Skip to main content
Erschienen in: Respiratory Research 1/2020

Open Access 01.12.2020 | Review

Tiotropium in chronic obstructive pulmonary disease – a review of clinical development

verfasst von: Antonio Anzueto, Marc Miravitlles

Erschienen in: Respiratory Research | Ausgabe 1/2020

Abstract

Background

Bronchodilators are the mainstay of pharmacological treatment in chronic obstructive pulmonary disease (COPD), and long-acting muscarinic antagonist (LAMA) monotherapy is recommended as initial treatment for Global Initiative for Chronic Obstructive Lung Disease (GOLD) groups B, C, and D.

Main body

Tiotropium bromide was the first LAMA available for COPD in clinical practice and, because of its long duration of action, is administered once daily. Tiotropium was initially available as an inhalation powder delivered via a dry-powder inhaler (DPI). Later, tiotropium also became available as an inhalation spray delivered via a soft mist inhaler (SMI). The SMI was designed to overcome or minimize some of the issues associated with other inhaler types (eg, the need for strong inspiratory airflow with DPIs). Results of short- and long-term randomized, controlled clinical trials of tiotropium in patients with COPD indicated tiotropium was safe and significantly improved lung function, health-related quality of life, and exercise endurance, and reduced dyspnea, lung hyperinflation, exacerbations, and use of rescue medication compared with placebo or active comparators. These positive efficacy findings triggered the evaluation of tiotropium in fixed-dose combination with olodaterol (a long-acting β2-agonist). In this review, we provide an overview of studies of tiotropium for the treatment of COPD, with a focus on pivotal studies.

Conclusion

Tiotropium is safe and efficacious as a long-term, once-daily LAMA for the maintenance treatment of COPD and for reducing COPD exacerbations. The SMI generates a low-velocity, long-duration aerosol spray with a high fine-particle fraction, which results in marked lung drug deposition. In addition, high inspiratory flow rates are not required.
Hinweise
An accompanying infographic is provided as Additional file 1.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12931-020-01407-y.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AUC0–24
area under the curve for the 24-h period post-morning dose
BID
twice daily
CI
confidence interval
COPD
chronic obstructive pulmonary disease
DPI
dry-powder inhaler
EU
European Union
FEV1
forced expiratory volume in 1 s
FVC
forced vital capacity
GLOW
glycopyrronium bromide in COPD airways
GOLD
Global Initiative for Chronic Obstructive Lung Disease
HCRU
health care resource utilization
HR
hazard ratio
HRQoL
health-related quality of life
IC
inspiratory capacity
ICS
inhaled corticosteroid
INHANCE
INdacaterol to Help Achieve New COPD treatment Excellence
INSPIRE
Investigating New Standards for Prophylaxis in Reducing Exacerbations
INTENSITY
INdacaterol Towards Establishment of cliNical SuperiorITY study
INTIME
INdacaterol & TIotropium: Measuring Efficacy
INVIGORATE
Indacaterol: Providing Opportunity to Re-engage Patients with Life
LABA
long-acting β2-agonist
LAMA
long-acting muscarinic antagonist
MISTRAL
Mesure de l’Influence de Spiriva® sur les Troubles Respiratoires Aigus à Long terme
OR
odds ratio
PEFR
peak expiratory flow rate
pMDI
pressurized metered-dose inhaler
POET-COPD
Prevention Of Exacerbations with Tiotropium in COPD
PY
patient-year
QD
once daily
QID
four times a day
RCT
randomized controlled trial
RR
rate ratio
SABA
short-acting β2-agonist
SAMA
short-acting muscarinic antagonist
SGRQ
St. George’s Respiratory Questionnaire
SMI
soft mist inhaler
TDI
transition dyspnea index
TIOSPIR
Tiotropium Safety and Performance In Respimat
TIPHON
Tiotropium: Influence sur la Perception de l’amélioration des activites Habituelles Objectivée par une echelle Numerique
UPLIFT
Understanding Potential Long-Term Impacts on Function with Tiotropium
US
United States

Background

Bronchodilators are the mainstay of pharmacological treatment in chronic obstructive pulmonary disease (COPD) [1]. The 2020 update of the Global Initiative for Chronic Obstructive Lung Disease (GOLD) strategy document includes a model for initiation of pharmacological treatment, which is based on assessment of symptoms and exacerbation risk according to the ABCD assessment tool [2]. After initial treatment, the GOLD committee recommends regular review of symptoms; assessment of inhaler technique and adherence, as well as non-pharmacological approaches; and treatment adjustment as needed. A separate follow-up treatment algorithm is provided that is based on the predominant treatable trait (dyspnea or exacerbations) and current treatment, and is independent of ABCD group at initiation of treatment [2]. Long-acting muscarinic antagonist (LAMA) monotherapy is recommended as initial treatment for GOLD groups B, C, and D [2]. Alternatively, long-acting β2-agonist (LABA) monotherapy can be used as initial treatment for GOLD group B, and dual bronchodilator therapy with a LABA/LAMA combination may be considered for group B patients with severe breathlessness. Dual bronchodilator therapy also is an initial treatment option for group D patients who are highly symptomatic (COPD Assessment Test [CAT] score > 20), and a combination of a LABA and inhaled corticosteroid (ICS) is an initial treatment option for group D patients with a blood eosinophil count of 300 cells/μL or more. In general, ICS-based therapy recommendations are guided by exacerbation history, blood eosinophil counts, and coexistent asthma in the GOLD 2020 update [2].
LAMAs inhibit the bronchoconstrictor effect of acetylcholine by prolonged binding to the M3 muscarinic receptors present on airway smooth muscles and faster dissociation from M2 receptors [2, 3]. Tiotropium bromide, the first LAMA available for COPD in clinical practice, with a dissociation half-life of 35 h from the M3 receptor, is structurally related to ipratropium [4, 5]. Ipratropium has a short duration of action, with a dissociation half-life of 0.3 h from the M3 receptor and requires four-times-a-day (QID) dosing, potentially affecting adherence to therapy [4, 5]. In contrast, tiotropium has a long duration of action, enabling once-daily (QD) dosing [5]. Tiotropium was first available as an inhalation powder delivered via a dry-powder inhaler (DPI; Spiriva® HandiHaler®; Boehringer Ingelheim Pharmaceuticals, Inc) and later became available as an inhalation spray delivered via a soft mist inhaler (SMI; Spiriva® Respimat®; Boehringer Ingelheim Pharmaceuticals, Inc). Tiotropium HandiHaler® and tiotropium Respimat® are indicated for long-term, QD maintenance treatment of bronchospasm associated with COPD and for reducing COPD exacerbations in the United States (US), European Union (EU), and other countries [6, 7]. A LABA/LAMA combination is used to leverage their different mechanisms of action. While LAMAs prevent bronchoconstriction, LABAs relax bronchial smooth muscle and cause bronchodilation by stimulating β2-adrenergic receptors in airway smooth muscle which trigger cellular pathways [8]. Tiotropium in a fixed-dose combination with the LABA olodaterol (Stiolto® Respimat® inhalation spray [Spiolto® Respimat® inhalation solution in Europe]; Boehringer Ingelheim Pharmaceuticals, Inc) is indicated for the long-term, QD maintenance treatment of patients with COPD [9, 10].
DPIs, pressurized metered-dose inhalers (pMDIs), and SMIs are used for delivery of inhaled COPD medications. However, patients with COPD might not be able to generate the inspiratory flow necessary for optimal drug de-aggregation and lung deposition with DPIs [11]. DPIs with varying internal resistances are available [11], and patients’ peak inspiratory flow should ideally be assessed before prescribing a DPI [12]. Further, the velocity of the aerosol from pMDIs and the inability of some patients with COPD to properly coordinate actuation and inhalation when using pMDIs can lead to considerable oropharyngeal drug deposition [11]. Use of a spacer or holding chamber with a pMDI helps slow the velocity of aerosolized particles; removes larger, nonrespirable particles; and reduces oropharyngeal deposition [13]. Respimat®, the only available SMI, is a propellant-free inhaler that uses mechanical energy to generate a fine, slow-moving mist [14]. Respimat® provides a higher lung drug deposition than pMDIs or DPIs [1517], delivers the dose of medication independent of the patient’s inspiratory effort [18], and requires minimal coordinated actuation and inhalation [19, 20], thereby making it suitable for a range of patients with COPD [14].
Cumulative results of clinical trials indicate that tiotropium significantly improves lung function (vs placebo [2133], ipratropium [27, 34], or salmeterol [35, 36]), reduces dyspnea (vs placebo [22, 28], salmeterol [35], or ipratropium [34]), reduces exacerbations (vs placebo [2224, 26, 28, 3638], ipratropium [34], or salmeterol [39]), and improves health-related quality of life (HRQoL; vs placebo [22, 23, 26, 28, 29, 32, 35, 36] or ipratropium [34]; Fig. 1). As reviewed here, most of the evidence base for use of tiotropium in COPD was derived from studies of the initial dry powder formulation. Results of studies of tiotropium as an inhalation solution delivered via an SMI confirmed its long-term efficacy benefits and safety profile. The clinical development of tiotropium in COPD culminated with evidence supporting its use in fixed-dose combination with olodaterol. The objective of this review is to comprehensively detail the key clinical trial evidence for use of tiotropium monotherapy in COPD.

Initial studies with tiotropium inhalation powder

Dose response and comparison with ipratropium

In a 4-week, dose-response study of tiotropium inhalation powder (4.5, 9, 18, and 36 μg QD) in patients with COPD (n = 169), all doses significantly improved lung function compared with placebo (day 29; difference in trough forced expiratory volume in 1 s [FEV1] response = 0.14 L, 0.11 L, 0.15 L, and 0.19 L, respectively; p < 0.05 for all) [31]. Similarly, forced vital capacity (FVC) was higher for all doses of tiotropium than with placebo (day 29; difference in trough FVC response = 0.31 L, 0.22 L, 0.37 L, and 0.22 L, respectively; p < 0.05 for 4.5-μg and 18.0-μg doses). All tiotropium doses also increased peak expiratory flow rate (PEFR) compared with placebo at all test points (morning, noon, and evening) [31]. Improvement in lung function was maintained throughout the treatment period. The overall safety profile of the four tiotropium doses was similar to that of placebo [31]. Based on these results, a dosage of 18 μg QD was evaluated in a 3-month, randomized controlled trial (RCT; n = 470) [21]. Again, tiotropium significantly improved lung function compared with placebo (day 92; difference in trough FEV1 response = 0.15 L; difference in trough FVC response = 0.28 L; p < 0.001 for both), and improvements were maintained throughout the treatment period. Tiotropium also significantly improved daily morning and evening PEFR and reduced symptoms of wheezing, shortness of breath, and “as-needed” albuterol use. Safety of tiotropium was similar to that of placebo, except for dry mouth, which was more common with tiotropium than with placebo (9.3% vs 1.6%; p < 0.05) [21]. Further, the efficacy and safety of tiotropium 18 μg QD was compared with that of ipratropium 40 μg QID (via a pMDI) in a 13-week study (n = 288) [40]. Tiotropium improved lung function compared with ipratropium throughout the treatment period (day 92; mean difference in trough FEV1 response = 0.13 L [p = 0.0001]; mean difference in trough FVC response = 0.21 L [p = 0.0003]). Tiotropium also consistently improved weekly mean morning and evening PEFR and reduced the use of rescue salbutamol. As observed in other studies, dry mouth was more common with tiotropium than with ipratropium (14.7% vs 10.3%) [40].

Pivotal trials

In a 6-month RCT (n = 623), tiotropium 18 μg QD compared with the LABA salmeterol 50 μg twice daily (BID; delivered via a pMDI) significantly improved lung function (week 24; difference in trough FEV1 response = 0.052 L; difference in trough FVC response = 0.112 L; p < 0.01 for both) and reduced dyspnea (week 24; difference in transition dyspnea index [TDI] focal score = 0.78 U; p < 0.05) [35]. Both active drugs significantly reduced the need for rescue albuterol compared with placebo (p < 0.0001). In this study, tiotropium also significantly improved HRQoL compared with placebo (week 24; difference in St. George’s Respiratory Questionnaire [SGRQ] total score = − 2.71 U; p < 0.05) but not salmeterol (difference = − 1.6 U; p > 0.05). Safety of tiotropium was similar to that of placebo and salmeterol, except for dry mouth, which was more common with tiotropium (10%) [35]. Health outcomes were evaluated in two 6-month RCTs (n = 1207) in which tiotropium 18 μg compared with placebo significantly reduced the number of exacerbations/patient-year (PY; 1.07 vs 1.49; p < 0.05), increased the time to first exacerbation (p ≤ 0.01), and improved HRQoL (difference in SGRQ total score = − 2.7 U; p < 0.01) during the treatment period [36]. Salmeterol did not significantly differ from placebo for these outcomes. Although both active drugs improved lung function, tiotropium did so to a greater extent than salmeterol. Dry mouth was more common with tiotropium (8.2%) than with salmeterol (1.7%) or placebo (2.3%) [36].
In two identical 12-month RCTs (n = 921), tiotropium 18 μg QD significantly improved lung function compared with placebo (difference in trough FEV1 response = 0.12–0.15 L [range over days 1–344]; p < 0.01) [22]. In addition, compared with patients who received placebo, those who received tiotropium also had significantly less dyspnea (difference in TDI focal score = 0.8–1.1 U [range over days 50–344]; p < 0.001), better health status scores (p < 0.05), and fewer COPD exacerbations/PY (0.76 vs 0.95; p = 0.045) and exacerbation-related hospitalizations (p < 0.05). In this and other tiotropium trials, patients were allowed to continue taking glucocorticoids during the study period. As observed in prior studies, incidence of dry mouth was higher with tiotropium than with placebo (16.0% vs 2.7%; p < 0.05). In two other identical 12-month RCTs (n = 535), tiotropium 18 μg QD compared with ipratropium 40 μg QID (delivered via a pMDI) significantly improved lung function (difference in trough FEV1 response = 0.15 L; p < 0.001) and reduced the use of rescue salbutamol (p < 0.05) at the end of the treatment period [34]. In addition, tiotropium compared with ipratropium significantly reduced dyspnea (day 364; difference in TDI focal score = 0.90 U; p = 0.001) and the number of exacerbations/PY (0.73 vs 0.96; p = 0.006); improved PEFR (difference in morning PEFR = 10–18 L/min [range over days 1–365]; difference in evening PEFR = 9–18 L/min [range over days 1–365]; p < 0.01) and HRQoL (day 364; difference in SGRQ total score = − 3.30; p = 0.004); and increased the time to first exacerbation (p = 0.008) and time to first exacerbation-related hospitalization (p = 0.048). These improvements were maintained throughout the treatment period. Collectively, results of these studies showed that QD tiotropium 18 μg was a safe and efficacious LAMA for maintenance treatment of COPD and for reducing exacerbations, leading to the approval of Spiriva® HandiHaler® in the EU (2002), US (2004), and other countries [41, 42].

