Skip to main content
Erschienen in: BMC Pulmonary Medicine 1/2021

Open Access 01.12.2021 | Study protocol

Opioids in patients with COPD and refractory dyspnea: literature review and design of a multicenter double blind study of low dosed morphine and fentanyl (MoreFoRCOPD)

verfasst von: Marlies van Dijk, Kris J. M. Mooren, Jan-Willem K. van den Berg, Wendy J. C. van Beurden-Moeskops, Roxane Heller-Baan, Sander M. de Hosson, Wai Yee Lam-Wong, Liesbeth Peters, Karin Pool, Huib A. M. Kerstjens

Erschienen in: BMC Pulmonary Medicine | Ausgabe 1/2021

Abstract

Background

Refractory dyspnea or breathlessness is a common symptom in patients with advanced chronic obstructive pulmonary disease (COPD), with a high negative impact on quality of life (QoL). Low dosed opioids have been investigated for refractory dyspnea in COPD and other life-limiting conditions, and some positive effects were demonstrated. However, upon first assessment of the literature, the quality of evidence in COPD seemed low or inconclusive, and focused mainly on morphine which may have more side effects than other opioids such as fentanyl. For the current publication we performed a systematic literature search. We searched for placebo-controlled randomized clinical trials investigating opioids for refractory dyspnea caused by COPD. We included trials reporting on dyspnea, health status and/or QoL. Three of fifteen trials demonstrated a significant positive effect of opioids on dyspnea. Only one of four trials reporting on QoL or health status, demonstrated a significant positive effect. Two-thirds of included trials investigated morphine. We found no placebo-controlled RCT on transdermal fentanyl. Subsequently, we hypothesized that both fentanyl and morphine provide a greater reduction of dyspnea than placebo, and that fentanyl has less side effects than morphine.

Methods

We describe the design of a robust, multi-center, double blind, double-dummy, cross-over, randomized, placebo-controlled clinical trial with three study arms investigating transdermal fentanyl 12 mcg/h and morphine sustained-release 10 mg b.i.d. The primary endpoint is change in daily mean dyspnea sensation measured on a numeric rating scale. Secondary endpoints are change in daily worst dyspnea, QoL, anxiety, sleep quality, hypercapnia, side effects, patient preference, and continued opioid use. Sixty patients with severe stable COPD and refractory dyspnea (FEV1 < 50%, mMRC ≥ 3, on optimal standard therapy) will be included.

Discussion

Evidence for opioids for refractory dyspnea in COPD is not as robust as usually appreciated. We designed a study comparing both the more commonly used opioid morphine, and transdermal fentanyl to placebo. The cross-over design will help to get a better impression of patient preferences. We believe our study design to investigate both sustained-release morphine and transdermal fentanyl for refractory dyspnea will provide valuable information for better treatment of refractory dyspnea in COPD.
Trial registration NCT03834363 (ClinicalTrials.gov), registred at 7 Feb 2019, https://​clinicaltrials.​gov/​ct2/​show/​NCT03834363.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12890-021-01647-8.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AE
Adverse event
CCMO
Central Committee on Research Involving Human Subjects
CCQ
Clinical COPD Questionnaire
CRQ
Chronic Respiratory Questionnaire
COPD
Chronic obstructive pulmonary disease
eGFR
Estimated Glomerular Filtration Rate
FEV1
Forced expiratory volume in 1 s
FVC
Forced vital capacity
GOLD
Global initiative for chronic obstructive lung disease
HR-QoL
Health Related Quality of Life
HADS-A
Hospital Anxiety and Depression Scale—anxiety subscale
IC
Informed consent
LAN
Lung Alliance The Netherlands
mMRC
Modified Medical Research Council Dyspnea Scale
MREC
Medical Research Ethics Committee
NRS
Numeric rating scale
SAE
Serious adverse event
SR
Sustained-release

