller RG. A comparison of maximal inspiratory pressure and forced vital capacity as potential criteria for initiating non-invasive ventilation in amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2007;8:106–11.CrossRefPubMed Mendoza M, Gelinas DF, Moore DH, Miller RG. A comparison of maximal inspiratory pressure and forced vital capacity as potential criteria for initiating non-invasive ventilation in amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2007;8:106–11.CrossRefPubMed
30.
Zurück zum Zitat Golparvar M, Ahmadi F, Saghaei M. Effects of progesterone on the ventilatory performance in adult trauma patients during partial support mechanical ventilation. Arch Iran Med. 2005;8:27–31. Golparvar M, Ahmadi F, Saghaei M. Effects of progesterone on the ventilatory performance in adult trauma patients during partial support mechanical ventilation. Arch Iran Med. 2005;8:27–31.
31.
Zurück zum Zitat Mackersie RC, Karagianes TG, Hoyt DB, Davis JW. Prospective evaluation of epidural and intravenous administration of fentanyl for pain control and restoration of ventilatory function following multiple rib fractures. J Trauma. 1991;31:443–9.CrossRefPubMed Mackersie RC, Karagianes TG, Hoyt DB, Davis JW. Prospective evaluation of epidural and intravenous administration of fentanyl for pain control and restoration of ventilatory function following multiple rib fractures. J Trauma. 1991;31:443–9.CrossRefPubMed
32.
Zurück zum Zitat Sosis M, Larijani GE, Marr AT. Priming with atracurium. Anesth Analg. 1987;66:329–32.PubMed Sosis M, Larijani GE, Marr AT. Priming with atracurium. Anesth Analg. 1987;66:329–32.PubMed
33.
Zurück zum Zitat Skorodin MS, Tenholder MF, Yetter B, et al. Magnesium sulfate in exacerbations of chronic obstructive pulmonary disease. Arch Intern Med. 1995;155:496–500.CrossRefPubMed Skorodin MS, Tenholder MF, Yetter B, et al. Magnesium sulfate in exacerbations of chronic obstructive pulmonary disease. Arch Intern Med. 1995;155:496–500.CrossRefPubMed
34.
Zurück zum Zitat Weisberg J, Wanger J, Olson J, et al. Megestrol acetate stimulates weight gain and ventilation in underweight COPD patients. Chest. 2002;121:1070–8.CrossRefPubMed Weisberg J, Wanger J, Olson J, et al. Megestrol acetate stimulates weight gain and ventilation in underweight COPD patients. Chest. 2002;121:1070–8.CrossRefPubMed
35.
Zurück zum Zitat Andreas S, Herrmann-Lingen C, Raupach T, et al. Angiotensin II blockers in obstructive pulmonary disease: a randomised controlled trial. Eur Respir J. 2006;27:972–9.PubMed Andreas S, Herrmann-Lingen C, Raupach T, et al. Angiotensin II blockers in obstructive pulmonary disease: a randomised controlled trial. Eur Respir J. 2006;27:972–9.PubMed
36.
Zurück zum Zitat Buyse GM, Goemans N, van den Hauwe M, Meier T. Effects of glucocorticoids and idebenone on respiratory function in patients with duchenne muscular dystrophy. Pediatr Pulmonol. 2013;48:912–20.CrossRefPubMed Buyse GM, Goemans N, van den Hauwe M, Meier T. Effects of glucocorticoids and idebenone on respiratory function in patients with duchenne muscular dystrophy. Pediatr Pulmonol. 2013;48:912–20.CrossRefPubMed
37.
Zurück zum Zitat Schmidt EP, Drachman DB, Wiener CM, Clawson L, Kimball R, Lechtzin N. Pulmonary predictors of survival in amyotrophic lateral sclerosis: use in clinical trial design. Muscle Nerve. 2006;33:127–32.CrossRefPubMed Schmidt EP, Drachman DB, Wiener CM, Clawson L, Kimball R, Lechtzin N. Pulmonary predictors of survival in amyotrophic lateral sclerosis: use in clinical trial design. Muscle Nerve. 2006;33:127–32.CrossRefPubMed
38.