Effect on lung hyperinflation, exercise endurance, exertional dyspnea, and HRQoL

Patients with COPD often experience hyperinflation, which results in reduced inspiratory capacity (IC), limited exercise capacity, and increased exertional dyspnea [2, 43]. A few RCTs were designed to specifically evaluate the effects of tiotropium on these parameters.
In a 4-week RCT (n = 81), tiotropium 18 μg QD significantly improved lung function and IC compared with placebo (mean differences at week 4: trough FEV1 = 0.16 L; trough FVC = 0.33 L; trough IC = 0.22 L; p < 0.01 for all) [44]. Similarly, in a 6-week RCT (n = 187), tiotropium significantly improved lung function compared with placebo, consistent with findings from prior studies [45]. Further, tiotropium compared with placebo significantly reduced lung hyperinflation (residual volume, p < 0.001; functional residual capacity, p < 0.001); increased vital capacity (p < 0.0001), IC (difference in trough response = 0.10 L; p < 0.05), and exercise endurance (difference in endurance time = 105 s [21%]; p < 0.01); and decreased exertional dyspnea (p < 0.01) on day 42 [45]. Change from baseline in all parameters was evident after the first treatment and persisted for the duration of the trial. In a subsequent 6-week study (n = 261), effects of tiotropium on lung hyperinflation, symptom-limited exercise tolerance, and exertional dyspnea were apparent at 2.25 h of treatment and lasted for 8 h after dosing on day 42 [46]. These findings were corroborated by results of another RCT (n = 100), where tiotropium 18 μg compared with placebo not only significantly improved trough FVC (difference = 0.20 L; p < 0.05) and trough IC (difference = 0.15 L; p < 0.05) but also significantly increased mean distance walked during the shuttle-walk test (difference = 36 m; p < 0.05) and improved HRQoL (difference in SGRQ total score = − 6.5; p = 0.026) after 12 weeks of treatment [32]. Further, in an RCT (n = 554) by the Tiotropium: Influence sur la Perception de l’amélioration des activites Habituelles Objectivée par une echelle Numerique (TIPHON) group—in addition to significantly improving lung function (difference in trough FEV1 = 0.10 L; p = 0.0001) and reducing exacerbations/PY (1.05 vs 1.83; p = 0.0287)—tiotropium compared with placebo significantly increased the proportion of patients achieving clinically relevant improvement in HRQoL (difference in responders = 10.9%; p = 0.029) after 9 months of treatment [23]. In contrast to the results of the above studies, in a 96-week RCT (n = 519), the difference in endurance time between tiotropium 18 μg QD and placebo was not statistically significant (tiotropium/placebo = 1.13; 95% CI = 0.97–1.32; p = 0.106). However, consistent with previous studies, tiotropium improved HRQoL at 96 weeks compared with placebo (difference in SGRQ total score = 4.03 U; p = 0.007) [47].

Effect on exacerbations

Patients with COPD, even those with mild disease, can experience exacerbations [4851], which account for a large proportion of total COPD burden on the healthcare system [52, 53]. Recommendations for reducing COPD exacerbations and treatment of stable COPD have been provided by GOLD [2], the European Respiratory Society/American Thoracic Society [54], and the Spanish Society of Pulmonology and Thoracic Surgery [55, 56]. The efficacy of tiotropium 18 μg QD in reducing exacerbations was evaluated in several short- and long-term studies, including those mentioned above (Table 1; summarized by Anzueto et al. [57]).
Table 1
Tiotropium reduces exacerbation and increases time to first exacerbation
Study
Comparator
Patients (N)
Study duration
Change from baseline in number of exacerbations
Change from baseline in patients with ≥ 1 exacerbation
Increase in time to first exacerbation
Casaburi 2002 [22]
Placebo
921
1 year
−20% (p = 0.045)
− 14% (p < 0.05)
p = 0.011
Vincken 2002 [34]
Ipratropium
535
1 year
−24% (p = 0.006)
−24% (p = 0.014)
p = 0.008
Brusasco 2003 [36]
Placebo
802a
6 months
−28% (p < 0.05)
−18% (p > 0.05)
p ≤ 0.01
Niewoehner 2005 [37]
Placebo
1829
6 months
−19% (p = 0.031)
−14% (p = 0.037)
p = 0.028
Dusser 2006 [24]
Placebo
1010
1 year
−35% (p < 0.001)
− 17% (p < 0.01)
p < 0.001
Tashkin 2008 [26]
Placebo
5993
4 years
− 14% (p < 0.001)
−2% (p = 0.35)
p < 0.001
Tonnel 2008 [23]
Placebo
554
9 months
−43% (p = 0.0287)
− 16% (p = 0.1013)
p = 0.0081
Bateman 2010 [28]
Placebo
1990
1 year
p < 0.01**
−16% (p < 0.01)b
p < 0.0001b
Bateman 2010 [29]
Placebo
3991
48 weeks
−21% (p < 0.0001)
− 18% (p < 0.0001)
p < 0.0001
Vogelmeier 2011 [39]
Salmeterol
7376
1 year
−11% (p = 0.002)c
−11% (p < 0.001)c
p < 0.001
**p < 0.01 for tiotropium 5 μg dose and p < 0.001 for tiotropium 10 μg dose
aThis number does not include the patients in the salmeterol group. bFor both tiotropium 5-μg and 10-μg doses. cRefers to moderate or severe exacerbations
In an RCT (n = 1829) conducted in a US Veterans Affairs setting, fewer patients treated with tiotropium than placebo experienced ≥1 exacerbation (difference = − 5.7%; p = 0.037) or exacerbation-related hospitalization (difference = − 3.0%; p = 0.056) after 6 months of treatment [37]. Analysis of secondary outcomes indicated that tiotropium significantly increased time to first exacerbation (p = 0.028) and reduced health care resource utilization (HCRU; frequency of hospitalizations, p = 0.047; antibiotic treatment days, p = 0.015; and unscheduled clinic visits, p = 0.019). Similar results were observed in another RCT (n = 1010; Mesure de l’Influence de Spiriva® sur les Troubles Respiratoires Aigus à Long terme [MISTRAL]) [24], where tiotropium compared with placebo significantly increased time to first exacerbation (p < 0.001) and reduced the number of exacerbations/PY (1.57 vs 2.41; p < 0.001), proportion of patients with ≥1 exacerbation (difference = − 10.4%; p < 0.01), and HCRU (concomitant respiratory medications, p < 0.0001; antibiotics, p < 0.001; and oral steroids, p < 0.01; and the number of unscheduled physician contacts, p < 0.05) after treatment for 1 year [24]. Tiotropium also significantly improved weekly morning PEFR (mean difference over 1 year = 25 L/min; p < 0.0001), trough FEV1 (mean difference = 0.12 L; p < 0.0001), FVC (mean difference = 0.17 L; p < 0.0001), and IC (mean difference = 0.14 L; p < 0.001) at the end of the treatment period. As observed in other studies, the safety of tiotropium was similar to that of placebo, except for dry mouth, which was more frequent with tiotropium (4.0%) than with placebo (1.4%) [24]. In addition, in the Prevention Of Exacerbations with Tiotropium in COPD (POET-COPD) trial (n = 7376), tiotropium 18 μg QD was significantly more efficacious than salmeterol 50 μg BID in increasing the time to first exacerbation (hazard ratio [HR] = 0.83 [ie, 17% reduction in risk of exacerbations with tiotropium]; p < 0.001) and reducing the annual rate of moderate or severe exacerbations (0.64 vs 0.72; rate ratio [RR] = 0.89 [11% reduction with tiotropium]; p = 0.002) and severe exacerbations (0.09 vs 0.13; RR = 0.73 [ie, 27% reduction with tiotropium]; p < 0.001) after 1 year of treatment [39]. The safety of tiotropium was similar to that of salmeterol. In a post hoc analysis of POET-COPD, tiotropium compared with salmeterol increased the time to first exacerbation and reduced the number of exacerbations in patients at low and high risk of exacerbation (time to first exacerbation: HR = 0.89; p = 0.1046 and HR = 0.84; p = 0.0002, respectively; number of exacerbations: RR = 0.89; p = 0.1768 and RR = 0.90; p = 0.0383, respectively) [58].

Longer-term studies

Longer-term studies were designed to expand upon findings from the 6- and 12-month RCTs. In a 2-year RCT (n = 841) of patients with early-stage COPD (ie, GOLD stage 1 [mild] or 2 [moderate]) in China, tiotropium HandiHaler® 18 μg QD compared with placebo significantly improved lung function throughout the 2-year period (range of mean differences in FEV1: before bronchodilator use = 0.127–0.169 L; after bronchodilator use = 0.071–0.133 L; p < 0.001 for both) [25]. In addition, in the Understanding Potential Long-Term Impacts on Function with Tiotropium (UPLIFT) trial (n = 5993), tiotropium compared with placebo significantly improved lung function (range of mean differences in FEV1: before bronchodilator use = 0.087–0.103 L; after bronchodilator use = 0.047–0.065 L; p < 0.001 for both), reduced the number of exacerbations/PY (0.73 vs 0.85; relative risk = 0.86 [ie, 14% reduction with tiotropium]; p < 0.001), increased the time to first exacerbation (p < 0.001), improved HRQoL (mean difference in SGRQ total score = − 2.7 U; p < 0.001), and reduced mortality (HR = 0.87; 95% confidence interval [CI] = 0.76–0.99) in patients with moderate-to-very severe COPD treated for 4 years [26]. Tiotropium significantly improved FEV1 and HRQoL compared with placebo throughout the trial.
Conflicting results were reported, however, with respect to the effect of tiotropium on annual decline in FEV1. Tiotropium compared with placebo significantly reduced the annual decline in FEV1 after bronchodilator use in the aforementioned 2-year RCT in China [25] (mean decline in FEV1 from day 30 to month 24: before bronchodilator use = 0.038 L/year vs 0.053 L/year [p = 0.06]; after bronchodilator use = 0.029 L/year vs 0.051 L/year [p = 0.006]) and in a retrospective analysis of two 1-year RCTs (mean decline in trough FEV1 from days 8 to 344 = 0.012 vs 0.058 L/year; p = 0.005) [59]. In UPLIFT, however, the annual decline in FEV1 was not significantly different between the tiotropium and placebo groups (mean decline in FEV1 from day 30 to month 48: before bronchodilator use = 0.030 L/year vs 0.030 L/year [p = 0.95]; after bronchodilator use = 0.040 L/year vs 0.042 L/year [p = 0.21]) [26]. In a pre-specified subgroup analysis of the UPLIFT trial in patients with GOLD stage II COPD, the annual decline in post-bronchodilator FEV1 was lower in the tiotropium group compared with the placebo group (mean decline in post-bronchodilator FEV1 from day 30 to month 48: 0.043 L/year vs 0.049 L/year [p = 0.024]), supporting benefits of early intervention in COPD [60].
Long-term effects of tiotropium as first-line maintenance medication in COPD were further evaluated in a secondary analysis of data from the UPLIFT trial [61]. In addition, various post hoc and subgroup analyses were conducted to assess the long-term efficacy of 4 years of treatment with tiotropium with regard to smoking status [62], sex [63], and baseline FEV1 ≥ 60% predicted [64]. In the latter analysis (n = 1210), tiotropium compared with placebo significantly improved lung function (difference in pre-bronchodilator FEV1 = 0.087–0.127 L [range over months 1–48]; difference in post-bronchodilator FEV1 = 0.038–0.084 L [range over months 1–48]; p ≤ 0.002 for both) and HRQoL (difference in SGRQ total score = − 2.0 to − 3.4 U; p < 0.05) and reduced the risk of exacerbations (HR = 0.83; 95% CI = 0.71–0.96) and mortality (HR = 0.66; 95% CI = 0.45–0.96) over the 4-year treatment period [64]. In another analysis of the UPLIFT data, patients taking ICS had significantly higher incidence rates of pneumonia (0.068 vs 0.056; p = 0.012) and COPD exacerbations (0.88 vs 0.62; p < 0.001) than patients not taking ICS [65]. An attenuated rate of pneumonia was observed in the tiotropium subgroup, in general, irrespective of ICS use (pneumonia incidence rate: fluticasone/placebo = 0.081; fluticasone/tiotropium = 0.073; other ICS/placebo = 0.062; other ICS/tiotropium = 0.055; no ICS/placebo = 0.055; and no ICS/tiotropium = 0.056), suggesting the adverse effects associated with the use of ICS might be mitigated with add-on tiotropium.

Efficacy and safety vs other therapies

Tiotropium vs ICS/LABA

Tiotropium 18 μg QD provided a bronchodilatory effect comparable to fluticasone/salmeterol 250/50 μg BID in a 6-week RCT (n = 107) of patients with moderate-to-very severe COPD [66]. In addition, in the Investigating New Standards for Prophylaxis in Reducing Exacerbations (INSPIRE) trial (n = 1323), fluticasone/salmeterol 500/50 μg BID was comparable to tiotropium 18 μg QD in reducing exacerbations (modeled annual exacerbation rate = 1.28 vs 1.32; p = 0.656) in patients with severe and very severe COPD after 2 years of treatment [67]. However, fluticasone/salmeterol was associated with an increased incidence of pneumonia compared with tiotropium (p = 0.008) [67].

