Skip to main content
Erschienen in: Advances in Therapy 10/2023

Open Access 17.08.2023 | Original Research

Understanding the Clinical Implications of Individual Patient Characteristics and Treatment Choice on the Risk of Exacerbation in Asthma Patients with Moderate–Severe Symptoms

verfasst von: Dave Singh, Sean Oosterholt, Ian Pavord, Gabriel Garcia, Abhijith PG, Oscar Della Pasqua

Erschienen in: Advances in Therapy | Ausgabe 10/2023

Abstract

Introduction

The assessment of future risk has become an important feature in the management of patients with asthma. However, the contribution of patient-specific characteristics and treatment choices to the risk of exacerbation is poorly understood. Here we evaluated the effect of interindividual baseline differences on the risk of exacerbation and treatment performance in patients receiving regular maintenance doses of inhaled corticosteroids (ICS) or ICS/long-acting beta-agonists (LABA) combination therapy.

Methods

Exacerbations and changes to asthma symptoms 5-item Asthma Control Questionnaire (ACQ-5) were simulated over a 12-month period using a time-to-event and a longitudinal model developed from phase III/IV studies in patients with moderate–severe asthma (N = 16,282). Simulations were implemented to explore treatment performance across different scenarios, including randomised designs and real-world settings. Treatment options included regular dosing with ICS monotherapy [fluticasone propionate (FP)] and combination therapy [fluticasone propionate/salmeterol (FP/SAL) or budesonide/formoterol (BUD/FOR)]. Exacerbation rate was analysed using the log-rank test. The cumulative incidence of events was summarised stratified by treatment.

Results

Being a woman, smoker, having higher baseline ACQ-5 and body mass index (BMI) and lower forced expiratory volume in the first second (FEV1) are associated with increased exacerbation risk (p < 0.01). This risk is bigger in winter because of the seasonal variation effect. Across the different scenarios, the use of FP/SAL resulted in a 10% lower annual incidence of exacerbations relative to FP or regular dosing BUD/FOR, independently of baseline characteristics. Similar differences in the annual incidence of exacerbations were also observed between treatments in obese patients (BMI ≥ 25–35 kg/m2) (p < 0.01) and in patients who do not achieve symptom control on FP monotherapy.

Conclusions

Individual baseline characteristics and treatment choices affect future risk. Achieving comparable levels of symptom control whilst on treatment does not imply comparable risk reduction, as shown by the lower exacerbation rates in FP/SAL vs. BUD/FOR-treated patients. These factors should be considered as a basis for personalised clinical management of patients with moderate–severe asthma.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s12325-023-02590-2.
Key Summary Points
Why carry out this study?
The assessment of future risk is an important feature of the clinical management of patients with moderate–severe asthma symptoms and is dependent on multiple interacting factors, including the incidence of previous exacerbations, the presence of type 2 inflammation, and poor asthma control.
The effect of interindividual differences in baseline characteristics and asthma treatment choices on future risk is poorly understood.
Current clinical guidelines are based on a stepwise approach that relies primarily on symptom control, disregarding the influence that individual patient characteristics and treatment choices may have on long-term outcomes.
What was learned from the study?
Being a woman, smoking habit, and interindividual differences in baseline 5-item Asthma Control Questionnaire (ACQ-5), body mass index (BMI), and forced expiratory volume (FEV1) alter the exacerbation risk, irrespective of treatment choice. This effect is further enhanced in winter because of seasonal variation.
After taking into account the contribution of interindividual baseline differences to exacerbation risk, it appears that regular dosing with fluticasone propionate/salmeterol (FP/SAL) results in a significantly lower exacerbation risk relative to fluticasone propionate (FP) or budesonide/formoterol (BUD/FOR), independently from baseline characteristics. This corresponds to 10% lower annual incidence of exacerbations.
Simulation scenarios in a real-world setting indicate that achieving comparable levels of symptom control whilst on treatment does not imply a comparable risk reduction. Such a difference may be associated with corticosteroid-specific properties, which vary between molecules.