Background

Refractory dyspnea or breathlessness is a common symptom in patients with advanced chronic obstructive pulmonary disease (COPD), with a prevalence of up to 94% in the last year of life [1, 2]. It is defined as persisting complaints of dyspnea despite optimal standard therapy including, but not limited to smoking cessation, education, inhaled bronchodilators and pulmonary physiotherapy [3]. Refractory dyspnea is known to severely impact quality of life and exercise tolerance, and to increase the risk of depression and anxiety [4]. As the prevalence of COPD is expected to rise during the upcoming decades [5], it is likely that the number of patients with COPD suffering from refractory dyspnea will also continue to grow.
Advanced treatments such as non-invasive ventilation, bronchoscopic lung volume reduction and lung transplantation can improve dyspnea and quality of life in patients with advanced COPD [6]. But these treatments are only available for a proportion of patients with advanced COPD, due to strict eligibility criteria, high health-care costs and sometimes scarcity. Therefore, there is still a need for more widely available treatments of refractory dyspnea. In this context low dosed opioids have previously been investigated, and some positive effect was demonstrated [79]. However, whether the quality of the evidence is sufficient is still a topic of discussion. Furthermore, despite a positive advice on opioids in palliative care guidelines for COPD, prescription appears to be low in clinical practice [1012].
We performed a systematic literature search with respect to opioids for refractory dyspnea in COPD, which we updated for the current publication to include all recent trials. We searched for placebo-controlled randomized clinical trials investigating any type of opioid prescribed for dyspnea reduction in COPD (at least 50% of participants). We included trials reporting on dyspnea, health status and/or quality of life. Additional details on the search strategy can be found in the online supplement (Additional file 1: Online supplement MoreFoRCOPD), including a flow chart on the number of records identified, screened and included.
Table 1 shows an overview of the trials we identified as a result of our search strategy. In total, fifteen trials were included. A statistically significant positive effect on dyspnea of opioid versus placebo was demonstrated only in three studies [7, 8, 13]. Since the majority of these studies included a small number of patients, the lack of statistically significant results may in part be explained by a low statistical power to detect a treatment effect. This assumption is supported by a meta-analysis published by Ekström et al. in 2015, in which a positive effect on dyspnea was found for both systemically administered and nebulized opioids (analyses of combined data of 8 and 4 trials, respectively) [14]. Nevertheless, the three largest studies in our table, which all have been published more recently, demonstrated no significant change in dyspnea for sustained-release morphine and oxycodone [1517]. While assessing this, it is important to note that in the studies of Currow et al. [15] and Ferriera et al. [16] (which were originally both part of a three-armed trial) all arms received immediate-release morphine as needed. For both studies, the immediate-release morphine was used significantly more frequently in the placebo group (8.7 vs. 5.8 and 7.0 vs. 4.2 daily doses, respectively) [15, 16] making an overall effect of the maintenance morphine more difficult to detect. Furthermore, in the study by Verberkt et al. there was a statistically significant effect on worst daily dyspnea measured on a numeric rating scale (NRS) in a subgroup of COPD patients with a modified Medical Research Council (mMRC) ≥ 3 (mean difference compared to placebo: − 1.33 (− 2.50 to − 0.16) points) [17].
Table 1
Overview of randomized clinical trials investigating the effect of opioids on dyspnea, quality of life or health status in COPD
References
Design
n (% COPD)
Setting
Comparison
Treatment duration
Breathlessness
Quality of life or health status
Measurement (scale)
Opioid
Placebo
Measurement (scale)
Opioid
Placebo
Woodcock et al. [18]
Cross-over
12 (100)
Outpatient
Dihydrocodeine
Single dose
VAS (0–10 cm)
45 min after med
5.54 ± 1.91
6.33 ± 2.0
Light et al. [19]
Cross-over
13 (100)
Outpatient
Oral morphine 0.8 mg/kg
Single dose
Borg score (0–10)
Rest
0.29 ± 0.58
0.13 ± 0.23
Jankelson et al. [20]
Cross-over
16 (100)
Outpatient
Nebulized morphine 20/40 mg
Single dose
Borg score (0–10)
After 6MWT
4.2 ± 2.1/4.3 ± 1.8
4.3 ± 2.2
Noseda et al. [21]
Cross-over
14 (79)#
Hospitalized
Nebulized morphine 10/20 mg ± oxygen
Single dose
VAS (− 100 to + 100%)
 + 33 ± 28/+ 43 ± 27
 + 42 ± 27
Jensen et al. [22]
Cross-over
12 (100)
Outpatient
Nebulized fentanyl 50 µg
Single dose
Borg score (0–10)
Isotime CPET
2.0 ± 0.5
2.6 ± 0.5
Abdallah et al. [13]
Cross-over
20 (100)
Outpatient
Morphine dose up to 10 mg
Single dose
Borg score (0–10)
Isotime CPET
3.0 ± 1.6*
4.2 ± 2.6
Iupati et al. [23]
Cross-over
Multicenter
21 (62)
Outpatient
Intranasal fentanyl 20 µg as needed
1 day
VAS (0–100 mm)
15 min after med
26 ± 21 (Δ29 ± 25)
21 ± 19 (Δ33 ± 24)
Abernethy et al. [7]
Cross-over
Multicenter
48 (88)#
Outpatient
SR morphine 20 mg od
4 days
VAS (0–100 mm)
Morning/evening
40.1 ± 24*/40.3 ± 23*
47.7 ± 26
49.9 ± 24
Data not presented
Data not presented
Data not presented
Janowiak et al. [24]
Cross-over
10 (100)
Hospitalized
Nebulized morphine 3–5 mg qid
4 days
VAS (0–100 mm)
Now (2dd)
Δ25.4 ± 9.0$
Δ6.3 ± 7.8
Johnson et al. [8]
Cross-over
18 (100)
Outpatient
Dihydrocodeine 15 mg as needed up to tds
7 days
VAS (0–10 cm)
Mean daily
4.6 ± 2.1*
5.6 ± 2.3
Currow et al. [15]
Parallel
Multicenter
284 (58)#
Outpatient
SR morphine 20 mg qd
All arms: morphine 2.5 mg as needed
7 days
VAS (0–100 mm)
Now (2dd)
Δ-5.00 ± 2.13
Δ-4.86 ± 2.07
EORTC-QLQ-C15 PAL (0–100)
Δ1.8 ± 2.2
Δ1.5 ± 2.2
Ferriera et al. [16]
Parallel
Multicenter
155 (60)#
Outpatient
Oxycodone 5 mg tds
All arms: morphine 2.5 mg as needed
7 days
VAS (0–100 mm)
Now
Δ-3.7 ± 2.9
Δ-9.0 ± 2.7
EORTC-QLQ-C15 PAL (0–100)
Δ-1.7 ± 3.1
Δ2.82 ± 3.1
Eiser et al. [25]
Cross-over
14 (100)
Outpatient
Diamorphine 2.5/5 mg
qid
14 days
VAS (0–10 cm)
7.0 ± 0.7/7.0 ± 0.8
6.5 ± 0.7
Verberkt et al. [17]
Parallel
Multicenter
124 (100)
Outpatient
SR morphine 10 mg 1-tds
28 days
NRS (0–10 points)
Mean
Δ-0.60 (− 1.55 to 0.35)
CAT (0–40)
Δ-2.18 (− 4.14 to − 0.2)*
Poole et al. [9]
Cross-over
16 (100)
Outpatient
SR morphine 10 mg od or bid
42 days
DBS (0–5)
2.22
2.26
CRQ (20–140)
∆2.08 ± 4.53
∆2.94 ± 3.46
Data presented as mean ± SD
od once a day, bid twice daily, tds three times a day, qid four times a day, SR sustained release, VAS visual analogue score, DBS daytime breathlessness score, NRS numeric rating scale, CRQ chronic respiratory questionnaire, EORTC-QLQ-C15 PAL Quality of life questionnaire developed by the European Organisation for Research and Treatment of Cancer, CAT COPD assessment test, CPET cardiopulmonary exercise testing, 6MWT 6-min walking test
*p < 0.05 opioid versus placebo, $p < 0.05 change after treatment. #Data not exclusively on COPD
Information on quality of life or health status was limited to four RCT’s. Of these, only the study by Verberkt et al. demonstrated a small positive, statistically significant effect on health status measured with the COPD assessment test (CAT) [17]. Our search identified no placebo-controlled RCT’s investigating transdermal fentanyl for refractory dyspnea in COPD.
Based on this assessment of available evidence, we designed a randomized, placebo-controlled clinical trial, on which we will further elaborate in the “Methods/design” section and “Discussion” section.