Tiotropium vs short-acting muscarinic antagonist/short-acting β2-agonist (SAMA/SABA) combinations

Patients with moderate-to-very severe COPD taking ipratropium/albuterol 36/206 μg QID delivered via a pMDI who switched to tiotropium 18 μg QD had significantly improved lung function compared with those who continued taking ipratropium/albuterol (difference in mean trough FEV1 at 84 days = 0.086 L; p < 0.0001) in a randomized, parallel-group, double-blind, double-dummy study (n = 676) [68]. The incidence of respiratory adverse events was lower in the tiotropium group than in the ipratropium/albuterol group [68]. Additionally, tiotropium significantly reduced the risk of exacerbations (p = 0.0086) and COPD-related referrals/hospitalizations (p = 0.004) compared with ipratropium/salbutamol at the end of treatment in a retrospective, 12-month, follow-up study (n = 4193) using the United Kingdom General Practice Research Database [69].

Tiotropium vs QD LABA

The efficacy of QD indacaterol was compared with that of QD tiotropium in several trials. Indacaterol 150 μg QD and indacaterol 300 μg QD delivered via a DPI (Onbrez® Breezhaler®; Novartis Pharmaceuticals) were at least as effective as tiotropium 18 μg QD in improving lung function and clinical outcomes in patients with moderate-to-severe COPD in short-term trials: INdacaterol & TIotropium: Measuring Efficacy (INTIME; indacaterol 150 or 300 μg; 14 days, n = 169) [70], INdacaterol Towards Establishment of cliNical SuperiorITY study (INTENSITY; indacaterol 150 μg; 12 weeks, n = 1598) [71], and Indacaterol to Help Achieve New COPD treatment Excellence (INHANCE; indacaterol 150 or 300 μg; 26 weeks, n = 1683) [72]. Further, indacaterol 150 μg QD was non-inferior to tiotropium 18 μg QD in improving lung function (week 12; least squares mean difference in trough FEV1 = − 0.011 L; p < 0.0001) in patients with severe COPD and a history of ≥1 exacerbation in the previous year in the 52-week Indacaterol: Providing Opportunity to Re-engage Patients with Life (INVIGORATE) trial (n = 3444) [73]. Tiotropium was superior to indacaterol in reducing exacerbations (week 52; annual rate = 0.73 vs 0.90; p < 0.001). Safety was similar between the treatment groups.

Tiotropium vs other LAMAs

In a 6-week RCT (n = 414), the efficacy of aclidinium 400 μg BID delivered via a DPI (Genuair®/Pressair™; AstraZeneca) was compared with that of placebo and tiotropium 18 μg QD in patients with moderate-to-severe COPD [74]. Both aclidinium and tiotropium compared with placebo significantly improved lung function (differences in change from baseline in FEV1 area under the curve for the 24-h period post-morning dose [AUC0–24] = 0.15 L and 0.14 L, respectively; both p < 0.0001) and COPD symptom scores (p < 0.0001 and p < 0.05, respectively) at the end of the treatment period. Aclidinium, but not tiotropium, reduced the severity of nighttime symptoms vs placebo (p < 0.05). Safety was similar between treatment groups. At week 12 of the glycopyrronium bromide in COPD airways trial 5 (GLOW 5; n = 657), lung function was comparable between glycopyrronium 50 μg QD (Breezhaler®) and tiotropium HandiHaler® 18 μg QD (week 12; least squares mean trough FEV1 = 1.405 L for both) [75]. Improvements in dyspnea, HRQoL, rescue medication use, and rate of COPD exacerbations, as well as safety, were also similar between groups.

Clinical development of tiotropium Respimat®

pMDIs generate aerosols with high velocity, which leads to considerable oropharyngeal drug deposition, and the necessary coordination of inhalation and actuation is difficult for some patients [76]. On the other hand, most DPIs require a strong inspiratory airflow, which also may be difficult for some patients [76].

Soft mist inhaler

The only available SMI, Respimat®, is a pocket-sized, propellant-free device that generates an aerosol from a drug solution effectively and consistently [76]. Respimat®, which was developed to overcome or minimize some of the limitations of other inhalers [77], uses mechanical energy to generate a fine, slow-moving aerosol cloud (mist) from the drug solution, requires minimal coordination of inhalation and actuation, and does not require high inspiratory flow rates [14]. Further, the fine-particle fraction (approximately 75%; particles > 1 to < 5.8 μm) of the mist is nearly twice that in the aerosol cloud emitted from most pMDIs and DPIs [76], and the mist has a lower velocity and a longer duration than the aerosol cloud of pMDIs (mean velocity at a 10-cm distance from the nozzle: SMI = 0.8 m/s; pMDIs = 2.0–8.4 m/s; mean duration: SMI = 1.5 s; pMDIs = 0.15–0.36 s) [20]. These factors contribute to reduced oropharyngeal drug deposition and increased drug deposition in the lungs [1517].

Dose response

Because Respimat® provides high drug lung deposition, lower doses of tiotropium were expected to provide comparable efficacy as the doses used with HandiHaler® [30]. A 3-week, dose-ranging RCT (n = 202) was conducted to compare various tiotropium doses (1.25 μg, 2.5 μg, 5 μg, 10 μg, or 20 μg QD) delivered via Respimat®, placebo delivered via Respimat®, tiotropium 18 μg delivered via HandiHaler®, and placebo delivered via HandiHaler® [30]. Tiotropium Respimat® 5 μg and 20 μg significantly improved lung function compared with placebo Respimat® (difference in trough FEV1 response = 0.17 L [both doses]; p < 0.05 for both) at the end of treatment. Tiotropium Respimat® 10 μg also improved lung function but not significantly (p = 0.06) more than placebo [30]. Safety was similar across treatment groups.

Pivotal studies

In a pre-specified pooled analysis of two 30-week studies (n = 207), tiotropium Respimat® 5 μg and 10 μg QD were superior to placebo in improving lung function (day 29; difference in trough FEV1 response = 0.126 L and 0.127 L, respectively; p < 0.0001 for both) and were non-inferior to tiotropium HandiHaler® 18 μg QD (day 29; difference in trough FEV1 response = 0.029 L and 0.031 L, respectively; p < 0.0001 for both) [78]. In two identical, short-term, randomized, active- and placebo-controlled trials (n = 719), the efficacy and safety of tiotropium Respimat® 5 μg and 10 μg QD were compared with that of ipratropium 36 μg QID delivered via a pMDI [27]. At week 12, both tiotropium doses significantly improved lung function compared with ipratropium (difference in trough FEV1 response = 0.064 L [p = 0.006] and 0.095 L [p < 0.001], respectively; difference in trough FVC response = 0.077 L [p > 0.01] and 0.125 L [p < 0.01], respectively) and compared with placebo (difference in trough FEV1 response = 0.118 L and 0.149 L, respectively [both p < 0.0001]; difference in trough FVC response = 0.132 L [p < 0.01] and 0.180 L [p < 0.0001], respectively) [27]. Both tiotropium doses also significantly reduced rescue medication use compared with placebo (p = 0.0061 and p < 0.0001, respectively); however, only the 10-μg dose was statistically superior to ipratropium (p = 0.04). The long-term efficacy and safety of tiotropium Respimat® 5 μg and 10 μg QD were evaluated in two identical RCTs (n = 1990) [28]. At 1 year of treatment, both tiotropium doses compared with placebo significantly improved lung function (difference in trough FEV1 response = 0.127 L and 0.150 L, respectively; both p < 0.0001) and HRQoL (difference in SGRQ total score = − 3.5 and – 3.8, respectively; both p < 0.0001) and reduced dyspnea (difference in TDI focal score = 1.05 and 1.08, respectively; both p < 0.0001) and mean COPD exacerbation rate/PY (odds ratio [OR] = 0.75 [p < 0.01] and 0.74 [p < 0.001], respectively). Significantly fewer patients experienced ≥1 exacerbation in the tiotropium groups than in the placebo group (difference = − 6.9 and − 7.2%, respectively; both p < 0.01). The incidence of gastrointestinal disorders was greater in the tiotropium groups than in the placebo group (dry mouth: 5 μg = 7.2%, 10 μg = 14.5%, placebo = 2.1%; constipation: 5 μg = 2.1%, 10 μg = 2.2%, placebo = 1.5%). In another 1-year RCT (n = 3991), the efficacy and safety of tiotropium Respimat® 5 μg QD were evaluated [29]. Patients were permitted to use usual therapy (any concurrent COPD medications except inhaled anticholinergics) to reflect closely the place of tiotropium in COPD management. At week 48, tiotropium 5 μg compared with placebo significantly improved lung function (adjusted mean difference in trough FEV1 = 0.102 L; adjusted mean difference in trough FVC = 0.168 L; both p < 0.0001) and improved HRQoL (adjusted mean difference in SGRQ total score = − 2.9 U; p < 0.0001). Further, the proportion of patients having ≥1 exacerbation was lower in the tiotropium group compared with the placebo group (35.3 and 43.1%, respectively; HR = 0.69 [ie, 31% reduction in the risk of exacerbations with tiotropium; p < 0.0001) [29]. Collectively, results of these studies showed that tiotropium Respimat® 5 μg QD was efficacious as maintenance treatment for COPD and for reducing exacerbations, leading to the approval of Spiriva® Respimat® in the EU (2007), US (2014), and other countries [79].

Observational studies

The effectiveness of tiotropium Respimat® 5 μg QD was evaluated in a 6-week open-label observational study (n = 1230) [80]. Tiotropium improved physical function as indicated by a significant increase in the mean Physical Function subdomain (PF-10) score (baseline, 49.0 points; week 6, 62.3 points; difference, 13.4 points [p < 0.001]). Further, the proportion of patients achieving a ≥10-point improvement in PF-10 score was not significantly different between smokers and non-smokers (61.4 and 61.6%, respectively; difference, p = 0.93). Adverse events were reported by 4.0% of patients, the most common being respiratory symptoms and dry mouth.