Introduction

The assessment of future risk defined as exacerbation events and subsequent deterioration in lung function has become an important feature of the clinical management of patients with moderate–severe asthma symptoms [14]. Future risk is dependent on multiple interacting factors, including the incidence of previous exacerbations, the presence of type 2 inflammation, and poor asthma control [5, 6]. However, the interaction between individual patient characteristics and asthma treatment choices with regard to modification of future risk is poorly understood. This is an important gap, as clinical practice guidelines focus on strategies that emphasise symptom control, rather than the influence that individual patient characteristics and treatment choices have on long-term outcomes [7, 8]. Consequently, potential differences in treatment performance in terms of future risk reduction, which influences long-term outcomes, are also overlooked. An approach can be envisaged where predictors of risk could be used to personalise treatment.
Here, we expand on the concept of clinical phenotypes, which has been widely discussed with regard to patients with severe (refractory) eosinophilic asthma [911]. In contrast to this subgroup of patients, different arguments have been used to explain heterogeneity in response to treatment in patients with moderate–severe asthma, for which no phenotypical evidence exists to support the use of a different approach to treatment. Yet, there have been limited efforts to characterise in an integrated manner the effect of patient baseline characteristics (i.e. treatable traits) and treatment choices on both immediate response (i.e. symptom control) and future risk [12, 13]. Moreover, it is difficult to accurately estimate such a correlation in a real-world setting [14, 15].
An alternative approach is the use of in silico modelling and simulation, and in particular clinical trial simulations. This methodology has been widely applied for the evaluation of novel drugs in early and late stages of development, as well as a tool for evidence synthesis and optimisation of the therapeutic use of medicines across different therapeutic areas [1618]. However, its application in asthma and respiratory medicine is relatively new [19, 20]. Recently, Oosterholt and colleagues used a model-based meta-analysis to explore the influence of treatable traits on the overall risk of exacerbation in patients with moderate–severe asthma symptoms [21]. A significant effect of interindividual differences in BMI, smoking status, sex, and ACQ-5 at baseline on the overall exacerbation risk was reported. This effect was independent of treatment or other extrinsic factors (e.g. season). These results underscore that consideration should be given to individual characteristics beyond symptom control levels in order to properly personalise asthma treatment to achieve optimal long-term outcomes. Moreover, their findings show differences in the response to treatment, which appear to reflect the pharmacological properties (e.g., lung residence time, systemic absorption, glucocorticoid receptor affinity) and therapeutic index of different corticosteroids [22]. The choice of ICS can therefore influence long-term outcomes, including exacerbation risk, which may be crucial for individuals at higher risk despite achieving a comparable level of symptom control [23].
As the control of multiple concurrent factors is not feasible in a prospective randomised controlled study or in a retrospective observational cohort, the use of computer simulations including virtual patient cohorts in a controlled [also known as clinical trial simulations (CTS)] or in a real-world setting [also known as not-in-trial simulations (NITS)] represents a unique opportunity to characterise the influence of each factor, and disentangle it from treatment effect [16]. Here we apply CTS and NITS to evaluate the effect of treatable traits and treatment choice on exacerbation risk in patients with moderate–severe asthma symptoms taking into account the immediate response to treatment as assessed by ACQ-5. Using a cohort of virtual patients with baseline characteristics comparable to those enrolled in actual clinical trials, the incidence and time-to-first event, i.e. moderate or severe exacerbation, was assessed for a range of scenarios including a parallel, controlled study design and a real-world setting. The analysis is based on a time-to-event (TTE) model that describes the time to first exacerbation, and a longitudinal model that describes the individual symptom trajectories over time, considering the concurrent effect of different baseline covariate factors [17, 21].