Zurück zum Zitat Baumann F, Henderson RD, Morrison SC, et al. Use of respiratory function tests to predict survival in amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2010;11:194–202.CrossRefPubMed Baumann F, Henderson RD, Morrison SC, et al. Use of respiratory function tests to predict survival in amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2010;11:194–202.CrossRefPubMed
39.
Zurück zum Zitat Gay PC, Westbrook PR, Daube JR, Litchy WJ, Windebank AJ, Iverson R. Effects of alterations in pulmonary function and sleep variables on survival in patients with amyotrophic lateral sclerosis. Mayo Clin Proc. 1991;66:686–94.CrossRefPubMed Gay PC, Westbrook PR, Daube JR, Litchy WJ, Windebank AJ, Iverson R. Effects of alterations in pulmonary function and sleep variables on survival in patients with amyotrophic lateral sclerosis. Mayo Clin Proc. 1991;66:686–94.CrossRefPubMed
40.
Zurück zum Zitat Chaudri MB, Liu C, Hubbard R, Jefferson D, Kinnear WJ. Relationship between supramaximal flow during cough and mortality in motor neurone disease. Eur Respir J. 2002;19:434–8.CrossRefPubMed Chaudri MB, Liu C, Hubbard R, Jefferson D, Kinnear WJ. Relationship between supramaximal flow during cough and mortality in motor neurone disease. Eur Respir J. 2002;19:434–8.CrossRefPubMed
41.
Zurück zum Zitat Lopes Almeida JP, Braga AC, Pinto A, et al. Respiratory predictors of ALS survival in home ventilated-compliant patients. Amyotroph Lateral Scler. 2012;13:186–7. Lopes Almeida JP, Braga AC, Pinto A, et al. Respiratory predictors of ALS survival in home ventilated-compliant patients. Amyotroph Lateral Scler. 2012;13:186–7.
42.
Zurück zum Zitat Benzo R, Siemion W, Novotny P, et al. Factors to inform clinicians about the end of life in severe chronic obstructive pulmonary disease. J Pain Symptom Manage. 2013;46:491–9.CrossRefPubMedPubMedCentral Benzo R, Siemion W, Novotny P, et al. Factors to inform clinicians about the end of life in severe chronic obstructive pulmonary disease. J Pain Symptom Manage. 2013;46:491–9.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Gray-Donald K, Gibbons L, Shapiro SH, Macklem PT, Martin JG. Nutritional status and mortality in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1996;153:961–6.CrossRefPubMed Gray-Donald K, Gibbons L, Shapiro SH, Macklem PT, Martin JG. Nutritional status and mortality in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1996;153:961–6.CrossRefPubMed
44.
Zurück zum Zitat Schols AM, Slangen J, Volovics L, Wouters EF. Weight loss is a reversible factor in the prognosis of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157:1791–7.CrossRefPubMed Schols AM, Slangen J, Volovics L, Wouters EF. Weight loss is a reversible factor in the prognosis of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157:1791–7.CrossRefPubMed
45.
Zurück zum Zitat Hodgev VA, Kostianev SS. Maximal inspiratory pressure predicts mortality in patients with chronic obstructive pulmonary disease in a five-year follow-up. Folia Med (Plovdiv). 2006;48:36–41. Hodgev VA, Kostianev SS. Maximal inspiratory pressure predicts mortality in patients with chronic obstructive pulmonary disease in a five-year follow-up. Folia Med (Plovdiv). 2006;48:36–41.
46.
Zurück zum Zitat Budweiser S, Jorres RA, Criee CP, et al. Prognostic value of mouth occlusion pressure in patients with chronic ventilatory failure. Respir Med. 2007;101:2343–51.CrossRefPubMed Budweiser S, Jorres RA, Criee CP, et al. Prognostic value of mouth occlusion pressure in patients with chronic ventilatory failure. Respir Med. 2007;101:2343–51.CrossRefPubMed
47.