Landmark safety trial

The TIOtropium Safety and Performance In Respimat® (TIOSPIR®) trial (n = 17,315) was conducted to confirm the safety and efficacy of tiotropium Respimat® in a large population of patients with COPD [81]. By having an adequate patient population size (> 17,000 patients at > 1200 investigator sites in 50 countries), broad inclusion criteria to closely reflect real-world patients with COPD, and sufficient treatment duration, analyses of all-cause mortality and time to first COPD exacerbation were possible. During a 2.3-year mean follow-up period, tiotropium Respimat® 5 μg and 2.5 μg QD were non-inferior to tiotropium HandiHaler® 18 μg QD in the risk of death (both p < 0.05) and not superior in the risk of exacerbation (p = 0.42 and p = 0.56, respectively) [81]. In patients with a history of cardiac arrhythmia, tiotropium Respimat® 2.5 μg and tiotropium HandiHaler®18 μg had a similar impact on survival as measured by all-cause mortality. In addition to these analyses, various post hoc analyses were conducted to assess the impact of geographical variations [82] on COPD outcomes, to evaluate spirometry outcomes [83], and to determine risk factors for exacerbations [84]. Tiotropium Respimat® 5 μg was associated with a similar improvement in trough FVC and a comparable rate of decline in FEV1 as tiotropium HandiHaler® 18 μg in the spirometry outcomes analysis [83]. In a multivariate analysis, baseline pulmonary maintenance medication was predictive of, and ICS use was associated with, increased exacerbation risk [84].
TIOSPIR® was conducted because a numerical, but not statistically significant, imbalance in the number of deaths had been noted in meta-analyses of Respimat® trials, between tiotropium Respimat® and placebo Respimat® or tiotropium HandiHaler®, particularly for patients with known cardiac disorders [8587]. Valid concerns were quickly raised, however, about the meta-analyses’ methodologies and conclusions, particularly the analysis conducted by Singh et al. [85], which included five of the aforementioned RCTs [2729, 8890]. For example, in that analysis, conclusions were based on three 1-year studies that had mortality imbalances (and not based on two 12-week studies that showed no imbalance) [88]. Additionally, in one of the studies with mortality imbalances, the placebo arm had an unusually low (0.77%) mortality rate compared with that observed in other studies (1.5–2.5% or higher) [88]. Further, fatal cases were incorrectly assigned to treatment groups [88, 91], and a 6-month trial of > 850 patients [92], which had only two deaths in the tiotropium Respimat® arm and five deaths in the placebo Respimat® arm, was not included [88, 91]. Moreover, two doses of tiotropium—the marketed 5-μg dose and a non-approved, unmarketed 10-μg dose—were included in the primary analysis [91, 93]. These and other factors provide a plausible explanation for the imbalances observed in the aforementioned meta-analyses. In TIOSPIR®, a similar impact on survival was observed between tiotropium Respimat® 2.5 μg and tiotropium HandiHaler® 18 μg, which expands upon findings from UPLIFT and reinforces the safety and efficacy of tiotropium in patients with COPD, regardless of cardiac arrhythmia history [26, 29, 81].
Lastly, results of an analysis of the tiotropium COPD clinical program (Table 2) showed that the characteristics of patients included in the RCTs were representative of “real-life” patient populations, thus demonstrating external validation of the results [94]. Further, tiotropium Respimat® 2.5 μg or 5 μg had similar exacerbation efficacy and safety to that of tiotropium HandiHaler® 18 μg [81].
Table 2
Tiotropium – summary table of evidence
Study
Patients (N)
Treatment arms
Primary endpoint results*
Proportion of patients with adverse events
Conclusion of the study
Casaburi 2000 [21]
470
• Tiotropium 18 μg QD
• Placebo
Trough FEV1 response:
0.11 L vs − 0.04 L (p < 0.001)
Overall adverse events:
61.6% vs 66.5%
Dry mouth:
9.3% vs 1.6% (p < 0.05)
Tiotropium was safe and effective
Casaburi 2002 [22]
921
• Tiotropium 18 μg QD
• Placebo
Trough FEV1 response:
0.11 L to 0.13 L (tiotropium; p < 0.01 vs placebo)
Overall adverse events:
90.0% vs 91.1%
Dry mouth:
16.0% vs 2.7% (p < 0.05)
Tiotropium significantly improved lung function and HRQoL and reduced dyspnea, COPD exacerbations, and hospitalizations
Vincken 2002 [34]
535
• Tiotropium 18 μg QD
• Ipratropium 40 μg QID
Trough FEV1 response:
0.12 L vs − 0.03 L (p < 0.001)
Adverse events leading to discontinuation:
10.1% vs 12.8%
Dry mouth:
12.1% vs 6.1% (p = 0.03)
Tiotropium significantly improved lung function and HRQoL and reduced dyspnea and COPD exacerbations compared with ipratropium
Donohue 2002 [35]
623**
• Tiotropium 18 μg QD
• Salmeterol 50 μg BID
Trough FEV1 response:
0.14 L vs 0.09 L (p < 0.01)
Dry mouth:
10% vs NA
Tiotropium significantly improved lung function and reduced dyspnea compared with salmeterol
Brusasco 2003 [36]
1207
• Tiotropium 18 μg QD
• Salmeterol 50 μg BID
• Placebo
COPD exacerbation rate: 1.07 vs 1.23 vs 1.49
(p < 0.05 for tiotropium vs placebo)
Dry mouth:
8.2% vs 1.7% vs 2.3%
Tiotropium significantly improved lung function compared with salmeterol; improved HRQoL and reduced dyspnea and COPD exacerbations compared with placebo
O’Donnell 2004 [45]
187
• Tiotropium 18 μg QD
• Placebo
Difference in endurance time between tiotropium and placebo:
105 s (p = 0.0098)
Overall adverse events:
36.7% vs 41.0%
Tiotropium significantly reduced lung hyperinflation at rest and exercise and improved exertional dyspnea and endurance time
Niewoehner 2005 [37]
1829
• Tiotropium 18 μg QD
• Placebo
Percentage of patients with ≥1 exacerbation:
27.9% vs 32.3% (p = 0.037)
Percentage of patients with COPD-related hospitalization:
7.0% vs 9.5% (p = 0.056)
Serious adverse events:
18% vs 17%
Tiotropium reduced COPD exacerbations, COPD-related hospitalization, and healthcare utilization compared with placebo
Dusser 2006 [24]
1010
• Tiotropium 18 μg QD
• Placebo
Percentage of patients with ≥1 exacerbation:
49.9% vs 60.3% (p < 0.01)
Overall adverse events:
46.4% vs 45.1%
Dry mouth:
4.0% vs 1.4%
Tiotropium significantly improved lung function and reduced COPD exacerbations and COPD-associated health resource use compared with placebo
Verkindre 2006 [32]
100
• Tiotropium 18 μg QD
• Placebo
Trough FVC:
Difference: 0.20 L (p < 0.05)
Adverse events leading to discontinuation:
2% vs 11%
Dry mouth:
2% vs 0%
Tiotropium significantly improved FVC, lung hyperinflation, walking distance, and HRQoL
Bateman 2008 [66]
107
• Tiotropium 18 μg QD
• Fluticasone/salmeterol 250/50 μg BID
FEV1 AUC0–12h:
1.55 L vs 1.57 L (p = 0.63)
Overall adverse events:
41.1% vs 43.1%
Dry mouth:
3.6% vs 3.9%
Tiotropium improved lung function similar to fluticasone/salmeterol combination
Tashkin 2008 [26]
5993
• Tiotropium 18 μg QD
• Placebo
Rate of decline in FEV1 before bronchodilation:
0.030 L vs 0.030 L (p = 0.95)
Rate of decline in FEV1 after bronchodilation:
0.040 L vs 0.042 L (p = 0.21)
Overall adverse events:
92.6% vs 92.3%
Tiotropium significantly improved lung function, improved HRQoL, reduced exacerbations, but did not significantly reduce rate of decline in FEV1 compared with placebo
Tonnel 2008 [23]
554
• Tiotropium 18 μg QD
• Placebo
Proportion of patients with improvement in HRQoLa:
59.1% vs 48.2% (p = 0.029)
Patients with ≥1 adverse event:
60.9% vs 67.0%
Dry mouth:
1.1% vs 0.7%
Tiotropium significantly improved lung function, improved HRQoL, and reduced exacerbations
Voshaar 2008 [27]
719
• Tiotropium 5 μg QD
• Tiotropium 10 μg QD
• Ipratropium 36 μg QID vs placebo
Trough FEV1 response treatment differences:
tiotropium 5 μg − placebo:
0.118 L (p < 0.0001)
tiotropium 10 μg − placebo: 0.149 L (p < 0.0001)
tiotropium 5 μg − ipratropium:
0.064 L (p = 0.006)
tiotropium 10 μg − ipratropium: 0.095 L (p < 0.0001)
Overall adverse events:
52.8% vs 60.0% vs 59.6% vs 59.1%
Dry mouth:
8.3% vs 10.0% vs 3.9% vs 2.2%
Tiotropium (via Respimat®) significantly improved lung function compared with ipratropium (pMDI) and placebo
Wedzicha 2008 [67]
1323
• Tiotropium 18 μg QD
• Fluticasone/salmeterol 500/50 μg BID
Modeled annual rate of exacerbations:
1.32 vs 1.28 (p = 0.656)
Overall adverse events: 62% vs 66%
Tiotropium was similar to fluticasone/salmeterol in exacerbation efficacy
Bateman 2010 [28]
1990
• Tiotropium 5 μg QD
• Tiotropium 10 μg QD
• Placebo
Trough FEV1 response:
tiotropium 5 μg vs placebo:
0.127 L (p < 0.0001)
tiotropium 10 μg vs placebo: 0.150 L (p < 0.0001)
Overall adverse events:
75.4% vs 78.7% vs 76.9%
Tiotropium (via Respimat®) significantly improved lung function and HRQoL and reduced dyspnea and exacerbations compared with placebo
Bateman 2010 [29]
3991
• Tiotropium 5 μg QD
• Placebo
Trough FEV1 response:
0.119 L vs 0.018 L (p < 0.0001)
Time to first exacerbation:
169 days vs 119 days (p < 0.0001)
Overall adverse events:
70.1% vs 69.3%;
Dry mouth:
3.1% vs 1.4%
Tiotropium (via Respimat®) significantly improved lung function and HRQoL and reduced exacerbations compared with placebo
Vogelmeier 2011 [39]
7376
• Tiotropium 18 μg QD
• Salmeterol 50 μg BID
Time to first exacerbation:
187 days vs 145 days (p < 0.001)
Serious adverse events:
14.7% vs 16.5%
Tiotropium significantly reduced exacerbations compared with salmeterol
Wise 2013 [81]
17,135
• Tiotropium 2.5 μg QD
• Tiotropium 5 μg QD
• Tiotropium 18 μg QD
Deaths:
7.7% vs 7.4% vs 7.7%
Proportion of patients with exacerbations:
49.4% vs 47.9% vs 48.9%
Serious adverse events:
33.8% vs 32.4% vs 32.4%
Tiotropium 2.5 μg or 5 μg (via Respimat®) was similar to tiotropium 18 μg (via HandiHaler®) in safety and exacerbation efficacy
AUC0–12h area under the curve from 0 to 12 h post-dose, BID twice a day, COPD chronic obstructive pulmonary disease, DPI dry-powder inhaler, FEV1 forced expiratory volume in 1 s, FVC forced vital capacity, HRQoL health-related quality of life, NA not available, pMDI pressurized metered-dose inhaler, QD once daily, QID four times a day, SGRQ St. George’s Respiratory Questionnaire
aReduction of at least four units in the SGRQ score
*For studies in which the primary endpoint was not specified, results of lung function are included.**Includes the total number of patients in the tiotropium, salmeterol, and placebo groups
Tiotropium 18 μg delivered via HandiHaler®; tiotropium 2.5 μg, 5 μg, and 10 μg delivered via Respimat®; other drugs delivered via a pMDI or a DPI

Clinical development of tiotropium/olodaterol Respimat®

As mentioned, the clinical development of tiotropium culminated with evidence supporting its use in a fixed-dose combination with olodaterol. The ToVITO® program, which involved more than 16,000 patients and is outside the scope of this review, was designed to evaluate the effect of tiotropium/olodaterol (2.5/5 μg and/or 5/5 μg) Respimat® on various efficacy parameters, as well as its safety. Results of these trials, and others, demonstrated that tiotropium/olodaterol provided a significant, incremental benefit over tiotropium monotherapy in alleviating COPD symptoms such as breathlessness, improving lung function and QoL, and reducing moderate and severe exacerbations (Table 3) [95102]. In the DYNAGITO® trial, tiotropium/olodaterol reduced the rate of moderate and severe exacerbations by 7% compared with tiotropium alone, which did not meet the predefined significance level of 0.01. However, in a post hoc analysis using multiple covariate models similar to those used in the SPARK and FLAME trials, an 11% reduction in moderate-to-severe exacerbations was observed [102, 103]. Results across trials included in the ToVITO® program showed no significant differences in the frequency of general and serious adverse events between tiotropium/olodaterol Respimat® and mono components [104].
Table 3
Tiotropium/olodaterol combination – summary table of evidence
Study
Patients (N)
Treatment arms
Primary endpoint results
Proportion of patients with adverse events
Conclusion of the study
Buhl 2015 [95]
5162
• Tiotropium+olodaterol 2.5/5 μg QD
• Tiotropium+olodaterol 5/5 μg QD
• Tiotropium 2.5 μg QD
• Tiotropium 5 μg QD
• Olodaterol 5 μg QD
• FEV1 AUC0–3 response:
o Tiotropium+olodaterol 2.5/5 μg vs olodaterol 5 μg, 0.115 L; vs tiotropium 2.5 μg, 0.111 L; and vs tiotropium 5 μg, 0.097 L (p < 0.0001 for all comparisons)
o Tiotropium+olodaterol 5/5 μg vs olodaterol 5 μg, 0.128 L and vs tiotropium 5 μg, 0.110 L (p < 0.0001 for both)
• Trough FEV1 response:
o Tiotropium+olodaterol 2.5/5 μg: vs olodaterol 5 μg, 0.062 L; vs tiotropium 2.5 μg, 0.045 L; vs tiotropium 5 μg, 0.038 L (p < 0.0001 for all comparisons)
o Tiotropium+olodaterol 5/5 μg: vs olodaterol 5 μg, 0.085 L; vs tiotropium 5 μg, 0.060 L (p < 0.0001 for both comparisons)
• SGRQ total score:
o Tiotropium+olodaterol 5/5 μg: vs olodaterol 5 μg, − 1.693 (p = 0.0022); vs tiotropium 5 μg, − 1.233 (p = 0.0252)
o Tiotropium+olodaterol 2.5/5 μg vs individual components was not significant for all comparisons
74.7% vs 74.0% vs 73.4% vs 73.3% vs 76.6%
Tiotropium+olodaterol improved lung function and HRQoL compared with monocomponents
Beeh 2015 [96]
259
• Tiotropium+olodaterol 2.5/5 μg QD
• Tiotropium+olodaterol 5/5 μg QD
• Tiotropium 2.5 μg QD
• Tiotropium 5 μg QD
• Olodaterol 5 μg QD
• Placebo
• FEV1 AUC0–24 response:
o Tiotropium+olodaterol 2.5/5 μg: vs olodaterol 5 μg, 0.111 L; vs tiotropium 2.5 μg, 0.124 L; vs tiotropium 5 μg, 0.107 L; vs placebo, 0.277 L (p < 0.001 for all comparisons)
o Tiotropium+olodaterol 5/5 μg: vs olodaterol 5 μg, 0.115 L; vs tiotropium 2.5 μg, 0.127 L; vs tiotropium 5 μg, 0.110 L; vs placebo, 0.280 L (p < 0.0001 for all comparisons)
36.0% vs 37.4% vs 39.4% vs 44.2% vs 37.7% vs 46.4%
Tiotropium+olodaterol improved lung function over 24 h compared with monocomponents
O’Donnell 2017 [97]
586
• Tiotropium+olodaterol 2.5/5 μg QD
• Tiotropium+olodaterol 5/5 μg QD
• Tiotropium 5 μg QD
• Olodaterol 5 μg QD
• Placebo
• Inspiratory capacity:
o Tiotropium+olodaterol 2.5/5 μg: vs olodaterol 5 μg, 0.090 L; vs tiotropium 5 μg, 0.092 L; vs placebo, 0.245 L (p < 0.0001 for all comparisons)
o Tiotropium+olodaterol 5/5 μg: vs olodaterol 5 μg, 0.099 L; vs tiotropium 5 μg, 0.101 L; vs placebo, 0.254 L (p < 0.0001 for all comparisons)
• Exercise endurance time during constant work-rate cycle ergometry (improvement):
o Tiotropium+olodaterol 2.5/5 μg: vs olodaterol 5 μg, 7.3% (p < 0.01); vs tiotropium 5 μg, 3.5%; vs placebo, 19.2% (p < 0.0001)
o Tiotropium+olodaterol 5/5 μg: vs olodaterol 5 μg, 5.6% (p < 0.05); vs tiotropium 5 μg, 1.9%; vs placebo, 17.3% (p < 0.0001)
36.3% vs 40.0% vs 38.3% vs 40.2% vs 40.8%
Tiotropium+olodaterol improved lung hyperinflation and exercise tolerance compared with monotherapies
Maltais 2018 [98]
404
• Tiotropium+olodaterol 2.5/5 μg QD
• Tiotropium+olodaterol 5/5 μg QD
• Placebo
• Endurance time during constant work-rate cycle ergometry:
o Tiotropium+olodaterol 5/5 μg vs placebo, 14% (p = 0.02)
o Tiotropium+olodaterol 2.5/5 μg vs placebo, 9% (p = 0.14)
54.9% vs 43.9% vs 50.8%
Tiotropium+olodaterol improved endurance time compared with placebo during cycle ergometry
Singh 2015 [99]
1621
• Tiotropium+olodaterol 2.5/5 μg QD
• Tiotropium+olodaterol 5/5 μg QD
• Tiotropium 5 μg QD
• Placebo
• SGRQ total score (difference):
o Tiotropium+olodaterol 5/5 μg: vs tiotropium 5 μg, − 2.10 (p < 0.01); vs placebo, − 4.67 (p < 0.0001)
o Tiotropium+olodaterol 2.5/5 μg: vs tiotropium 5 μg, − 1.27; vs placebo, − 3.85 (p < 0.0001)
• FEV1 AUC0–3 response:
o Both tiotropium+olodaterol 2.5/5 μg and 5/5 μg significantly improved (p < 0.0001) FEV1 AUC0–3 response compared with placebo and tiotropium 5 μg
OTEMTO 1: 42.6% vs 44.8% vs 44.3% vs 51.5%
OTEMTO 2: 45.5% vs 43.1% vs 45.8% vs 46.0%
Tiotropium+olodaterol improved lung function and QoL compared with placebo and tiotropium
Beeh 2016 [100]
229
• Tiotropium+olodaterol 2.5/5 μg QD
• Tiotropium+olodaterol 5/5 μg QD
• Salmeterol/fluticasone 50/500 μg BID
• Salmeterol/fluticasone 50/250 μg BID
• FEV1 AUC0–12 response:
o 0.295 L vs 0.317 L vs 0.188 L vs 0.192 L (p < 0.0001 for comparisons of tiotropium+olodaterol vs salmeterol/fluticasone)
34.4% vs 33.9% vs 37.0% vs 29.7%
Tiotropium+olodaterol QD provided superior improvement in lung function compared with salmeterol/fluticasone BID
Troosters 2018 [101]
303
• Tiotropium+olodaterol 5/5 μg QD
• Tiotropium+olodaterol 5/5 μg QD plus 8 weeks of ExT
• Tiotropium 5 μg
• Placebo
• Exercise endurance time by shuttle walk test (increase):
o Tiotropium+olodaterol 5/5 μg QD vs placebo, 29.2% (p = 0.0109)
o Tiotropium+olodaterol 5/5 μg QD plus 8 weeks of ExT vs placebo, 45.8% (p = 0.0002)
o Tiotropium 5 μg vs placebo, 4.1% (p = 0.6895)
57.9% vs 64.5% vs 67.1% vs 61.3%
In patients taking part in a self-management behavior-modification program, tiotropium+olodaterol improved exercise endurance time compared with placebo
Calverley 2018 [102]
7880
• Tiotropium+olodaterol 5/5 μg QD
• Tiotropium 5 μg QD
• Rate of moderate and severe COPD exacerbations:
o 0.90 vs 0.97 (rate ratio, 0.93; p = 0.0498)
74% vs 75%
Tiotropium+olodaterol reduced exacerbation rate compared with tiotropium, but not to a significant extent
AUC0–24 area under the curve from 0 to 24 h post-dose, AUC0–12 area under the curve from 0 to 12 h post-dose, AUC0–3 area under the curve from 0 to 3 h post-dose, BID twice a day, COPD chronic obstructive pulmonary disease, ExT exercise training, FEV1 forced expiratory volume in 1 s, HRQoL health-related quality of life, QD once daily, QoL quality of life, SGRQ St. George’s Respiratory Questionnaire
Tiotropium will continue to be the first-line treatment for newly diagnosed COPD, in particular in mild to moderate cases and infrequent exacerbators. We hope that early diagnosis will be more frequent in the future and these newly diagnosed cases, probably with milder disease, might be optimal candidates for treatment with tiotropium/olodaterol if new studies demonstrate that optimal early bronchodilation results in improved long-term outcomes.