Methods/design

Overview

We designed a robust, multi-center, double blind, double-dummy, cross-over, randomized, placebo-controlled clinical trial with three study arms investigating transdermal fentanyl and sustained-release morphine. We hypothesize that both fentanyl and morphine provide a reduction of dyspnea which is greater than placebo, and that fentanyl has less side effects than morphine. A total of 60 patients with severe stable COPD and refractory dyspnea will be included in this study in ten Dutch hospitals. Patients will be recruited at the outpatient clinic of each participating hospital by chest physicians. The study is registered at clinicaltrials.gov (NCT03834363), where a full list of participating hospitals can be found, and the protocol is approved by the UMCG Ethics committee. Written informed consent will be obtained from all participants and the study will be performed in accordance with the Declaration of Helsinki.

Study duration and treatment

The study duration is 6 weeks for each participant, divided in three periods of 2 weeks. During each period the participant is treated for 11 days. During the first 3 days of every treatment period no study medication is used, to wash out medication of any previous treatment period. The fentanyl patches are dosed 12 µg/h and changed every 3 days. The morphine sustained-released capsules are dosed 10 mg b.i.d. Both an antiemetic (metoclopramide 10 mg as needed, up to thrice daily) and laxative (macrogol/electrolytes 13.7 g, started once daily, more or less sachets as needed) are prescribed. In total, there are four study visits. A complete study flowchart can be found in Fig. 1. After the end of the study treatment patients can discuss with their chest physician whether they would like to continue with low dosed morphine or transdermal fentanyl. At the time of this decision, the participants and physician are still blinded to the study treatment.

In- and exclusion criteria

All in- and exclusion criteria can be found in Table 2. In general, patients with COPD Gold class III or IV and a modified Medical Research Council score (mMRC) ≥ 3 who perceive dyspnea despite optimal standard therapy according to GOLD and the Dutch guideline for diagnosis and treatment of COPD can be included. If there is comorbidity substantially contributing to the breathlessness, for example severe heart failure, patients are excluded. Participants who have a moderate or severe exacerbation (requiring oral corticosteroids, antibiotics and/or hospital admission) during participation are discontinued from the trial. If they are stable for 8 weeks after recovery from the exacerbation, they are allowed to restart the study once more.
Table 2
In- and exclusion criteria
Inclusion criteria
Age ≥ 40 years
Read, understood and signed the Informed Consent form
COPD GOLD class III or IV, according to GOLD criteria
 Post-bronchodilatation FEV1/FVC ≤ 70% and FEV1 < 50% pred.*
Complaints of refractory dyspnea as established by patient and doctor
mMRC score ≥ 3
Life expectancy of ≥ 2 months
Optimized standard therapy according to Dutch LAN guideline for diagnosis and treatment of COPD
Exclusion criteria
Other severe disease with chronic pain or chronic dyspnea (a non-susbstantial component of left sided heart failure is acceptable)
Current use of opioids for whatever indication
Allergy/intolerance for opioids
Psychiatric disease, not related to severe COPD
Exacerbation of COPD 8 weeks prior to inclusion or between screening and randomization
Problematic (leading to medical help or social problems) substance abuse during the last 5 years
Active malignancy, with the exception of planocellular or basal cell carcinoma of the skin
eGFR < 15 ml/min*
*Measured within 6 months of screening
COPD chronic obstructive pulmonary disease, FEV1 forced expiratory volume in 1 s, FVC forced vital capacity, GOLD global initiative for chronic obstructive lung disease, LAN Lung Alliance The Netherlands, mMRC modified Medical Research Council Dyspnea Scale, eGFR estimated Glomerular Filtration Rate