Zurück zum Zitat Meyer FJ, Borst MM, Zugck C, et al. Respiratory muscle dysfunction in congestive heart failure: clinical correlation and prognostic significance. Circulation. 2001;103:2153–8.CrossRefPubMed Meyer FJ, Borst MM, Zugck C, et al. Respiratory muscle dysfunction in congestive heart failure: clinical correlation and prognostic significance. Circulation. 2001;103:2153–8.CrossRefPubMed
48.
Zurück zum Zitat Frankenstein L, Nelles M, Meyer FJ, et al. Validity, prognostic value and optimal cutoff of respiratory muscle strength in patients with chronic heart failure changes with beta-blocker treatment. Eur J Cardiovasc Prev Rehabil. 2009;16:424–9.CrossRefPubMed Frankenstein L, Nelles M, Meyer FJ, et al. Validity, prognostic value and optimal cutoff of respiratory muscle strength in patients with chronic heart failure changes with beta-blocker treatment. Eur J Cardiovasc Prev Rehabil. 2009;16:424–9.CrossRefPubMed
49.
Zurück zum Zitat Ionescu AA, Chatham K, Davies CA, Nixon LS, Enright S, Shale DJ. Inspiratory muscle function and body composition in cystic fibrosis. Am J Respir Crit Care Med. 1998;158:1271–6.CrossRefPubMed Ionescu AA, Chatham K, Davies CA, Nixon LS, Enright S, Shale DJ. Inspiratory muscle function and body composition in cystic fibrosis. Am J Respir Crit Care Med. 1998;158:1271–6.CrossRefPubMed
50.
Zurück zum Zitat Marroni CA, Galant LH. Functional capacity, respiratory muscle strength, and oxygen consumption predict mortality in patients with cirrhosis. J Hepatol. 2014;60:S245 [Abstract]. Marroni CA, Galant LH. Functional capacity, respiratory muscle strength, and oxygen consumption predict mortality in patients with cirrhosis. J Hepatol. 2014;60:S245 [Abstract].
51.
Zurück zum Zitat van der Palen J, Rea TD, Manolio TA, et al. Respiratory muscle strength and the risk of incident cardiovascular events. Thorax. 2004;59:1063–7.CrossRefPubMedPubMedCentral van der Palen J, Rea TD, Manolio TA, et al. Respiratory muscle strength and the risk of incident cardiovascular events. Thorax. 2004;59:1063–7.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Polkey MI, Lyall RA, Yang K, Johnson E, Leigh PN, Moxham J. Respiratory muscle strength as a predictive biomarker for survival in amyotrophic lateral sclerosis. Am J Respir Crit Care Med. 2017;195:86–95.CrossRefPubMed Polkey MI, Lyall RA, Yang K, Johnson E, Leigh PN, Moxham J. Respiratory muscle strength as a predictive biomarker for survival in amyotrophic lateral sclerosis. Am J Respir Crit Care Med. 2017;195:86–95.CrossRefPubMed
53.
Zurück zum Zitat Phillips MF, Smith PE, Carroll N, Edwards RH, Calverley PM. Nocturnal oxygenation and prognosis in Duchenne muscular dystrophy. Am J Respir Crit Care Med. 1999;160:198–202.CrossRefPubMed Phillips MF, Smith PE, Carroll N, Edwards RH, Calverley PM. Nocturnal oxygenation and prognosis in Duchenne muscular dystrophy. Am J Respir Crit Care Med. 1999;160:198–202.CrossRefPubMed
54.
Zurück zum Zitat Frankenstein L, Meyer FJ, Sigg C, et al. Is serial determination of inspiratory muscle strength a useful prognostic marker in chronic heart failure? Eur J Cardiovasc Prev Rehabil. 2008;15:156–61.CrossRefPubMed Frankenstein L, Meyer FJ, Sigg C, et al. Is serial determination of inspiratory muscle strength a useful prognostic marker in chronic heart failure? Eur J Cardiovasc Prev Rehabil. 2008;15:156–61.CrossRefPubMed
55.