Conclusions

Treatment goals for COPD include reduction in symptoms and future risk of exacerbations [1, 2, 105]. Per GOLD recommendations, LAMA monotherapy is recommended as initial treatment for GOLD groups B, C, and D. LABA/LAMA combination therapy may be considered in group B patients with severe breathlessness and is an initial treatment option for group D patients who are highly symptomatic [2]. No other LAMA has been proven superior to tiotropium, making it an optimal LAMA as monotherapy and as the backbone LAMA in LABA/LAMA combination therapy. In most comparative studies of tiotropium and placebo, ipratropium, or salmeterol, tiotropium provided significant beneficial effects on lung function, including improvements in FEV1 and FVC [2136]. Tiotropium also significantly improved exacerbation-related outcomes such as reduction in the number of exacerbations/PY, reduction in the proportion of patients experiencing ≥1 exacerbation and exacerbation-related hospitalizations, increase in the time to first exacerbation, and reduction in HCRU compared with placebo and salmeterol [2224, 26, 28, 34, 3639]. In addition, tiotropium treatment improved HRQoL and significantly reduced dyspnea, need for “as-needed” SABA use, and lung hyperinflation, resulting in improvement in exertional dyspnea and exercise endurance [2123, 26, 28, 29, 32, 3436, 45, 46]. The long-term efficacy of tiotropium was demonstrated in the UPLIFT trial. Finally, tiotropium was comparable to ICS/LABA (fluticasone/salmeterol) in improving lung function and reducing exacerbations [66, 67] and had a greater effect on exacerbation rates than LABAs [39, 73]. Because long-term use of ICS is associated with systemic and local side effects, tiotropium is a suitable alternative to ICS/LABA combinations. Overall, tiotropium is safe and efficacious as a long-term, QD LAMA for the maintenance treatment of COPD and for reducing COPD exacerbations or for the maintenance treatment of COPD as part of long-term, QD, fixed-dose LAMA/LABA (tiotropium/olodaterol) [6, 7, 9].

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12931-020-01407-y.

Acknowledgements

The authors meet the criteria for authorship as recommended by the International Committee of Medical Journal Editors (ICMJE). The authors received no direct compensation related to the development of the manuscript. Writing, editorial support, and formatting assistance was provided by Sudha Korwar, PhD, Suchita Nath-Sain, PhD, and Maribeth Bogush, PhD, of Cactus Life Sciences (part of Cactus Communications), which was contracted and funded by Boehringer Ingelheim Pharmaceuticals, Inc. (BIPI). BIPI was given the opportunity to review the manuscript for medical and scientific accuracy as well as intellectual property considerations.
Not applicable.
Not applicable.