Methods

Study Subjects

The current investigation describes the results from computer modelling and simulations and as such does not involve human participants. Patient baseline characteristics used for the prediction of exacerbations and time course of symptom control scores were obtained from the pooled population enrolled in the clinical trials listed in the Supplementary Material, all of which have been performed according to relevant ethical and clinical guidelines. All participants enrolled into the original clinical trials have given informed consent. Re-use of the data for the purpose of the current investigation is in alignment with the terms of the informed consent.
All clinical data used for the development and validation of the time-to-event and longitudinal models, as well as those required for generating baseline characteristics for the virtual cohorts were derived from clinical trials, which have been performed according to the Declaration of Helsinki and were approved by the required ethics committee(s) and/or ethics review board(s).

Data Source

The data available for the development and evaluation of the statistical models used in the current analysis were obtained from nine clinical trials in adults with moderate–severe asthma (ADA109055, ADA109057, HZA113091, HZA115150, SAM40027, SAM40056, SAM40065, SAM40086, SAS115359), treated with regular fixed ICS dosing (not maintenance and reliever therapy) with fluticasone propionate (FP) monotherapy, combination therapy with salmeterol (FP/SAL), or budesonide/formoterol (BUD/FOR). These studies were selected on the basis of the availability of accurate individual patient exacerbation event records, clinical and demographic baseline details (Table S1). The dataset includes 1816 observed exacerbation events (first only) from 16,282 adult subjects who were randomised to receive FP (n = 7490), FP/SAL (n = 8049) or BUD/FOR (n = 743) over a period of up to 1 year. An overview of the clinical study protocols along with treatment details and eventual deviations is shown in Table S2. Likewise, the data used for the development of the longitudinal model describing individual ACQ-5 trajectories consisted of a subset of the studies available for the implementation of the time-to-event model. Only those studies that had longitudinal ACQ-5 data (i.e., at baseline and throughout the course of treatment) were pooled for the purpose of the current analysis. An overview of the selected population is summarised in the Appendix, Supplementary Material.

Clinical Trial Simulations

Final parameters of the TTE model previously developed by Oosterholt and colleagues (Table 1) were used for the implementation of the simulation scenarios, which assess the potential implications of treatable traits and treatment choices on the risk of exacerbation [21]. In parallel, the time course of symptoms (ACQ-5) was evaluated using a longitudinal model based on a similar patient population. The TTE model is a fully parametric hazard (survival) model that describes the time to the first moderate or severe exacerbation, defined as the use of systemic corticosteroids for at least 3 days OR in-patient hospitalisation OR emergency department visit due to asthma requiring systemic corticosteroids. The predicted log hazard is a linear sum of functions of smoking status, ACQ-5 BMI, percentage predicted FEV1 (FEV1p) at baseline and sex in relation to treatment choice, and a seasonal factor that includes the effect of winter. Similarly, the longitudinal model characterises the individual ACQ-5 trajectories, taking into account the effect of covariates. These models allow for an integrated estimation of the effect of clinical and demographic baseline characteristics, along with the effect of treatment with ICS monotherapy and ICS/LABA combination therapy.
Table 1
Parameter estimates of the final model describing the time to first exacerbation in patients with moderate–severe asthma
Covariate
Parametera
Estimate
SE
RSE
(%)
Bootstrap median
(5th–95th percentiles)
 