Zurück zum Zitat Habedank D, Meyer FJ, Hetzer R, Anker SD, Ewert R. Relation of respiratory muscle strength, cachexia and survival in severe chronic heart failure. J Cachexia Sarcopenia Muscle. 2013;4:277–85.CrossRefPubMedPubMedCentral Habedank D, Meyer FJ, Hetzer R, Anker SD, Ewert R. Relation of respiratory muscle strength, cachexia and survival in severe chronic heart failure. J Cachexia Sarcopenia Muscle. 2013;4:277–85.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Hui D, Bansal S, Morgado M, Dev R, Chisholm G, Bruera E. Phase angle for prognostication of survival in patients with advanced cancer: preliminary findings. Cancer. 2014;120:2207–14.CrossRefPubMedPubMedCentral Hui D, Bansal S, Morgado M, Dev R, Chisholm G, Bruera E. Phase angle for prognostication of survival in patients with advanced cancer: preliminary findings. Cancer. 2014;120:2207–14.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Jackson M, Smith I, King M, Shneerson J. Long term non-invasive domiciliary assisted ventilation for respiratory failure following thoracoplasty. Thorax. 1994;49:915–9.CrossRefPubMedPubMedCentral Jackson M, Smith I, King M, Shneerson J. Long term non-invasive domiciliary assisted ventilation for respiratory failure following thoracoplasty. Thorax. 1994;49:915–9.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Nizet TA, van den Elshout FJ, Heijdra YF, van de Ven MJ, Mulder PG, Folgering HT. Survival of chronic hypercapnic COPD patients is predicted by smoking habits, comorbidity, and hypoxemia. Chest. 2005;127:1904–10.CrossRefPubMed Nizet TA, van den Elshout FJ, Heijdra YF, van de Ven MJ, Mulder PG, Folgering HT. Survival of chronic hypercapnic COPD patients is predicted by smoking habits, comorbidity, and hypoxemia. Chest. 2005;127:1904–10.CrossRefPubMed
59.
Zurück zum Zitat White AC, Terrin N, Miller KB, Ryan HF. Impaired respiratory and skeletal muscle strength in patients prior to hematopoietic stem-cell transplantation. Chest. 2005;128:145–52.CrossRefPubMed White AC, Terrin N, Miller KB, Ryan HF. Impaired respiratory and skeletal muscle strength in patients prior to hematopoietic stem-cell transplantation. Chest. 2005;128:145–52.CrossRefPubMed
60.
Zurück zum Zitat Polkey MI, Kyroussis D, Hamnegard CH, Mills GH, Green M, Moxham J. Diaphragm strength in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1996;154:1310–7.CrossRefPubMed Polkey MI, Kyroussis D, Hamnegard CH, Mills GH, Green M, Moxham J. Diaphragm strength in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1996;154:1310–7.CrossRefPubMed
61.
Zurück zum Zitat Euteneuer S, Windisch W, Suchi S, Kohler D, Jones PW, Schonhofer B. Health-related quality of life in patients with chronic respiratory failure after long-term mechanical ventilation. Respir Med. 2006;100:477–86.CrossRefPubMed Euteneuer S, Windisch W, Suchi S, Kohler D, Jones PW, Schonhofer B. Health-related quality of life in patients with chronic respiratory failure after long-term mechanical ventilation. Respir Med. 2006;100:477–86.CrossRefPubMed
62.
Zurück zum Zitat Dellborg C, Olofson J, Midgren B, Caro O, Skoogh BE, Sullivan M. Quality of life in patients with chronic alveolar hypoventilation. Eur Respir J. 2002;19:113–20.CrossRefPubMed Dellborg C, Olofson J, Midgren B, Caro O, Skoogh BE, Sullivan M. Quality of life in patients with chronic alveolar hypoventilation. Eur Respir J. 2002;19:113–20.CrossRefPubMed
63.
Zurück zum Zitat Trojan DA, Arnold DL, Shapiro S, et al. Fatigue in post-poliomyelitis syndrome: association with disease-related, behavioral, and psychosocial factors. PM R. 2009;1:442–9.CrossRefPubMed Trojan DA, Arnold DL, Shapiro S, et al. Fatigue in post-poliomyelitis syndrome: association with disease-related, behavioral, and psychosocial factors. PM R. 2009;1:442–9.CrossRefPubMed
64.