Outcome measurements

The primary outcome measurement is change in mean daily dyspnea sensation as measured on the numeric rating scale for Dyspnea [26]. Secondary outcome measurements are change in worst daily dyspnea sensation, health-related quality of life, anxiety, sleep quality, occurrence of respiratory depression and side effects, patient preference and continued opioid use. A more extensive description of the outcome measures can be found in Table 3. Patients who drop out will be followed as much as possible for vital status, hospitalization, and start of open label opioids during the intended 6 weeks period of the study.
Table 3
Outcome measurements
 
Measurement
Frequency of measurement
Primary outcome measure
  
Change in mean dyspnea sensation
Numeric rating scale [26]
Once daily in patient diary
Secondary outcome measures
  
Change in worst dyspnea sensation
Numeric rating scale [26]
Once daily in patient diary
Change in Health-Related Quality of Life
CCQ [27]
Once daily in patient diary
CRQ [28]
During each study visit
CRQ-mastery domain
During each study visit
HADS-A [29]
Open en named side effects
During each study visit
Once daily in patient diary and asked during study visits
Anxiety
Side effects
Change in hypercapnia, HCO3 and pH
Change in sleep quality
Patient preference
Continued opioid use
Capillary blood gas analysis
Numeric rating scale [30]
Asked during the final study visit
Asked 3 months after the end of study
During each study visit
Once daily in patient diary
Once
Once
CCQ clinical COPD questionnaire, CRQ chronic respiratory questionnaire, HADS-A hospital anxiety and depression scale—anxiety subscale

Randomization and unblinding

Randomization is tailor made for this study using a web based program (ALEA® DM version 17.1). Randomization can be performed online by the research team of each participating hospital. Participants will be randomized equally between the six possible treatment sequences, stratified for study location. Unblinding only occurs in the case of patient emergencies and at the conclusion of the study. Health authorities will be granted access to unblinded data if needed. The pharmacist on call of each participating hospital can unblind a participant using the web based program if requested by the researcher because of a patient emergency.

Statistical analysis

For the power calculation the difference in primary endpoint between fentanyl and placebo was used. The Minimal Clinical Important Difference (MCID) for the NRS score is 1 point, the standard deviation is 2.0 points [31]. With a two-sided alpha = 0.05 and a power of 0.90 in a cross-over design, 44 participants who complete the study are needed. Because this is a fragile patient group, we will aim to recruit 60 participants.
The primary endpoint analysis will be on an intention to treat basis and therefore all patients randomized. The primary endpoint is the NRS mean dyspnea score which we will treat as a continuous variable for day 7–14. This will not be calculated if less dan 2 days are available. Since it is a three way cross-over, the data for the available periods will also be used of not all periods were completed. No imputation will be used for the primary endpoint. There will be two comparisons: the difference in the mean dyspnea score of day 7–14 for fentanyl versus placebo and for morphine versus placebo. In this way, the risk of any remaining effect from the previous treatment periods influencing the outcome will be optimally reduced. The analysis will be by Student’s t-test. The analyses of secondary endpoints will be done by Student’s t-tests (or non-parametric tests where needed) or chi square, following the same scheme of main comparisons as for the primary endpoints. The analysis of side effects will be done by comparison of proportions of side effects by chi square tests between all three arms. Composite questionnaire data will be primarily analysed by total sum scores. Additionally, per protocol analyses will be performed. The study is not powered to determine equivalence of dyspnea relief of fentanyl compared to morphine: that comparison will consist of descriptive statistics only.

Safety

All (serious) adverse events will be monitored. The sponsor will report serious adverse events (SAEs) through the Dutch web portal ToetsingOnline to the accredited Ethics committee that approved the protocol, within 7 days of first knowledge for SAEs that result in death or are life threatening followed by a period of maximum of 8 days to complete the initial preliminary report. All other SAEs will be reported within a period of maximum 15 days after the sponsor has first knowledge of the serious adverse events. This is a short term study with 60 patients, entered parallel in a multi-centre study. Therefore, and since opioids in the form of morphine are in the guidelines, we will not perform interim analyses, even though the patient population of patients with severe COPD and in a palliative setting is at increased risk of death. For the same reasons, no Data Safety Monitoring Board (DSMB) will be instituted.

Study timeline

The study has started in November 2019. At this point the first participant was included at the University Medical Center Groningen. For the other participating hospitals the start of inclusion was delayed by one or more months because of a delay in the production of research medication and a delay in the issuing of a permit for scientific research with opioids for the participating hospital pharmacies. Unfortunately, starting March 2020 the inclusion was alternately put on hold or restricted in each participating hospital due to the COVID-19 pandemic. We aim to include all patients by the end of 2021, but whether this will be achieved is strongly depended on the course of the COVID-19 pandemic.