Competing interests

AA has received consultant fees from Boehringer Ingelheim, AstraZeneca, GlaxoSmithKline, and Novartis.
MM has received speaker fees from AstraZeneca, Boehringer Ingelheim, Chiesi, Cipla, Menarini, Rovi, Bial, Sandoz, Zambon, CSL Behring, Grifols, and Novartis; consulting fees from AstraZeneca, Boehringer Ingelheim, Chiesi, GlaxoSmithKline, Bial, Gebro Pharma, Kamada, CSL Behring, Laboratorios Esteve, Ferrer, Mereo Biopharma, Verona Pharma, TEVA, pH Pharma, Novartis, Sanofi, and Grifols; and research grants from GlaxoSmithKline and Grifols.
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.
Literatur
1.
Zurück zum Zitat Vogelmeier CF, Criner GJ, Martínez FJ, Anzueto A, Barnes PJ, Bourbeau J, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease 2017 report: GOLD executive summary. Arch Bronconeumol. 2017;53:128–49.PubMed Vogelmeier CF, Criner GJ, Martínez FJ, Anzueto A, Barnes PJ, Bourbeau J, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease 2017 report: GOLD executive summary. Arch Bronconeumol. 2017;53:128–49.PubMed
2.
Zurück zum Zitat Global Initiative for Chronic Obstructive Lung Disease (GOLD). Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease (2020 report). https://goldcopd.org/. Accessed 3 Feb 2020. Global Initiative for Chronic Obstructive Lung Disease (GOLD). Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease (2020 report). https://​goldcopd.​org/​. Accessed 3 Feb 2020.
3.
Zurück zum Zitat Meurs H, Oenema TA, Kistemaker LE, Gosens R. A new perspective on muscarinic receptor antagonism in obstructive airways diseases. Curr Opin Pharmacol. 2013;13:316–23.PubMed Meurs H, Oenema TA, Kistemaker LE, Gosens R. A new perspective on muscarinic receptor antagonism in obstructive airways diseases. Curr Opin Pharmacol. 2013;13:316–23.PubMed
4.
Zurück zum Zitat Disse B, Speck GA, Rominger KL, Witek TJ Jr, Hammer R. Tiotropium (Spiriva): mechanistical considerations and clinical profile in obstructive lung disease. Life Sci. 1999;64:457–64.PubMed Disse B, Speck GA, Rominger KL, Witek TJ Jr, Hammer R. Tiotropium (Spiriva): mechanistical considerations and clinical profile in obstructive lung disease. Life Sci. 1999;64:457–64.PubMed
5.
Zurück zum Zitat Panning CA, DeBisschop M. Tiotropium: an inhaled, long-acting anticholinergic drug for chronic obstructive pulmonary disease. Pharmacotherapy. 2003;23:183–9.PubMed Panning CA, DeBisschop M. Tiotropium: an inhaled, long-acting anticholinergic drug for chronic obstructive pulmonary disease. Pharmacotherapy. 2003;23:183–9.PubMed
8.
Zurück zum Zitat Malerba M, Foci V, Patrucco F, Pochetti P, Nardin M, Pelaia C, et al. Single Inhaler LABA/LAMA for COPD. Front Pharmacol. 2019;10:390.PubMedPubMedCentral Malerba M, Foci V, Patrucco F, Pochetti P, Nardin M, Pelaia C, et al. Single Inhaler LABA/LAMA for COPD. Front Pharmacol. 2019;10:390.PubMedPubMedCentral
11.
Zurück zum Zitat Ibrahim M, Verma R, Garcia-Contreras L. Inhalation drug delivery devices: technology update. Med Devices (Auckl). 2015;8:131–9. Ibrahim M, Verma R, Garcia-Contreras L. Inhalation drug delivery devices: technology update. Med Devices (Auckl). 2015;8:131–9.
12.
Zurück zum Zitat Mahler DA. Peak inspiratory flow rate as a criterion for dry powder inhaler use in chronic obstructive pulmonary disease. Ann Am Thorac Soc. 2017;14:1103–7.PubMed Mahler DA. Peak inspiratory flow rate as a criterion for dry powder inhaler use in chronic obstructive pulmonary disease. Ann Am Thorac Soc. 2017;14:1103–7.PubMed
13.
Zurück zum Zitat Vincken W, Levy ML, Scullion J, Usmani OS, Dekhuijzen PNR, Corrigan CJ. Spacer devices for inhaled therapy: why use them, and how? ERJ Open Res. 2018;4:00065–2018.PubMedPubMedCentral Vincken W, Levy ML, Scullion J, Usmani OS, Dekhuijzen PNR, Corrigan CJ. Spacer devices for inhaled therapy: why use them, and how? ERJ Open Res. 2018;4:00065–2018.PubMedPubMedCentral
14.
15.
Zurück zum Zitat Newman SP, Brown J, Steed KP, Reader SJ, Kladders H. Lung deposition of fenoterol and flunisolide delivered using a novel device for inhaled medicines: comparison of RESPIMAT with conventional metered-dose inhalers with and without spacer devices. Chest. 1998;113:957–63.PubMed Newman SP, Brown J, Steed KP, Reader SJ, Kladders H. Lung deposition of fenoterol and flunisolide delivered using a novel device for inhaled medicines: comparison of RESPIMAT with conventional metered-dose inhalers with and without spacer devices. Chest. 1998;113:957–63.PubMed
16.
Zurück zum Zitat Brand P, Hederer B, Austen G, Dewberry H, Meyer T. Higher lung deposition with Respimat soft mist inhaler than HFA-MDI in COPD patients with poor technique. Int J Chron Obstruct Pulmon Dis. 2008;3:763–70.PubMedPubMedCentral Brand P, Hederer B, Austen G, Dewberry H, Meyer T. Higher lung deposition with Respimat soft mist inhaler than HFA-MDI in COPD patients with poor technique. Int J Chron Obstruct Pulmon Dis. 2008;3:763–70.PubMedPubMedCentral
17.
Zurück zum Zitat Pitcairn G, Reader S, Pavia D, Newman S. Deposition of corticosteroid aerosol in the human lung by Respimat soft mist inhaler compared to deposition by metered dose inhaler or by Turbuhaler dry powder inhaler. J Aerosol Med. 2005;18:264–72.PubMed Pitcairn G, Reader S, Pavia D, Newman S. Deposition of corticosteroid aerosol in the human lung by Respimat soft mist inhaler compared to deposition by metered dose inhaler or by Turbuhaler dry powder inhaler. J Aerosol Med. 2005;18:264–72.PubMed
18.
Zurück zum Zitat Zierenberg B. Optimizing the in vitro performance of Respimat. J Aerosol Med. 1999;12(Suppl 1):S19–24.PubMed Zierenberg B. Optimizing the in vitro performance of Respimat. J Aerosol Med. 1999;12(Suppl 1):S19–24.PubMed
19.
Zurück zum Zitat Dalby R, Spallek M, Voshaar T. A review of the development of Respimat soft mist inhaler. Int J Pharm. 2004;283:1–9.PubMed Dalby R, Spallek M, Voshaar T. A review of the development of Respimat soft mist inhaler. Int J Pharm. 2004;283:1–9.PubMed
20.
Zurück zum Zitat Hochrainer D, Hölz H, Kreher C, Scaffidi L, Spallek M, Wachtel H. Comparison of the aerosol velocity and spray duration of Respimat soft mist inhaler and pressurized metered dose inhalers. J Aerosol Med. 2005;18:273–82.PubMed Hochrainer D, Hölz H, Kreher C, Scaffidi L, Spallek M, Wachtel H. Comparison of the aerosol velocity and spray duration of Respimat soft mist inhaler and pressurized metered dose inhalers. J Aerosol Med. 2005;18:273–82.PubMed
21.
Zurück zum Zitat Casaburi R, Briggs D Jr, Donohue JF, Serby CW, Menjoge SS, Witek T Jr. The spirometric efficacy of once-daily dosing with tiotropium in stable COPD: a 13-week multicenter trial. The US Tiotropium Study Group. Chest. 2000;118:1294–302. Casaburi R, Briggs D Jr, Donohue JF, Serby CW, Menjoge SS, Witek T Jr. The spirometric efficacy of once-daily dosing with tiotropium in stable COPD: a 13-week multicenter trial. The US Tiotropium Study Group. Chest. 2000;118:1294–302.
22.
Zurück zum Zitat Casaburi R, Mahler DA, Jones PW, Wanner A, San PG, ZuWallack RL, et al. A long-term evaluation of once-daily inhaled tiotropium in chronic obstructive pulmonary disease. Eur Respir J. 2002;19:217–24.PubMed Casaburi R, Mahler DA, Jones PW, Wanner A, San PG, ZuWallack RL, et al. A long-term evaluation of once-daily inhaled tiotropium in chronic obstructive pulmonary disease. Eur Respir J. 2002;19:217–24.PubMed
23.
Zurück zum Zitat Tonnel AB, Perez T, Grosbois JM, Verkindre C, Bravo ML, Brun M, et al. Effect of tiotropium on health-related quality of life as a primary efficacy endpoint in COPD. Int J Chron Obstruct Pulmon Dis. 2008;3:301–10.PubMedPubMedCentral Tonnel AB, Perez T, Grosbois JM, Verkindre C, Bravo ML, Brun M, et al. Effect of tiotropium on health-related quality of life as a primary efficacy endpoint in COPD. Int J Chron Obstruct Pulmon Dis. 2008;3:301–10.PubMedPubMedCentral
24.
Zurück zum Zitat Dusser D, Bravo ML, Iacono P. The effect of tiotropium on exacerbations and airflow in patients with COPD. Eur Respir J. 2006;27:547–55.PubMed Dusser D, Bravo ML, Iacono P. The effect of tiotropium on exacerbations and airflow in patients with COPD. Eur Respir J. 2006;27:547–55.PubMed
25.
Zurück zum Zitat Zhou Y, Zhong NS, Li X, Chen S, Zheng J, Zhao D, et al. Tiotropium in early-stage chronic obstructive pulmonary disease. N Engl J Med. 2017;377:923–35.PubMed Zhou Y, Zhong NS, Li X, Chen S, Zheng J, Zhao D, et al. Tiotropium in early-stage chronic obstructive pulmonary disease. N Engl J Med. 2017;377:923–35.PubMed
26.
Zurück zum Zitat Tashkin DP, Celli B, Senn S, Burkhart D, Kesten S, Menjoge S, et al. A 4-year trial of tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2008;359:1543–54.PubMed Tashkin DP, Celli B, Senn S, Burkhart D, Kesten S, Menjoge S, et al. A 4-year trial of tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2008;359:1543–54.PubMed
27.
Zurück zum Zitat Voshaar T, Lapidus R, Maleki-Yazdi R, Timmer W, Rubin E, Lowe L, et al. A randomized study of tiotropium Respimat soft mist inhaler vs. ipratropium pMDI in COPD. Respir Med. 2008;102:32–41.PubMed Voshaar T, Lapidus R, Maleki-Yazdi R, Timmer W, Rubin E, Lowe L, et al. A randomized study of tiotropium Respimat soft mist inhaler vs. ipratropium pMDI in COPD. Respir Med. 2008;102:32–41.PubMed
28.
Zurück zum Zitat Bateman E, Singh D, Smith D, Disse B, Towse L, Massey D, et al. Efficacy and safety of tiotropium Respimat SMI in COPD in two 1-year randomized studies. Int J Chron Obstruct Pulmon Dis. 2010;5:197–208.PubMedPubMedCentral Bateman E, Singh D, Smith D, Disse B, Towse L, Massey D, et al. Efficacy and safety of tiotropium Respimat SMI in COPD in two 1-year randomized studies. Int J Chron Obstruct Pulmon Dis. 2010;5:197–208.PubMedPubMedCentral
29.
Zurück zum Zitat Bateman ED, Tashkin D, Siafakas N, Dahl R, Towse L, Massey D, et al. A one-year trial of tiotropium Respimat plus usual therapy in COPD patients. Respir Med. 2010;104:1460–72.PubMed Bateman ED, Tashkin D, Siafakas N, Dahl R, Towse L, Massey D, et al. A one-year trial of tiotropium Respimat plus usual therapy in COPD patients. Respir Med. 2010;104:1460–72.PubMed
30.
Zurück zum Zitat Caillaud D, Le Merre C, Martinat Y, Aguilaniu B, Pavia D. A dose-ranging study of tiotropium delivered via Respimat soft mist inhaler or HandiHaler in COPD patients. Int J Chron Obstruct Pulmon Dis. 2007;2:559–65.PubMedPubMedCentral Caillaud D, Le Merre C, Martinat Y, Aguilaniu B, Pavia D. A dose-ranging study of tiotropium delivered via Respimat soft mist inhaler or HandiHaler in COPD patients. Int J Chron Obstruct Pulmon Dis. 2007;2:559–65.PubMedPubMedCentral
31.
Zurück zum Zitat Littner MR, Ilowite JS, Tashkin DP, Friedman M, Serby CW, Menjoge SS, et al. Long-acting bronchodilation with once-daily dosing of tiotropium (Spiriva) in stable chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2000;161:1136–42.PubMed Littner MR, Ilowite JS, Tashkin DP, Friedman M, Serby CW, Menjoge SS, et al. Long-acting bronchodilation with once-daily dosing of tiotropium (Spiriva) in stable chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2000;161:1136–42.PubMed
32.
Zurück zum Zitat Verkindre C, Bart F, Aguilaniu B, Fortin F, Guérin JC, Le Merre C, et al. The effect of tiotropium on hyperinflation and exercise capacity in chronic obstructive pulmonary disease. Respiration. 2006;73:420–7.PubMed Verkindre C, Bart F, Aguilaniu B, Fortin F, Guérin JC, Le Merre C, et al. The effect of tiotropium on hyperinflation and exercise capacity in chronic obstructive pulmonary disease. Respiration. 2006;73:420–7.PubMed
33.
Zurück zum Zitat Chan CK, Maltais F, Sigouin C, Haddon JM, Ford GT. A randomized controlled trial to assess the efficacy of tiotropium in Canadian patients with chronic obstructive pulmonary disease. Can Respir J. 2007;14:465–72.PubMedPubMedCentral Chan CK, Maltais F, Sigouin C, Haddon JM, Ford GT. A randomized controlled trial to assess the efficacy of tiotropium in Canadian patients with chronic obstructive pulmonary disease. Can Respir J. 2007;14:465–72.PubMedPubMedCentral
34.
Zurück zum Zitat Vincken W, van Noord JA, Greefhorst AP, Bantje TA, Kesten S, Korducki L, et al. Improved health outcomes in patients with COPD during 1 yr's treatment with tiotropium. Eur Respir J. 2002;19:209–16.PubMed Vincken W, van Noord JA, Greefhorst AP, Bantje TA, Kesten S, Korducki L, et al. Improved health outcomes in patients with COPD during 1 yr's treatment with tiotropium. Eur Respir J. 2002;19:209–16.PubMed
35.
Zurück zum Zitat Donohue JF, van Noord JA, Bateman ED, Langley SJ, Lee A, Witek TJ Jr, et al. A 6-month, placebo-controlled study comparing lung function and health status changes in COPD patients treated with tiotropium or salmeterol. Chest. 2002;122:47–55.PubMed Donohue JF, van Noord JA, Bateman ED, Langley SJ, Lee A, Witek TJ Jr, et al. A 6-month, placebo-controlled study comparing lung function and health status changes in COPD patients treated with tiotropium or salmeterol. Chest. 2002;122:47–55.PubMed
36.
Zurück zum Zitat Brusasco V, Hodder R, Miravitlles M, Korducki L, Towse L, Kesten S. Health outcomes following treatment for six months with once daily tiotropium compared with twice daily salmeterol in patients with COPD. Thorax. 2003;58:399–404.PubMedPubMedCentral Brusasco V, Hodder R, Miravitlles M, Korducki L, Towse L, Kesten S. Health outcomes following treatment for six months with once daily tiotropium compared with twice daily salmeterol in patients with COPD. Thorax. 2003;58:399–404.PubMedPubMedCentral
37.
Zurück zum Zitat Niewoehner DE, Rice K, Cote C, Paulson D, Cooper JA Jr, Korducki L, et al. Prevention of exacerbations of chronic obstructive pulmonary disease with tiotropium, a once-daily inhaled anticholinergic bronchodilator: a randomized trial. Ann Intern Med. 2005;143:317–26.PubMed Niewoehner DE, Rice K, Cote C, Paulson D, Cooper JA Jr, Korducki L, et al. Prevention of exacerbations of chronic obstructive pulmonary disease with tiotropium, a once-daily inhaled anticholinergic bronchodilator: a randomized trial. Ann Intern Med. 2005;143:317–26.PubMed
38.
Zurück zum Zitat Powrie DJ, Wilkinson TM, Donaldson GC, Jones P, Scrine K, Viel K, et al. Effect of tiotropium on sputum and serum inflammatory markers and exacerbations in COPD. Eur Respir J. 2007;30:472–8.PubMed Powrie DJ, Wilkinson TM, Donaldson GC, Jones P, Scrine K, Viel K, et al. Effect of tiotropium on sputum and serum inflammatory markers and exacerbations in COPD. Eur Respir J. 2007;30:472–8.PubMed