Zurück zum Zitat Allen SM, Hunt B, Green M. Fall in vital capacity with posture. Br J Dis Chest. 1985;79:267–71.CrossRefPubMed Allen SM, Hunt B, Green M. Fall in vital capacity with posture. Br J Dis Chest. 1985;79:267–71.CrossRefPubMed
65.
Zurück zum Zitat Braun NM, Arora NS, Rochester DF. Respiratory muscle and pulmonary function in polymyositis and other proximal myopathies. Thorax. 1983;38:616–23.CrossRefPubMedPubMedCentral Braun NM, Arora NS, Rochester DF. Respiratory muscle and pulmonary function in polymyositis and other proximal myopathies. Thorax. 1983;38:616–23.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Hart N, Cramer D, Ward SP, et al. Effect of pattern and severity of respiratory muscle weakness on carbon monoxide gas transfer and lung volumes. Eur Respir J. 2002;20:996–1002.CrossRefPubMed Hart N, Cramer D, Ward SP, et al. Effect of pattern and severity of respiratory muscle weakness on carbon monoxide gas transfer and lung volumes. Eur Respir J. 2002;20:996–1002.CrossRefPubMed
67.
Zurück zum Zitat DePalo VA, McCool FD. Respiratory muscle evaluation of the patient with neuromuscular disease. Semin Respir Crit Care Med. 2002;23:201–9.CrossRefPubMed DePalo VA, McCool FD. Respiratory muscle evaluation of the patient with neuromuscular disease. Semin Respir Crit Care Med. 2002;23:201–9.CrossRefPubMed
68.
Zurück zum Zitat Jackson CE, Rosenfeld J, Moore DH, et al. A preliminary evaluation of a prospective study of pulmonary function studies and symptoms of hypoventilation in ALS/MND patients. J Neurol Sci. 2001;191:75–8.CrossRefPubMed Jackson CE, Rosenfeld J, Moore DH, et al. A preliminary evaluation of a prospective study of pulmonary function studies and symptoms of hypoventilation in ALS/MND patients. J Neurol Sci. 2001;191:75–8.CrossRefPubMed
69.
Zurück zum Zitat Inal-Ince D, Savci S, Arikan H, et al. Effects of scoliosis on respiratory muscle strength in patients with neuromuscular disorders. Spine J. 2009;9:981–6.CrossRefPubMed Inal-Ince D, Savci S, Arikan H, et al. Effects of scoliosis on respiratory muscle strength in patients with neuromuscular disorders. Spine J. 2009;9:981–6.CrossRefPubMed
70.
Zurück zum Zitat Prigent H, Orlikowski D, Letilly N, et al. Vital capacity versus maximal inspiratory pressure in patients with Guillain-Barre syndrome and myasthenia gravis. Neurocrit Care. 2012;17:236–9.CrossRefPubMed Prigent H, Orlikowski D, Letilly N, et al. Vital capacity versus maximal inspiratory pressure in patients with Guillain-Barre syndrome and myasthenia gravis. Neurocrit Care. 2012;17:236–9.CrossRefPubMed
71.
Zurück zum Zitat Gordon PH, Corcia P, Lacomblez L, et al. Defining survival as an outcome measure in amyotrophic lateral sclerosis. Arch Neurol. 2009;66:758–61.CrossRefPubMed Gordon PH, Corcia P, Lacomblez L, et al. Defining survival as an outcome measure in amyotrophic lateral sclerosis. Arch Neurol. 2009;66:758–61.CrossRefPubMed
72.
Zurück zum Zitat Abresch R, McDonald C, Han J, et al. Pulmonary function characteristics of boys with duchenne and Becker muscular dystrophy by age groups and steroid use: One-year data from the CINRG longitudinal study project. Neurology. 2010;74:A219. Abresch R, McDonald C, Han J, et al. Pulmonary function characteristics of boys with duchenne and Becker muscular dystrophy by age groups and steroid use: One-year data from the CINRG longitudinal study project. Neurology. 2010;74:A219.
73.