Discussion

Optimal reduction of dyspnea in patients with severe COPD is an important way to improve quality of life, yet can be very challenging. From our assessment of the literature, we found that even though opioids have found their way into COPD guidelines as a treatment option for refractory dyspnea, the evidence base can still be considered inconclusive. Furthermore, the majority of research has focused on morphine and we identified no placebo-controlled RCT investigating transdermal fentanyl. However, trials investigating fentanyl in the short-acting form, suggest that fentanyl could give a reduction of dyspnea [32, 33]. Additionally, studies on pain treatment indicate that patients may prefer transdermal fentanyl and experience less side effects in comparison to oral morphine [34]. Therefore, we believe that our current multi-center, double blind, cross-over, placebo-controlled study design to investigate sustained-release morphine and transdermal fentanyl for refractory dyspnea will provide valuable information on patient preference and the effectiveness of transdermal fentanyl and sustained-release morphine for refractory dyspnea in COPD.
By choosing a cross-over design for this study the participant is his or her own control, thus reducing the variability and the number of patients needed to participate. Additionally, this design helps to get a better impression of patient preferences. On the other hand, because of the cross-over design the treatment duration is 6 weeks instead of 11 days (which it would be if this study had a parallel design). This prolonged study duration will most likely increase the risk of participants that have to be discontinued from the trial because of the occurrence of COPD exacerbations, which occur frequently in advanced COPD. For this reason we aim to include 60 participants, which is sixteen more than the 44 participants calculated from the power analysis which need to fully complete the study. Furthermore, patients experiencing an exacerbation will discontinue the trial, but may be included once more if they are clinically stable for at least 8 weeks.
There are indications that prescription of opioids for refractory dyspnea in COPD can be a loaded topic for both patient and doctors, amongst others because of associations with terminal disease, possible adverse effects and addiction [10]. Although this has not been formally investigated in patients, we believe education is important to address any questions or worries patients may have regarding opioids. Therefore, both an animated short film for patients and their loved ones on facts and myths about opioids (developed by Indiveo B.V.) as well as an information leaflet with the same content are tested during our study. At the end of the trial, feedback from the participants will be used to adjust the animation and leaflet and these will be made widely available for patients with COPD. Additionally, both patients and physicians participating in the study are asked to share their experiences with opioids for refractory dyspnea in COPD during regional congresses and meetings.

Acknowledgements

The investigators would like to thank Daisy J. Janssen for advising on the research protocol. The investigators would like to thank the members of the investigational teams in all participating hospitals for all the time and effort it takes to carry out this study.

Protocol version

Protocol version 14, date 14th February 2020, has been approved by the ethics committee of the University Medical Center Groningen Medical Research.

Declarations

The study is designed in accordance with the Declaration of Helsinki and approved by the ethics committee of the University Medical Center Groningen Medical Research Ethics Committee (Netherlands). All amendments will be communicated with relevant parties. Written informed consent will be obtained from all participants by a member of the research team of each participating hospital. Each participant will be covered by insurance in case of unexpected harm from participating in the trial.
The ethical approval and patient information include consent to publish collected data.