39.
Zurück zum Zitat Vogelmeier C, Hederer B, Glaab T, Schmidt H, Rutten-van Mölken MP, Beeh KM, et al. Tiotropium versus salmeterol for the prevention of exacerbations of COPD. N Engl J Med. 2011;364:1093–103.PubMed Vogelmeier C, Hederer B, Glaab T, Schmidt H, Rutten-van Mölken MP, Beeh KM, et al. Tiotropium versus salmeterol for the prevention of exacerbations of COPD. N Engl J Med. 2011;364:1093–103.PubMed
40.
Zurück zum Zitat van Noord JA, Bantje TA, Eland ME, Korducki L, Cornelissen PJ. A randomised controlled comparison of tiotropium and ipratropium in the treatment of chronic obstructive pulmonary disease. The Dutch Tiotropium Study Group. Thorax. 2000;55:289–94. van Noord JA, Bantje TA, Eland ME, Korducki L, Cornelissen PJ. A randomised controlled comparison of tiotropium and ipratropium in the treatment of chronic obstructive pulmonary disease. The Dutch Tiotropium Study Group. Thorax. 2000;55:289–94.
42.
Zurück zum Zitat Rice KL, Kunisaki KM, Niewoehner DE. Role of tiotropium in the treatment of COPD. Int J Chron Obstruct Pulmon Dis. 2007;2:95–105.PubMedPubMedCentral Rice KL, Kunisaki KM, Niewoehner DE. Role of tiotropium in the treatment of COPD. Int J Chron Obstruct Pulmon Dis. 2007;2:95–105.PubMedPubMedCentral
43.
Zurück zum Zitat Anzueto A, Miravitlles M. Pathophysiology of dyspnea in COPD. Postgrad Med. 2017;129:366–74.PubMed Anzueto A, Miravitlles M. Pathophysiology of dyspnea in COPD. Postgrad Med. 2017;129:366–74.PubMed
44.
Zurück zum Zitat Celli B, ZuWallack R, Wang S, Kesten S. Improvement in resting inspiratory capacity and hyperinflation with tiotropium in COPD patients with increased static lung volumes. Chest. 2003;124:1743–8.PubMed Celli B, ZuWallack R, Wang S, Kesten S. Improvement in resting inspiratory capacity and hyperinflation with tiotropium in COPD patients with increased static lung volumes. Chest. 2003;124:1743–8.PubMed
45.
Zurück zum Zitat O'Donnell DE, Flüge T, Gerken F, Hamilton A, Webb K, Aguilaniu B, et al. Effects of tiotropium on lung hyperinflation, dyspnoea and exercise tolerance in COPD. Eur Respir J. 2004;23:832–40.PubMed O'Donnell DE, Flüge T, Gerken F, Hamilton A, Webb K, Aguilaniu B, et al. Effects of tiotropium on lung hyperinflation, dyspnoea and exercise tolerance in COPD. Eur Respir J. 2004;23:832–40.PubMed
46.
Zurück zum Zitat Maltais F, Hamilton A, Marciniuk D, Hernandez P, Sciurba FC, Richter K, et al. Improvements in symptom-limited exercise performance over 8 h with once-daily tiotropium in patients with COPD. Chest. 2005;128:1168–78.PubMed Maltais F, Hamilton A, Marciniuk D, Hernandez P, Sciurba FC, Richter K, et al. Improvements in symptom-limited exercise performance over 8 h with once-daily tiotropium in patients with COPD. Chest. 2005;128:1168–78.PubMed
47.
Zurück zum Zitat Cooper CB, Celli BR, Jardim JR, Wise RA, Legg D, Guo J, et al. Treadmill endurance during 2-year treatment with tiotropium in patients with COPD: a randomized trial. Chest. 2013;144:490–7.PubMed Cooper CB, Celli BR, Jardim JR, Wise RA, Legg D, Guo J, et al. Treadmill endurance during 2-year treatment with tiotropium in patients with COPD: a randomized trial. Chest. 2013;144:490–7.PubMed
48.
Zurück zum Zitat Seemungal TA, Hurst JR, Wedzicha JA. Exacerbation rate, health status and mortality in COPD--a review of potential interventions. Int J Chron Obstruct Pulmon Dis. 2009;4:203–23.PubMedPubMedCentral Seemungal TA, Hurst JR, Wedzicha JA. Exacerbation rate, health status and mortality in COPD--a review of potential interventions. Int J Chron Obstruct Pulmon Dis. 2009;4:203–23.PubMedPubMedCentral
49.
Zurück zum Zitat Wallace AE, Kaila S, Bayer V, Shaikh A, Shinde MU, Willey VJ, et al. Health care resource utilization and exacerbation rates in patients with COPD stratified by disease severity in a commercially insured population. J Manag Care Spec Pharm. 2019;25:205–17.PubMed Wallace AE, Kaila S, Bayer V, Shaikh A, Shinde MU, Willey VJ, et al. Health care resource utilization and exacerbation rates in patients with COPD stratified by disease severity in a commercially insured population. J Manag Care Spec Pharm. 2019;25:205–17.PubMed
50.
Zurück zum Zitat de Miguel-Diez J, Hernandez-Vazquez J, Lopez-de-Andres A, Alvaro-Meca A, Hernandez-Barrera V, Jimenez-Garcia R. Analysis of environmental risk factors for chronic obstructive pulmonary disease exacerbation: a case-crossover study (2004-2013). PLoS One. 2019;14:e0217143.PubMedPubMedCentral de Miguel-Diez J, Hernandez-Vazquez J, Lopez-de-Andres A, Alvaro-Meca A, Hernandez-Barrera V, Jimenez-Garcia R. Analysis of environmental risk factors for chronic obstructive pulmonary disease exacerbation: a case-crossover study (2004-2013). PLoS One. 2019;14:e0217143.PubMedPubMedCentral
51.
Zurück zum Zitat Farias R, Sedeno M, Beaucage D, Drouin I, Ouellet I, Joubert A, et al. Innovating the treatment of COPD exacerbations: a phone interactive telesystem to increase COPD action plan adherence. BMJ Open Respir Res. 2019;6:e000379.PubMedPubMedCentral Farias R, Sedeno M, Beaucage D, Drouin I, Ouellet I, Joubert A, et al. Innovating the treatment of COPD exacerbations: a phone interactive telesystem to increase COPD action plan adherence. BMJ Open Respir Res. 2019;6:e000379.PubMedPubMedCentral
52.
Zurück zum Zitat Anzueto A, Miravitlles M. Chronic obstructive pulmonary disease exacerbations: a need for action. Am J Med. 2018;131:15–22.PubMed Anzueto A, Miravitlles M. Chronic obstructive pulmonary disease exacerbations: a need for action. Am J Med. 2018;131:15–22.PubMed
53.
Zurück zum Zitat Miravitlles M, Garcia-Polo C, Domenech A, Villegas G, Conget F, de la Roza C. Clinical outcomes and cost analysis of exacerbations in chronic obstructive pulmonary disease. Lung. 2013;191:523–30.PubMed Miravitlles M, Garcia-Polo C, Domenech A, Villegas G, Conget F, de la Roza C. Clinical outcomes and cost analysis of exacerbations in chronic obstructive pulmonary disease. Lung. 2013;191:523–30.PubMed
54.
Zurück zum Zitat Wedzicha JA, Calverley PMA, Albert RK, Anzueto A, Criner GJ, Hurst JR, et al. Prevention of COPD exacerbations: a European Respiratory Society/American Thoracic Society guideline. Eur Respir J. 2017;50:1602265. Wedzicha JA, Calverley PMA, Albert RK, Anzueto A, Criner GJ, Hurst JR, et al. Prevention of COPD exacerbations: a European Respiratory Society/American Thoracic Society guideline. Eur Respir J. 2017;50:1602265.
55.
Zurück zum Zitat Miravitlles M, Soler-Cataluña JJ, Calle M, Molina J, Almagro P, Quintano JA, et al. Spanish guidelines for Management of Chronic Obstructive Pulmonary Disease (GesEPOC) 2017. Pharmacological treatment of stable phase. Arch Bronconeumol. 2017;53:324–35.PubMed Miravitlles M, Soler-Cataluña JJ, Calle M, Molina J, Almagro P, Quintano JA, et al. Spanish guidelines for Management of Chronic Obstructive Pulmonary Disease (GesEPOC) 2017. Pharmacological treatment of stable phase. Arch Bronconeumol. 2017;53:324–35.PubMed
56.
Zurück zum Zitat Alcazar Navarrete B, Ancochea Bermudez J, Garcia-Rio F, Izquierdo Alonso JL, Miravitlles M, Rodriguez Gonzalez-Moro JM, et al. Patients with chronic obstructive pulmonary disease exacerbations: recommendations for diagnosis. Treatment and Care Arch Bronconeumol. 2019;55:478–87.PubMed Alcazar Navarrete B, Ancochea Bermudez J, Garcia-Rio F, Izquierdo Alonso JL, Miravitlles M, Rodriguez Gonzalez-Moro JM, et al. Patients with chronic obstructive pulmonary disease exacerbations: recommendations for diagnosis. Treatment and Care Arch Bronconeumol. 2019;55:478–87.PubMed
57.
Zurück zum Zitat Anzueto A, Miravitlles M. Efficacy of tiotropium in the prevention of exacerbations of COPD. Ther Adv Respir Dis. 2009;3:103–11.PubMed Anzueto A, Miravitlles M. Efficacy of tiotropium in the prevention of exacerbations of COPD. Ther Adv Respir Dis. 2009;3:103–11.PubMed
58.
Zurück zum Zitat Vogelmeier CF, Asijee GM, Kupas K, Beeh KM. Tiotropium and salmeterol in COPD patients at risk of exacerbations: a post hoc analysis from POET-COPD®. Adv Ther. 2015;32:537–47.PubMedPubMedCentral Vogelmeier CF, Asijee GM, Kupas K, Beeh KM. Tiotropium and salmeterol in COPD patients at risk of exacerbations: a post hoc analysis from POET-COPD®. Adv Ther. 2015;32:537–47.PubMedPubMedCentral
59.
Zurück zum Zitat Anzueto A, Tashkin D, Menjoge S, Kesten S. One-year analysis of longitudinal changes in spirometry in patients with COPD receiving tiotropium. Pulm Pharmacol Ther. 2005;18:75–81.PubMed Anzueto A, Tashkin D, Menjoge S, Kesten S. One-year analysis of longitudinal changes in spirometry in patients with COPD receiving tiotropium. Pulm Pharmacol Ther. 2005;18:75–81.PubMed
60.
Zurück zum Zitat Decramer M, Celli B, Kesten S, Lystig T, Mehra S, Tashkin DP, et al. Effect of tiotropium on outcomes in patients with moderate chronic obstructive pulmonary disease (UPLIFT): a prespecified subgroup analysis of a randomised controlled trial. Lancet. 2009;374:1171–8.PubMed Decramer M, Celli B, Kesten S, Lystig T, Mehra S, Tashkin DP, et al. Effect of tiotropium on outcomes in patients with moderate chronic obstructive pulmonary disease (UPLIFT): a prespecified subgroup analysis of a randomised controlled trial. Lancet. 2009;374:1171–8.PubMed
61.
Zurück zum Zitat Troosters T, Celli B, Lystig T, Kesten S, Mehra S, Tashkin DP, et al. Tiotropium as a first maintenance drug in COPD: secondary analysis of the UPLIFT trial. Eur Respir J. 2010;36:65–73.PubMed Troosters T, Celli B, Lystig T, Kesten S, Mehra S, Tashkin DP, et al. Tiotropium as a first maintenance drug in COPD: secondary analysis of the UPLIFT trial. Eur Respir J. 2010;36:65–73.PubMed
62.
Zurück zum Zitat Tashkin DP, Celli B, Kesten S, Lystig T, Mehra S, Decramer M. Long-term efficacy of tiotropium in relation to smoking status in the UPLIFT trial. Eur Respir J. 2010;35:287–94.PubMed Tashkin DP, Celli B, Kesten S, Lystig T, Mehra S, Decramer M. Long-term efficacy of tiotropium in relation to smoking status in the UPLIFT trial. Eur Respir J. 2010;35:287–94.PubMed
63.
Zurück zum Zitat Tashkin D, Celli B, Kesten S, Lystig T, Decramer M. Effect of tiotropium in men and women with COPD: results of the 4-year UPLIFT trial. Respir Med. 2010;104:1495–504.PubMed Tashkin D, Celli B, Kesten S, Lystig T, Decramer M. Effect of tiotropium in men and women with COPD: results of the 4-year UPLIFT trial. Respir Med. 2010;104:1495–504.PubMed
64.
Zurück zum Zitat Tashkin DP, Celli BR, Decramer M, Lystig T, Liu D, Kesten S. Efficacy of tiotropium in COPD patients with FEV1 ≥ 60% participating in the UPLIFT® trial. COPD. 2012;9:289–96.PubMed Tashkin DP, Celli BR, Decramer M, Lystig T, Liu D, Kesten S. Efficacy of tiotropium in COPD patients with FEV1 ≥ 60% participating in the UPLIFT® trial. COPD. 2012;9:289–96.PubMed
65.
Zurück zum Zitat Morjaria JB, Rigby A, Morice AH. Inhaled corticosteroid use and the risk of pneumonia and COPD exacerbations in the UPLIFT study. Lung. 2017;195:281–8.PubMedPubMedCentral Morjaria JB, Rigby A, Morice AH. Inhaled corticosteroid use and the risk of pneumonia and COPD exacerbations in the UPLIFT study. Lung. 2017;195:281–8.PubMedPubMedCentral
66.
Zurück zum Zitat Bateman ED, van Dyk M, Sagriotis A. Comparable spirometric efficacy of tiotropium compared with salmeterol plus fluticasone in patients with COPD: a pilot study. Pulm Pharmacol Ther. 2008;21:20–5.PubMed Bateman ED, van Dyk M, Sagriotis A. Comparable spirometric efficacy of tiotropium compared with salmeterol plus fluticasone in patients with COPD: a pilot study. Pulm Pharmacol Ther. 2008;21:20–5.PubMed
67.
Zurück zum Zitat Wedzicha JA, Calverley PM, Seemungal TA, Hagan G, Ansari Z, Stockley RA, et al. The prevention of chronic obstructive pulmonary disease exacerbations by salmeterol/fluticasone propionate or tiotropium bromide. Am J Respir Crit Care Med. 2008;177:19–26.PubMed Wedzicha JA, Calverley PM, Seemungal TA, Hagan G, Ansari Z, Stockley RA, et al. The prevention of chronic obstructive pulmonary disease exacerbations by salmeterol/fluticasone propionate or tiotropium bromide. Am J Respir Crit Care Med. 2008;177:19–26.PubMed
68.
Zurück zum Zitat Niewoehner DE, Lapidus R, Cote C, Sharafkhaneh A, Plautz M, Johnson P, et al. Therapeutic conversion of the combination of ipratropium and albuterol to tiotropium in patients with chronic obstructive pulmonary disease. Pulm Pharmacol Ther. 2009;22:587–92.PubMed Niewoehner DE, Lapidus R, Cote C, Sharafkhaneh A, Plautz M, Johnson P, et al. Therapeutic conversion of the combination of ipratropium and albuterol to tiotropium in patients with chronic obstructive pulmonary disease. Pulm Pharmacol Ther. 2009;22:587–92.PubMed
69.
Zurück zum Zitat Griffin J, Lee S, Caiado M, Kesten S, Price D. Comparison of tiotropium bromide and combined ipratropium/salbutamol for the treatment of COPD: a UK general practice research database 12-month follow-up study. Prim Care Respir J. 2008;17:104–10.PubMedPubMedCentral Griffin J, Lee S, Caiado M, Kesten S, Price D. Comparison of tiotropium bromide and combined ipratropium/salbutamol for the treatment of COPD: a UK general practice research database 12-month follow-up study. Prim Care Respir J. 2008;17:104–10.PubMedPubMedCentral
70.
Zurück zum Zitat Vogelmeier C, Ramos-Barbon D, Jack D, Piggott S, Owen R, Higgins M, et al. Indacaterol provides 24-hour bronchodilation in COPD: a placebo-controlled blinded comparison with tiotropium. Respir Res. 2010;11:135.PubMedPubMedCentral Vogelmeier C, Ramos-Barbon D, Jack D, Piggott S, Owen R, Higgins M, et al. Indacaterol provides 24-hour bronchodilation in COPD: a placebo-controlled blinded comparison with tiotropium. Respir Res. 2010;11:135.PubMedPubMedCentral
71.
Zurück zum Zitat Buhl R, Dunn LJ, Disdier C, Lassen C, Amos C, Henley M, et al. Blinded 12-week comparison of once-daily indacaterol and tiotropium in COPD. Eur Respir J. 2011;38:797–803.PubMed Buhl R, Dunn LJ, Disdier C, Lassen C, Amos C, Henley M, et al. Blinded 12-week comparison of once-daily indacaterol and tiotropium in COPD. Eur Respir J. 2011;38:797–803.PubMed
72.
Zurück zum Zitat Donohue JF, Fogarty C, Lötvall J, Mahler DA, Worth H, Yorgancioglu A, et al. Once-daily bronchodilators for chronic obstructive pulmonary disease: indacaterol versus tiotropium. Am J Respir Crit Care Med. 2010;182:155–62.PubMed Donohue JF, Fogarty C, Lötvall J, Mahler DA, Worth H, Yorgancioglu A, et al. Once-daily bronchodilators for chronic obstructive pulmonary disease: indacaterol versus tiotropium. Am J Respir Crit Care Med. 2010;182:155–62.PubMed