Zurück zum Zitat van der Ploeg AT, Clemens PR, Corzo D, et al. A randomized study of alglucosidase alfa in late-onset Pompe’s disease. N Engl J Med. 2010;362:1396–406.CrossRefPubMed van der Ploeg AT, Clemens PR, Corzo D, et al. A randomized study of alglucosidase alfa in late-onset Pompe’s disease. N Engl J Med. 2010;362:1396–406.CrossRefPubMed
74.
Zurück zum Zitat van der Ploeg AT, Barohn R, Carlson L, et al. Open-label extension study following the late-onset treatment study (LOTS) of alglucosidase Alfa. Mol Genet Metab. 2012;107:456–61.CrossRefPubMed van der Ploeg AT, Barohn R, Carlson L, et al. Open-label extension study following the late-onset treatment study (LOTS) of alglucosidase Alfa. Mol Genet Metab. 2012;107:456–61.CrossRefPubMed
75.
Zurück zum Zitat Cheah BC, Boland RA, Brodaty NE, et al. INSPIRATIonAL—INSPIRAtory muscle training in amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2009;10:384–92.CrossRefPubMed Cheah BC, Boland RA, Brodaty NE, et al. INSPIRATIonAL—INSPIRAtory muscle training in amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2009;10:384–92.CrossRefPubMed
76.
Zurück zum Zitat van der Beek NA, van Capelle CI, van der Velden-van Etten KI, et al. Rate of progression and predictive factors for pulmonary outcome in children and adults with Pompe disease. Mol Genet Metab. 2011;104:129–36.CrossRefPubMed van der Beek NA, van Capelle CI, van der Velden-van Etten KI, et al. Rate of progression and predictive factors for pulmonary outcome in children and adults with Pompe disease. Mol Genet Metab. 2011;104:129–36.CrossRefPubMed
77.
Zurück zum Zitat De Troyer A, Borenstein S, Cordier R. Analysis of lung volume restriction in patients with respiratory muscle weakness. Thorax. 1980;35:603–10.CrossRefPubMedPubMedCentral De Troyer A, Borenstein S, Cordier R. Analysis of lung volume restriction in patients with respiratory muscle weakness. Thorax. 1980;35:603–10.CrossRefPubMedPubMedCentral
78.
Zurück zum Zitat Chiang LL, Yu CT, Liu CY, Lo YL, Kuo HP, Lin HC. Six-month nocturnal nasal positive pressure ventilation improves respiratory muscle capacity and exercise endurance in patients with chronic hypercapnic respiratory failure. J Formos Med Assoc. 2006;105:459–67.CrossRefPubMed Chiang LL, Yu CT, Liu CY, Lo YL, Kuo HP, Lin HC. Six-month nocturnal nasal positive pressure ventilation improves respiratory muscle capacity and exercise endurance in patients with chronic hypercapnic respiratory failure. J Formos Med Assoc. 2006;105:459–67.CrossRefPubMed
79.
Zurück zum Zitat Shioya T, Satake M, Sato K, et al. Long-term effect of the beta2-receptor agonist procaterol on daily life performance and exercise capacity in patients with stable chronic obstructive pulmonary disease. Clinical study with special reference to health-related quality of life and activities of daily living. Arzneimittelforschung. 2008;58:24–8.PubMed Shioya T, Satake M, Sato K, et al. Long-term effect of the beta2-receptor agonist procaterol on daily life performance and exercise capacity in patients with stable chronic obstructive pulmonary disease. Clinical study with special reference to health-related quality of life and activities of daily living. Arzneimittelforschung. 2008;58:24–8.PubMed
80.
Zurück zum Zitat O’Brien K, Geddes EL, Reid WD, Brooks D, Crowe J. Inspiratory muscle training compared with other rehabilitation interventions in chronic obstructive pulmonary disease: a systematic review update. J Cardiopulm Rehabil Prev. 2008;28:128–41.CrossRefPubMed O’Brien K, Geddes EL, Reid WD, Brooks D, Crowe J. Inspiratory muscle training compared with other rehabilitation interventions in chronic obstructive pulmonary disease: a systematic review update. J Cardiopulm Rehabil Prev. 2008;28:128–41.CrossRefPubMed
81.