Competing interests

MD, KMM, JWB, WB, RH, SMH, WYL, LEP, KP, HAK report no competing interests related to this study.
Open AccessThis 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.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge
Literatur
1.
Zurück zum Zitat Carette H, Zysman M, Morelot-Panzini C, Perrin J, Gomez E, Guillaumot A, et al. Prevalence and management of chronic breathlessness in COPD in a tertiary care center. BMC Pulm Med. 2019;19(1):95.CrossRef Carette H, Zysman M, Morelot-Panzini C, Perrin J, Gomez E, Guillaumot A, et al. Prevalence and management of chronic breathlessness in COPD in a tertiary care center. BMC Pulm Med. 2019;19(1):95.CrossRef
2.
Zurück zum Zitat Edmonds P, Karlsen S, Khan S, Addington-Hall J. A comparison of the palliative care needs of patients dying from chronic respiratory diseases and lung cancer. Palliat Med. 2001;15(4):287–95.CrossRef Edmonds P, Karlsen S, Khan S, Addington-Hall J. A comparison of the palliative care needs of patients dying from chronic respiratory diseases and lung cancer. Palliat Med. 2001;15(4):287–95.CrossRef
3.
Zurück zum Zitat Mahler DA, Selecky PA, Harrod CG, Benditt JO, Carrieri-Kohlman V, Curtis JR, et al. American College of Chest Physicians consensus statement on the management of dyspnea in patients with advanced lung or heart disease. Chest. 2010;137(3):674–91.CrossRef Mahler DA, Selecky PA, Harrod CG, Benditt JO, Carrieri-Kohlman V, Curtis JR, et al. American College of Chest Physicians consensus statement on the management of dyspnea in patients with advanced lung or heart disease. Chest. 2010;137(3):674–91.CrossRef
4.
Zurück zum Zitat Janssen DJ, Wouters EF, Spruit MA. Psychosocial consequences of living with breathlessness due to advanced disease. Curr Opin Support Palliat Care. 2015;9(3):232–7.CrossRef Janssen DJ, Wouters EF, Spruit MA. Psychosocial consequences of living with breathlessness due to advanced disease. Curr Opin Support Palliat Care. 2015;9(3):232–7.CrossRef
5.
Zurück zum Zitat Singh D, Agusti A, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease: the GOLD science committee report 2019. Eur Respir J. 2019;53(5):190. Singh D, Agusti A, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease: the GOLD science committee report 2019. Eur Respir J. 2019;53(5):190.
6.
Zurück zum Zitat van Dijk M, Gan CT, Koster TD, Wijkstra PJ, Slebos DJ, Kerstjens HAM, et al. Treatment of severe stable COPD: the multidimensional approach of treatable traits. ERJ Open Res. 2020;6(3):00322–2019.CrossRef van Dijk M, Gan CT, Koster TD, Wijkstra PJ, Slebos DJ, Kerstjens HAM, et al. Treatment of severe stable COPD: the multidimensional approach of treatable traits. ERJ Open Res. 2020;6(3):00322–2019.CrossRef
7.
Zurück zum Zitat Abernethy AP, Currow DC, Frith P, Fazekas BS, McHugh A, Bui C. Randomised, double blind, placebo controlled crossover trial of sustained release morphine for the management of refractory dyspnoea. BMJ. 2003;327(7414):523–8.CrossRef Abernethy AP, Currow DC, Frith P, Fazekas BS, McHugh A, Bui C. Randomised, double blind, placebo controlled crossover trial of sustained release morphine for the management of refractory dyspnoea. BMJ. 2003;327(7414):523–8.CrossRef
8.
Zurück zum Zitat Johnson MA, Woodcock AA, Geddes DM. Dihydrocodeine for breathlessness in “pink puffers.” Br Med J (Clin Res Ed). 1983;286(6366):675–7.CrossRef Johnson MA, Woodcock AA, Geddes DM. Dihydrocodeine for breathlessness in “pink puffers.” Br Med J (Clin Res Ed). 1983;286(6366):675–7.CrossRef
9.
Zurück zum Zitat Poole PJ, Veale AG, Black PN. The effect of sustained-release morphine on breathlessness and quality of life in severe chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157(6 Pt 1):1877–80.CrossRef Poole PJ, Veale AG, Black PN. The effect of sustained-release morphine on breathlessness and quality of life in severe chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157(6 Pt 1):1877–80.CrossRef
10.
Zurück zum Zitat Janssen DJ, de Hosson SM, bij de Vaate E, Mooren KJ, Baas AA. Attitudes toward opioids for refractory dyspnea in COPD among Dutch chest physicians. Chron Respir Dis. 2015;12(2):85–92.CrossRef Janssen DJ, de Hosson SM, bij de Vaate E, Mooren KJ, Baas AA. Attitudes toward opioids for refractory dyspnea in COPD among Dutch chest physicians. Chron Respir Dis. 2015;12(2):85–92.CrossRef
11.
Zurück zum Zitat Ahmadi Z, Bernelid E, Currow DC, Ekstrom M. Prescription of opioids for breathlessness in end-stage COPD: a national population-based study. Int J Chron Obstruct Pulmon Dis. 2016;11:2651–7.CrossRef Ahmadi Z, Bernelid E, Currow DC, Ekstrom M. Prescription of opioids for breathlessness in end-stage COPD: a national population-based study. Int J Chron Obstruct Pulmon Dis. 2016;11:2651–7.CrossRef
12.
Zurück zum Zitat Currow DC, Johnson MJ, Pollack A, Ferreira DH, Kochovska S, Ekstrom M, et al. Breathlessness and opioid prescribing in COPD in general practice: a cross-sectional, observational study. ERJ Open Res. 2020;6(2):00299–2019.CrossRef Currow DC, Johnson MJ, Pollack A, Ferreira DH, Kochovska S, Ekstrom M, et al. Breathlessness and opioid prescribing in COPD in general practice: a cross-sectional, observational study. ERJ Open Res. 2020;6(2):00299–2019.CrossRef
13.