73.
Zurück zum Zitat Decramer ML, Chapman KR, Dahl R, Frith P, Devouassoux G, Fritscher C, et al. Once-daily indacaterol versus tiotropium for patients with severe chronic obstructive pulmonary disease (INVIGORATE): a randomised, blinded, parallel-group study. Lancet Respir Med. 2013;1:524–33.PubMed Decramer ML, Chapman KR, Dahl R, Frith P, Devouassoux G, Fritscher C, et al. Once-daily indacaterol versus tiotropium for patients with severe chronic obstructive pulmonary disease (INVIGORATE): a randomised, blinded, parallel-group study. Lancet Respir Med. 2013;1:524–33.PubMed
74.
Zurück zum Zitat Beier J, Kirsten AM, Mróz R, Segarra R, Chuecos F, Caracta C, et al. Efficacy and safety of aclidinium bromide compared with placebo and tiotropium in patients with moderate-to-severe chronic obstructive pulmonary disease: results from a 6-week, randomized, controlled phase IIIb study. COPD. 2013;10:511–22.PubMedPubMedCentral Beier J, Kirsten AM, Mróz R, Segarra R, Chuecos F, Caracta C, et al. Efficacy and safety of aclidinium bromide compared with placebo and tiotropium in patients with moderate-to-severe chronic obstructive pulmonary disease: results from a 6-week, randomized, controlled phase IIIb study. COPD. 2013;10:511–22.PubMedPubMedCentral
75.
Zurück zum Zitat Chapman KR, Beeh KM, Beier J, Bateman ED, D'Urzo A, Nutbrown R, et al. A blinded evaluation of the efficacy and safety of glycopyrronium, a once-daily long-acting muscarinic antagonist, versus tiotropium, in patients with COPD: the GLOW5 study. BMC Pulm Med. 2014;14:4.PubMedPubMedCentral Chapman KR, Beeh KM, Beier J, Bateman ED, D'Urzo A, Nutbrown R, et al. A blinded evaluation of the efficacy and safety of glycopyrronium, a once-daily long-acting muscarinic antagonist, versus tiotropium, in patients with COPD: the GLOW5 study. BMC Pulm Med. 2014;14:4.PubMedPubMedCentral
76.
Zurück zum Zitat Dalby RN, Eicher J, Zierenberg B. Development of Respimat® Soft Mist Inhaler™ and its clinical utility in respiratory disorders. Med Devices (Auckl). 2011;4:145–55. Dalby RN, Eicher J, Zierenberg B. Development of Respimat® Soft Mist Inhaler™ and its clinical utility in respiratory disorders. Med Devices (Auckl). 2011;4:145–55.
77.
Zurück zum Zitat Wachtel H, Kattenbeck S, Dunne S, Disse B. The Respimat® development story: patient-centered innovation. Pulm Ther. 2017;3:19–30. Wachtel H, Kattenbeck S, Dunne S, Disse B. The Respimat® development story: patient-centered innovation. Pulm Ther. 2017;3:19–30.
78.
Zurück zum Zitat van Noord JA, Cornelissen PJ, Aumann JL, Platz J, Mueller A, Fogarty C. The efficacy of tiotropium administered via Respimat soft mist inhaler or HandiHaler in COPD patients. Respir Med. 2009;103:22–9.PubMed van Noord JA, Cornelissen PJ, Aumann JL, Platz J, Mueller A, Fogarty C. The efficacy of tiotropium administered via Respimat soft mist inhaler or HandiHaler in COPD patients. Respir Med. 2009;103:22–9.PubMed
80.
Zurück zum Zitat Rau-Berger H, Mitfessel H, Glaab T. Tiotropium Respimat® improves physical functioning in chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2010;5:367–73.PubMedPubMedCentral Rau-Berger H, Mitfessel H, Glaab T. Tiotropium Respimat® improves physical functioning in chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2010;5:367–73.PubMedPubMedCentral
81.
Zurück zum Zitat Wise RA, Anzueto A, Cotton D, Dahl R, Devins T, Disse B, et al. Tiotropium Respimat inhaler and the risk of death in COPD. N Engl J Med. 2013;369:1491–501.PubMed Wise RA, Anzueto A, Cotton D, Dahl R, Devins T, Disse B, et al. Tiotropium Respimat inhaler and the risk of death in COPD. N Engl J Med. 2013;369:1491–501.PubMed
82.
Zurück zum Zitat Anzueto A, Calverley PMA, Mueller A, Metzdorf N, Haensel M, Jardim JR, et al. Demographic characteristics and clinical outcomes in patients from Latin America versus the rest of the world: a TIOSPIR® post-hoc analysis. Arch Bronconeumol. 2018;54:140–8.PubMed Anzueto A, Calverley PMA, Mueller A, Metzdorf N, Haensel M, Jardim JR, et al. Demographic characteristics and clinical outcomes in patients from Latin America versus the rest of the world: a TIOSPIR® post-hoc analysis. Arch Bronconeumol. 2018;54:140–8.PubMed
83.
Zurück zum Zitat Anzueto A, Wise R, Calverley P, Dusser D, Tang W, Metzdorf N, et al. The tiotropium safety and performance in Respimat® (TIOSPIR®) trial: spirometry outcomes. Respir Res. 2015;16:107.PubMedPubMedCentral Anzueto A, Wise R, Calverley P, Dusser D, Tang W, Metzdorf N, et al. The tiotropium safety and performance in Respimat® (TIOSPIR®) trial: spirometry outcomes. Respir Res. 2015;16:107.PubMedPubMedCentral
84.
Zurück zum Zitat Calverley PM, Tetzlaff K, Dusser D, Wise RA, Mueller A, Metzdorf N, et al. Determinants of exacerbation risk in patients with COPD in the TIOSPIR study. Int J Chron Obstruct Pulmon Dis. 2017;12:3391–405.PubMedPubMedCentral Calverley PM, Tetzlaff K, Dusser D, Wise RA, Mueller A, Metzdorf N, et al. Determinants of exacerbation risk in patients with COPD in the TIOSPIR study. Int J Chron Obstruct Pulmon Dis. 2017;12:3391–405.PubMedPubMedCentral
85.
Zurück zum Zitat Singh S, Loke YK, Enright PL, Furberg CD. Mortality associated with tiotropium mist inhaler in patients with chronic obstructive pulmonary disease: systematic review and meta-analysis of randomised controlled trials. BMJ. 2011;342:d3215.PubMedPubMedCentral Singh S, Loke YK, Enright PL, Furberg CD. Mortality associated with tiotropium mist inhaler in patients with chronic obstructive pulmonary disease: systematic review and meta-analysis of randomised controlled trials. BMJ. 2011;342:d3215.PubMedPubMedCentral
86.
Zurück zum Zitat Dong YH, Lin HH, Shau WY, Wu YC, Chang CH, Lai MS. Comparative safety of inhaled medications in patients with chronic obstructive pulmonary disease: systematic review and mixed treatment comparison meta-analysis of randomised controlled trials. Thorax. 2013;68:48–56.PubMed Dong YH, Lin HH, Shau WY, Wu YC, Chang CH, Lai MS. Comparative safety of inhaled medications in patients with chronic obstructive pulmonary disease: systematic review and mixed treatment comparison meta-analysis of randomised controlled trials. Thorax. 2013;68:48–56.PubMed
87.
Zurück zum Zitat Karner C, Chong J, Poole P. Tiotropium versus placebo for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2012:CD009285. Karner C, Chong J, Poole P. Tiotropium versus placebo for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2012:CD009285.
88.
Zurück zum Zitat Bateman ED. Tiotropium Respimat increases the risk of mortality: con. Eur Respir J. 2013;42:590–3.PubMed Bateman ED. Tiotropium Respimat increases the risk of mortality: con. Eur Respir J. 2013;42:590–3.PubMed
89.
Zurück zum Zitat Barnes NC, Jones PW, Davis KJ. Safety of tiotropium through the Handihaler: why did meta-analyses and database studies appear to give a false alarm? Thorax. 2014;69:598–9.PubMed Barnes NC, Jones PW, Davis KJ. Safety of tiotropium through the Handihaler: why did meta-analyses and database studies appear to give a false alarm? Thorax. 2014;69:598–9.PubMed
91.
Zurück zum Zitat Disse B, Metzdorf N, Martin A, Cerasoli F, Leimer I. Mortality associated with tiotropium mist inhaler? A critical appraisal of the authors' selection and use of previously communicated tiotropium Respimat data BMJ 2011;342:d3215. Disse B, Metzdorf N, Martin A, Cerasoli F, Leimer I. Mortality associated with tiotropium mist inhaler? A critical appraisal of the authors' selection and use of previously communicated tiotropium Respimat data BMJ 2011;342:d3215.
92.
Zurück zum Zitat Abrahams R, Moroni-Zentgraf P, Ramsdell J, Schmidt H, Joseph E, Karpel J. Safety and efficacy of the once-daily anticholinergic BEA2180 compared with tiotropium in patients with COPD. Respir Med. 2013;107:854–62.PubMed Abrahams R, Moroni-Zentgraf P, Ramsdell J, Schmidt H, Joseph E, Karpel J. Safety and efficacy of the once-daily anticholinergic BEA2180 compared with tiotropium in patients with COPD. Respir Med. 2013;107:854–62.PubMed
93.
Zurück zum Zitat Andersen L, Høimark L, Nielsen LP. Respimat mist inhaler safety. BMJ (online). 2011;342:d3215. Andersen L, Høimark L, Nielsen LP. Respimat mist inhaler safety. BMJ (online). 2011;342:d3215.
94.
Zurück zum Zitat Miravitlles M, Price D, Rabe KF, Schmidt H, Metzdorf N, Celli B. Comorbidities of patients in tiotropium clinical trials: comparison with observational studies of patients with chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2015;10:549–64.PubMedPubMedCentral Miravitlles M, Price D, Rabe KF, Schmidt H, Metzdorf N, Celli B. Comorbidities of patients in tiotropium clinical trials: comparison with observational studies of patients with chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2015;10:549–64.PubMedPubMedCentral
95.
Zurück zum Zitat Buhl R, Maltais F, Abrahams R, Bjermer L, Derom E, Ferguson G, et al. Tiotropium and olodaterol fixed-dose combination versus mono-components in COPD (GOLD 2-4). Eur Respir J. 2015;45:969–79.PubMedPubMedCentral Buhl R, Maltais F, Abrahams R, Bjermer L, Derom E, Ferguson G, et al. Tiotropium and olodaterol fixed-dose combination versus mono-components in COPD (GOLD 2-4). Eur Respir J. 2015;45:969–79.PubMedPubMedCentral
96.
Zurück zum Zitat Beeh KM, Westerman J, Kirsten AM, Hebert J, Grönke L, Hamilton A, et al. The 24-h lung-function profile of once-daily tiotropium and olodaterol fixed-dose combination in chronic obstructive pulmonary disease. Pulm Pharmacol Ther. 2015;32:53–9.PubMed Beeh KM, Westerman J, Kirsten AM, Hebert J, Grönke L, Hamilton A, et al. The 24-h lung-function profile of once-daily tiotropium and olodaterol fixed-dose combination in chronic obstructive pulmonary disease. Pulm Pharmacol Ther. 2015;32:53–9.PubMed
97.
Zurück zum Zitat O'Donnell DE, Casaburi R, Frith P, Kirsten A, De Sousa D, Hamilton A, et al. Effects of combined tiotropium/olodaterol on inspiratory capacity and exercise endurance in COPD. Eur Respir J. 2017;49:1601348. O'Donnell DE, Casaburi R, Frith P, Kirsten A, De Sousa D, Hamilton A, et al. Effects of combined tiotropium/olodaterol on inspiratory capacity and exercise endurance in COPD. Eur Respir J. 2017;49:1601348.
98.
Zurück zum Zitat Maltais F, O'Donnell D, Gáldiz Iturri JB, Kirsten AM, Singh D, Hamilton A, et al. Effect of 12 weeks of once-daily tiotropium/olodaterol on exercise endurance during constant work-rate cycling and endurance shuttle walking in chronic obstructive pulmonary disease. Ther Adv Respir Dis. 2018;12:1753465818755091. Maltais F, O'Donnell D, Gáldiz Iturri JB, Kirsten AM, Singh D, Hamilton A, et al. Effect of 12 weeks of once-daily tiotropium/olodaterol on exercise endurance during constant work-rate cycling and endurance shuttle walking in chronic obstructive pulmonary disease. Ther Adv Respir Dis. 2018;12:1753465818755091.
99.
Zurück zum Zitat Singh D, Ferguson GT, Bolitschek J, Grönke L, Hallmann C, Bennett N, et al. Tiotropium + olodaterol shows clinically meaningful improvements in quality of life. Respir Med. 2015;109:1312–9.PubMed Singh D, Ferguson GT, Bolitschek J, Grönke L, Hallmann C, Bennett N, et al. Tiotropium + olodaterol shows clinically meaningful improvements in quality of life. Respir Med. 2015;109:1312–9.PubMed
100.
Zurück zum Zitat Beeh KM, Derom E, Echave-Sustaeta J, Grönke L, Hamilton A, Zhai D, et al. The lung function profile of once-daily tiotropium and olodaterol via Respimat® is superior to that of twice-daily salmeterol and fluticasone propionate via Accuhaler® (ENERGITO® study). Int J Chron Obstruct Pulmon Dis. 2016;11:193–205.PubMedPubMedCentral Beeh KM, Derom E, Echave-Sustaeta J, Grönke L, Hamilton A, Zhai D, et al. The lung function profile of once-daily tiotropium and olodaterol via Respimat® is superior to that of twice-daily salmeterol and fluticasone propionate via Accuhaler® (ENERGITO® study). Int J Chron Obstruct Pulmon Dis. 2016;11:193–205.PubMedPubMedCentral
101.
Zurück zum Zitat Troosters T, Maltais F, Leidy N, Lavoie KL, Sedeno M, Janssens W, et al. Effect of bronchodilation, exercise training, and behavior modification on symptoms and physical activity in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2018;198:1021–32. Troosters T, Maltais F, Leidy N, Lavoie KL, Sedeno M, Janssens W, et al. Effect of bronchodilation, exercise training, and behavior modification on symptoms and physical activity in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2018;198:1021–32.
102.
Zurück zum Zitat Calverley PMA, Anzueto AR, Carter K, Grönke L, Hallmann C, Jenkins C, et al. Tiotropium and olodaterol in the prevention of chronic obstructive pulmonary disease exacerbations (DYNAGITO): a double-blind, randomised, parallel-group, active-controlled trial. Lancet Respir Med. 2018;6:337–44.PubMed Calverley PMA, Anzueto AR, Carter K, Grönke L, Hallmann C, Jenkins C, et al. Tiotropium and olodaterol in the prevention of chronic obstructive pulmonary disease exacerbations (DYNAGITO): a double-blind, randomised, parallel-group, active-controlled trial. Lancet Respir Med. 2018;6:337–44.PubMed
103.
Zurück zum Zitat Suissa S. Improving the efficiency of randomized trials: the DYNAGITO example. COPD. 2020;17:4–6. Suissa S. Improving the efficiency of randomized trials: the DYNAGITO example. COPD. 2020;17:4–6.
104.
Zurück zum Zitat Miravitlles M, Urrutia G, Mathioudakis AG, Ancochea J. Efficacy and safety of tiotropium and olodaterol in COPD: a systematic review and meta-analysis. Respir Res. 2017;18:196.PubMedPubMedCentral Miravitlles M, Urrutia G, Mathioudakis AG, Ancochea J. Efficacy and safety of tiotropium and olodaterol in COPD: a systematic review and meta-analysis. Respir Res. 2017;18:196.PubMedPubMedCentral
105.
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;53:1900164. 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;53:1900164.
Metadaten
Titel
Tiotropium in chronic obstructive pulmonary disease – a review of clinical development
verfasst von
Antonio Anzueto
Marc Miravitlles
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2020
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-020-01407-y

Weitere Artikel der Ausgabe 1/2020

Respiratory Research 1/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

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