Zurück zum Zitat Borghi-Silva A, Mendes RG, Toledo AC, et al. Adjuncts to physical training of patients with severe COPD: oxygen or noninvasive ventilation? Respir Care. 2010;55:885–94.PubMed Borghi-Silva A, Mendes RG, Toledo AC, et al. Adjuncts to physical training of patients with severe COPD: oxygen or noninvasive ventilation? Respir Care. 2010;55:885–94.PubMed
82.
Zurück zum Zitat van Wetering CR, Hoogendoorn M, Broekhuizen R, et al. Efficacy and costs of nutritional rehabilitation in muscle-wasted patients with chronic obstructive pulmonary disease in a community-based setting: a prespecified subgroup analysis of the INTERCOM trial. J Am Med Dir Assoc. 2010;11:179–87.CrossRefPubMed van Wetering CR, Hoogendoorn M, Broekhuizen R, et al. Efficacy and costs of nutritional rehabilitation in muscle-wasted patients with chronic obstructive pulmonary disease in a community-based setting: a prespecified subgroup analysis of the INTERCOM trial. J Am Med Dir Assoc. 2010;11:179–87.CrossRefPubMed
83.
Zurück zum Zitat Deering BM, Fullen B, Egan C, et al. Acupuncture as an adjunct to pulmonary rehabilitation. J Cardiopulm Rehabil Prev. 2011;31:392–9.CrossRefPubMed Deering BM, Fullen B, Egan C, et al. Acupuncture as an adjunct to pulmonary rehabilitation. J Cardiopulm Rehabil Prev. 2011;31:392–9.CrossRefPubMed
84.
Zurück zum Zitat Pleguezuelos E, Perez ME, Guirao L, et al. Effects of whole body vibration training in patients with severe chronic obstructive pulmonary disease. Respirology. 2013;18:1028–34.CrossRefPubMed Pleguezuelos E, Perez ME, Guirao L, et al. Effects of whole body vibration training in patients with severe chronic obstructive pulmonary disease. Respirology. 2013;18:1028–34.CrossRefPubMed
85.
Zurück zum Zitat Wetzel JL, Fry DK, Pfalzer LA. Six-minute walk test for persons with mild or moderate disability from multiple sclerosis: performance and explanatory factors. Physiother Can. 2011;63:166–80.CrossRefPubMedPubMedCentral Wetzel JL, Fry DK, Pfalzer LA. Six-minute walk test for persons with mild or moderate disability from multiple sclerosis: performance and explanatory factors. Physiother Can. 2011;63:166–80.CrossRefPubMedPubMedCentral
86.
Zurück zum Zitat Dimitriadis Z, Kapreli E, Konstantinidou I, Oldham J, Strimpakos N. Test/retest reliability of maximum mouth pressure measurements with the MicroRPM in healthy volunteers. Respir Care. 2011;56:776–82.CrossRefPubMed Dimitriadis Z, Kapreli E, Konstantinidou I, Oldham J, Strimpakos N. Test/retest reliability of maximum mouth pressure measurements with the MicroRPM in healthy volunteers. Respir Care. 2011;56:776–82.CrossRefPubMed
87.
Zurück zum Zitat Hamnegard CH, Wragg S, Kyroussis D, Aquilina R, Moxham J, Green M. Portable measurement of maximum mouth pressures. Eur Respir J. 1994;7:398–401.CrossRefPubMed Hamnegard CH, Wragg S, Kyroussis D, Aquilina R, Moxham J, Green M. Portable measurement of maximum mouth pressures. Eur Respir J. 1994;7:398–401.CrossRefPubMed
88.
Zurück zum Zitat McElvaney G, Blackie S, Morrison NJ, Wilcox PG, Fairbarn MS, Pardy RL. Maximal static respiratory pressures in the normal elderly. Am Rev Respir Dis. 1989;139:277–81.CrossRefPubMed McElvaney G, Blackie S, Morrison NJ, Wilcox PG, Fairbarn MS, Pardy RL. Maximal static respiratory pressures in the normal elderly. Am Rev Respir Dis. 1989;139:277–81.CrossRefPubMed
89.