Zurück zum Zitat Abdallah SJ, Wilkinson-Maitland C, Saad N, Li PZ, Smith BM, Bourbeau J, et al. Effect of morphine on breathlessness and exercise endurance in advanced COPD: a randomised crossover trial. Eur Respir J. 2017;50(4):1701235.CrossRef Abdallah SJ, Wilkinson-Maitland C, Saad N, Li PZ, Smith BM, Bourbeau J, et al. Effect of morphine on breathlessness and exercise endurance in advanced COPD: a randomised crossover trial. Eur Respir J. 2017;50(4):1701235.CrossRef
14.
Zurück zum Zitat Ekstrom M, Nilsson F, Abernethy AA, Currow DC. Effects of opioids on breathlessness and exercise capacity in chronic obstructive pulmonary disease. A systematic review. Ann Am Thorac Soc. 2015;12(7):1079–92.CrossRef Ekstrom M, Nilsson F, Abernethy AA, Currow DC. Effects of opioids on breathlessness and exercise capacity in chronic obstructive pulmonary disease. A systematic review. Ann Am Thorac Soc. 2015;12(7):1079–92.CrossRef
15.
Zurück zum Zitat Currow D, Louw S, McCloud P, Fazekas B, Plummer J, McDonald CF, et al. Regular, sustained-release morphine for chronic breathlessness: a multicentre, double-blind, randomised, placebo-controlled trial. Thorax. 2020;75(1):50–6.CrossRef Currow D, Louw S, McCloud P, Fazekas B, Plummer J, McDonald CF, et al. Regular, sustained-release morphine for chronic breathlessness: a multicentre, double-blind, randomised, placebo-controlled trial. Thorax. 2020;75(1):50–6.CrossRef
16.
Zurück zum Zitat Ferreira DH, Louw S, McCloud P, Fazekas B, McDonald CF, Agar MR, et al. Controlled-release oxycodone versus placebo in the treatment of chronic breathlessness—a multisite randomized placebo controlled trial. J Pain Symptom Manag. 2020;59(3):581–9.CrossRef Ferreira DH, Louw S, McCloud P, Fazekas B, McDonald CF, Agar MR, et al. Controlled-release oxycodone versus placebo in the treatment of chronic breathlessness—a multisite randomized placebo controlled trial. J Pain Symptom Manag. 2020;59(3):581–9.CrossRef
17.
Zurück zum Zitat Verberkt CA, van den Beuken-van Everdingen MHJ, Schols J, Hameleers N, Wouters EFM, Janssen DJA. Effect of sustained-release morphine for refractory breathlessness in chronic obstructive pulmonary disease on health status: a randomized clinical trial. JAMA Intern Med. 2020;180(10):1306–14.CrossRef Verberkt CA, van den Beuken-van Everdingen MHJ, Schols J, Hameleers N, Wouters EFM, Janssen DJA. Effect of sustained-release morphine for refractory breathlessness in chronic obstructive pulmonary disease on health status: a randomized clinical trial. JAMA Intern Med. 2020;180(10):1306–14.CrossRef
18.
Zurück zum Zitat Woodcock AA, Gross ER, Gellert A, Shah S, Johnson M, Geddes DM. Effects of dihydrocodeine, alcohol, and caffeine on breathlessness and exercise tolerance in patients with chronic obstructive lung disease and normal blood gases. N Engl J Med. 1981;305(27):1611–6.CrossRef Woodcock AA, Gross ER, Gellert A, Shah S, Johnson M, Geddes DM. Effects of dihydrocodeine, alcohol, and caffeine on breathlessness and exercise tolerance in patients with chronic obstructive lung disease and normal blood gases. N Engl J Med. 1981;305(27):1611–6.CrossRef
19.
Zurück zum Zitat Light RW, Muro JR, Sato RI, Stansbury DW, Fischer CE, Brown SE. Effects of oral morphine on breathlessness and exercise tolerance in patients with chronic obstructive pulmonary disease. Am Rev Respir Dis. 1989;139(1):126–33.CrossRef Light RW, Muro JR, Sato RI, Stansbury DW, Fischer CE, Brown SE. Effects of oral morphine on breathlessness and exercise tolerance in patients with chronic obstructive pulmonary disease. Am Rev Respir Dis. 1989;139(1):126–33.CrossRef
20.
Zurück zum Zitat Jankelson D, Hosseini K, Mather LE, Seale JP, Young IH. Lack of effect of high doses of inhaled morphine on exercise endurance in chronic obstructive pulmonary disease. Eur Respir J. 1997;10(10):2270–4.CrossRef Jankelson D, Hosseini K, Mather LE, Seale JP, Young IH. Lack of effect of high doses of inhaled morphine on exercise endurance in chronic obstructive pulmonary disease. Eur Respir J. 1997;10(10):2270–4.CrossRef
21.
Zurück zum Zitat Noseda A, Carpiaux JP, Markstein C, Meyvaert A, de Maertelaer V. Disabling dyspnoea in patients with advanced disease: lack of effect of nebulized morphine. Eur Respir J. 1997;10(5):1079–83.CrossRef Noseda A, Carpiaux JP, Markstein C, Meyvaert A, de Maertelaer V. Disabling dyspnoea in patients with advanced disease: lack of effect of nebulized morphine. Eur Respir J. 1997;10(5):1079–83.CrossRef
22.
Zurück zum Zitat Jensen D, Alsuhail A, Viola R, Dudgeon DJ, Webb KA, O’Donnell DE. Inhaled fentanyl citrate improves exercise endurance during high-intensity constant work rate cycle exercise in chronic obstructive pulmonary disease. J Pain Symptom Manag. 2012;43(4):706–19.CrossRef Jensen D, Alsuhail A, Viola R, Dudgeon DJ, Webb KA, O’Donnell DE. Inhaled fentanyl citrate improves exercise endurance during high-intensity constant work rate cycle exercise in chronic obstructive pulmonary disease. J Pain Symptom Manag. 2012;43(4):706–19.CrossRef
23.
Zurück zum Zitat Iupati S, Bridge R, Allan S, Hewitt D. Intranasal fentanyl versus placebo for treatment of episodic breathlessness in hospice patients with advanced nonmalignant diseases. J Pain Symptom Manag. 2020;61:1035–41.CrossRef Iupati S, Bridge R, Allan S, Hewitt D. Intranasal fentanyl versus placebo for treatment of episodic breathlessness in hospice patients with advanced nonmalignant diseases. J Pain Symptom Manag. 2020;61:1035–41.CrossRef