Zurück zum Zitat Hart N, Polkey MI, Sharshar T, et al. Limitations of sniff nasal pressure in patients with severe neuromuscular weakness. J Neurol Neurosurg Psychiatry. 2003;74:1685–7.CrossRefPubMedPubMedCentral Hart N, Polkey MI, Sharshar T, et al. Limitations of sniff nasal pressure in patients with severe neuromuscular weakness. J Neurol Neurosurg Psychiatry. 2003;74:1685–7.CrossRefPubMedPubMedCentral
90.
Zurück zum Zitat Morgan RK, McNally S, Alexander M, Conroy R, Hardiman O, Costello RW. Use of sniff nasal-inspiratory force to predict survival in amyotrophic lateral sclerosis. Am J Respir Crit Care Med. 2005;171:269–74.CrossRefPubMed Morgan RK, McNally S, Alexander M, Conroy R, Hardiman O, Costello RW. Use of sniff nasal-inspiratory force to predict survival in amyotrophic lateral sclerosis. Am J Respir Crit Care Med. 2005;171:269–74.CrossRefPubMed
91.
Zurück zum Zitat Khirani S, Ramirez A, Aubertin G, et al. Respiratory muscle decline in Duchenne muscular dystrophy. Pediatr Pulmonol. 2014;49:473–81.CrossRefPubMed Khirani S, Ramirez A, Aubertin G, et al. Respiratory muscle decline in Duchenne muscular dystrophy. Pediatr Pulmonol. 2014;49:473–81.CrossRefPubMed
92.
Zurück zum Zitat Fitting JW, Paillex R, Hirt L, Aebischer P, Schluep M. Sniff nasal pressure: a sensitive respiratory test to assess progression of amyotrophic lateral sclerosis. Ann Neurol. 1999;46:887–93.CrossRefPubMed Fitting JW, Paillex R, Hirt L, Aebischer P, Schluep M. Sniff nasal pressure: a sensitive respiratory test to assess progression of amyotrophic lateral sclerosis. Ann Neurol. 1999;46:887–93.CrossRefPubMed
93.
Zurück zum Zitat Stefanutti D, Benoist MR, Scheinmann P, Chaussain M, Fitting JW. Usefulness of sniff nasal pressure in patients with neuromuscular or skeletal disorders. Am J Respir Crit Care Med. 2000;162:1507–11.CrossRefPubMed Stefanutti D, Benoist MR, Scheinmann P, Chaussain M, Fitting JW. Usefulness of sniff nasal pressure in patients with neuromuscular or skeletal disorders. Am J Respir Crit Care Med. 2000;162:1507–11.CrossRefPubMed
94.
Zurück zum Zitat Terzi N, Orlikowski D, Fermanian C, et al. Measuring inspiratory muscle strength in neuromuscular disease: one test or two? Eur Respir J. 2008;31:93–8.CrossRefPubMed Terzi N, Orlikowski D, Fermanian C, et al. Measuring inspiratory muscle strength in neuromuscular disease: one test or two? Eur Respir J. 2008;31:93–8.CrossRefPubMed
95.
Zurück zum Zitat Gruis KL, Lechtzin N. Respiratory therapies for amyotrophic lateral sclerosis: a primer. Muscle Nerve. 2012;46:313–31.CrossRefPubMed Gruis KL, Lechtzin N. Respiratory therapies for amyotrophic lateral sclerosis: a primer. Muscle Nerve. 2012;46:313–31.CrossRefPubMed
Metadaten
Titel
Maximum inspiratory pressure as a clinically meaningful trial endpoint for neuromuscular diseases: a comprehensive review of the literature
verfasst von
Benedikt Schoser
Edward Fong
Tarekegn Geberhiwot
Derralynn Hughes
John T. Kissel
Shyam C. Madathil
David Orlikowski
Michael I. Polkey
Mark Roberts
Harm A. W. M. Tiddens
Peter Young
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Orphanet Journal of Rare Diseases / Ausgabe 1/2017
Elektronische ISSN: 1750-1172
DOI
https://doi.org/10.1186/s13023-017-0598-0

Weitere Artikel der Ausgabe 1/2017

Orphanet Journal of Rare Diseases 1/2017 Zur Ausgabe