24.
Zurück zum Zitat Janowiak P, Krajnik M, Podolec Z, Bandurski T, Damps-Konstanska I, Sobanski P, et al. Dosimetrically administered nebulized morphine for breathlessness in very severe chronic obstructive pulmonary disease: a randomized, controlled trial. BMC Pulm Med. 2017;17(1):186.CrossRef Janowiak P, Krajnik M, Podolec Z, Bandurski T, Damps-Konstanska I, Sobanski P, et al. Dosimetrically administered nebulized morphine for breathlessness in very severe chronic obstructive pulmonary disease: a randomized, controlled trial. BMC Pulm Med. 2017;17(1):186.CrossRef
25.
Zurück zum Zitat Eiser N, Denman WT, West C, Luce P. Oral diamorphine: lack of effect on dyspnoea and exercise tolerance in the “pink puffer” syndrome. Eur Respir J. 1991;4(8):926–31.PubMed Eiser N, Denman WT, West C, Luce P. Oral diamorphine: lack of effect on dyspnoea and exercise tolerance in the “pink puffer” syndrome. Eur Respir J. 1991;4(8):926–31.PubMed
26.
Zurück zum Zitat Gift AG, Narsavage G. Validity of the numeric rating scale as a measure of dyspnea. Am J Crit Care. 1998;7(3):200–4.CrossRef Gift AG, Narsavage G. Validity of the numeric rating scale as a measure of dyspnea. Am J Crit Care. 1998;7(3):200–4.CrossRef
27.
Zurück zum Zitat van der Molen T, Willemse BW, Schokker S, ten Hacken NH, Postma DS, Juniper EF. Development, validity and responsiveness of the Clinical COPD questionnaire. Health Qual Life Outcomes. 2003;1:13.CrossRef van der Molen T, Willemse BW, Schokker S, ten Hacken NH, Postma DS, Juniper EF. Development, validity and responsiveness of the Clinical COPD questionnaire. Health Qual Life Outcomes. 2003;1:13.CrossRef
28.
Zurück zum Zitat Wijkstra PJ, TenVergert EM, Van Altena R, Otten V, Postma DS, Kraan J, et al. Reliability and validity of the chronic respiratory questionnaire (CRQ). Thorax. 1994;49(5):465–7.CrossRef Wijkstra PJ, TenVergert EM, Van Altena R, Otten V, Postma DS, Kraan J, et al. Reliability and validity of the chronic respiratory questionnaire (CRQ). Thorax. 1994;49(5):465–7.CrossRef
29.
Zurück zum Zitat Zigmond AS, Snaith RP. The hospital anxiety and depression scale. Acta Psychiatr Scand. 1983;67(6):361–70.CrossRef Zigmond AS, Snaith RP. The hospital anxiety and depression scale. Acta Psychiatr Scand. 1983;67(6):361–70.CrossRef
30.
Zurück zum Zitat Cappelleri JC, Bushmakin AG, McDermott AM, Sadosky AB, Petrie CD, Martin S. Psychometric properties of a single-item scale to assess sleep quality among individuals with fibromyalgia. Health Qual Life Outcomes. 2009;7:54.CrossRef Cappelleri JC, Bushmakin AG, McDermott AM, Sadosky AB, Petrie CD, Martin S. Psychometric properties of a single-item scale to assess sleep quality among individuals with fibromyalgia. Health Qual Life Outcomes. 2009;7:54.CrossRef
31.
Zurück zum Zitat Johnson MJ, Bland JM, Oxberry SG, Abernethy AP, Currow DC. Clinically important differences in the intensity of chronic refractory breathlessness. J Pain Symptom Manag. 2013;46(6):957–63.CrossRef Johnson MJ, Bland JM, Oxberry SG, Abernethy AP, Currow DC. Clinically important differences in the intensity of chronic refractory breathlessness. J Pain Symptom Manag. 2013;46(6):957–63.CrossRef
32.
Zurück zum Zitat Hui D, Hernandez F, Larsson L, Liu D, Kilgore K, Naberhuis J, et al. Prophylactic fentanyl sublingual spray for episodic exertional dyspnea in cancer patients: a pilot double-blind randomized controlled trial. J Pain Symptom Manag. 2019;58(4):605–13.CrossRef Hui D, Hernandez F, Larsson L, Liu D, Kilgore K, Naberhuis J, et al. Prophylactic fentanyl sublingual spray for episodic exertional dyspnea in cancer patients: a pilot double-blind randomized controlled trial. J Pain Symptom Manag. 2019;58(4):605–13.CrossRef
33.
Zurück zum Zitat Simon ST, Koskeroglu P, Gaertner J, Voltz R. Fentanyl for the relief of refractory breathlessness: a systematic review. J Pain Symptom Manag. 2013;46(6):874–86.CrossRef Simon ST, Koskeroglu P, Gaertner J, Voltz R. Fentanyl for the relief of refractory breathlessness: a systematic review. J Pain Symptom Manag. 2013;46(6):874–86.CrossRef
34.
Zurück zum Zitat Payne R, Mathias SD, Pasta DJ, Wanke LA, Williams R, Mahmoud R. Quality of life and cancer pain: satisfaction and side effects with transdermal fentanyl versus oral morphine. J Clin Oncol. 1998;16(4):1588–93.CrossRef Payne R, Mathias SD, Pasta DJ, Wanke LA, Williams R, Mahmoud R. Quality of life and cancer pain: satisfaction and side effects with transdermal fentanyl versus oral morphine. J Clin Oncol. 1998;16(4):1588–93.CrossRef
Metadaten
Titel
Opioids in patients with COPD and refractory dyspnea: literature review and design of a multicenter double blind study of low dosed morphine and fentanyl (MoreFoRCOPD)
verfasst von
Marlies van Dijk
Kris J. M. Mooren
Jan-Willem K. van den Berg
Wendy J. C. van Beurden-Moeskops
Roxane Heller-Baan
Sander M. de Hosson
Wai Yee Lam-Wong
Liesbeth Peters
Karin Pool
Huib A. M. Kerstjens
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
BMC Pulmonary Medicine / Ausgabe 1/2021
Elektronische ISSN: 1471-2466
DOI
https://doi.org/10.1186/s12890-021-01647-8

Weitere Artikel der Ausgabe 1/2021

BMC Pulmonary Medicine 1/2021 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.