Base hazard (θBASE)
0.188
0.0045
2.4
0.188 (0.170–0.206)
ACQ-5
fHAZACQ-5
(fractional increase in hazard per unit ACQ-5)
0.207
0.0629
30.4
0.199 (0.112–0.319)
Sex
fHAZfemale
(fractional increase in hazard relative to male %)
0.327
0.0831
25.4
0.325 (0.218–0.440)
BMI
fHAZBMI
(fractional increase in hazard per kg/m2)
0.0279
0.007
25.2
0.029 (0.020–0.036)
FEV1
fHAZFEV1
(fractional change in hazard per % change in FEV1)
− 0.00834
0.002
24.0
− 0.0083 (− 0.0118 to − 0.0045)
Smoking
fHAZcurrent smoker
(fractional increase in hazard relative to never smoked)
0.51
0.121
23.7
0.510 (0.297–0.732)
fHAZformer smoker
(fractional increase in hazard relative to never smoked)
0.268
0.0715
26.7
0.264 (0.146–0.419)
Season
fHAZamplitude
(fractional change between season)
0.304
0.0006
0.2
0.302 (0.252–0.358)
Phase shift (years)
0.27
0.0006
0.2
0.262 (0.234–0.306)
Treatment
fHAZBUD/FOR
(fractional increase in hazard relative to FP)
0.321
0.106
33.0
0.334 (0.112–0.536)
fHAZ FP/SAL
(fractional increase in hazard relative to FP)
− 0.308
0.0348
11.3
− 0.311 (− 0.362 to − 0.251)
The base hazard is described using FP monotherapy as the reference treatment. With permission from Oosterholt et al. [21]
ACQ-5 5-item Asthma Control Questionnaire, BMI body mass index, BUD/FOR budesonide/formoterol, FEV1 forced expiratory volume, FP fluticasone propionate, FP/SAL fluticasone propionate/salmeterol, RSE relative standard error, SE standard error
aTime-to-event model—the effects of baseline covariates, treatment and seasonal variation are described by an additive function to the base hazard. Point estimates are depicted by thetas (\(\Theta\)): \(Hazard = \theta _{{BASE}} *\left( {1 + \theta _{{previous\;smoker }} } \right)*\left( {1 + \theta _{{current\;smoker }} } \right)*\left( {1 + \left( {BMI - 27.6} \right)*\theta _{{BMI}} } \right) *\left( {1 + \left( {FEV1_{P} - 73} \right)*\theta _{{FEV1_{P}}} } \right)*\left( {1 + \left( {\text{ACQ-}5_{{baseline}} - 2} \right) \cdot \theta _{{ACQ-5}} } \right)*\left( {1 + \theta _{{FEMALE}} } \right) *\left( {1 + \theta _{{\frac{{BUD}}{{FOR}}}} } \right)*\left( {1 + \theta _{{\frac{{FP}}{{SAL}}}} } \right)*e^{{\theta _{{amp}} \cdot \sin \left( {calendar\;time + \theta _{{phase}} } \right)}}\)  
Treatment arms were defined in a way that interventions reflect the stepwise approach to the management of patients with moderate–severe asthma symptoms, i.e. starting with ICS monotherapy and progressing to regular ICS/LABA combination therapy. Only non-responders to ICS monotherapy were assigned to ICS/LABA. For scenarios in which treatment changes were necessary, response was defined as an improvement in symptom control scores, as predicted by the longitudinal model describing the individual ACQ-5 trajectories over the course of treatment (see Appendix, Supplementary Material for further details on model development and validation).
From a pharmacological perspective, these scenarios represent the effect of a drug with disease-modifying, bronchoprotective, anti-inflammatory properties (i.e. ICS) prior to the addition of a drug with primarily symptomatic, bronchodilatory properties (i.e. LABA). An outline of the clinical trial simulation workflow is shown in Fig. 1. Full details of the protocol design characteristics along with the key assumptions used for the evaluation of the effect of baseline characteristics and treatment choices on exacerbation risk are summarised in Table S4.

Simulation Scenarios

For each simulation scenario baseline characteristics were randomly sampled from 1500 patients from the pooled population of adults (N = 16,282) with moderate–severe asthma. Treatment was assumed to be independent of baseline characteristics and was assigned prior to the simulations. All scenarios included treatment for the period of 1 year, except for the scenario on the seasonal effect which had a duration of 6 months. Each scenario was repeated 500 times, and for each replicate, patient baseline characteristics were re-sampled from the pooled population. Kaplan–Meier estimates of the simulated exacerbation events were summarised both numerically and graphically per simulation scenario. Where appropriate, heat maps were used to explore the effects of individual covariates on the overall risk of exacerbation.
Figure 1 summarises the scenarios that were evaluated to disentangle the effect of relevant demographic and clinical baseline characteristics from that of treatment. To ensure alignment with clinical criteria, results were stratified by baseline covariates according to the following groups or categories: ACQ-5—well controlled (≤ 0.75), not well controlled (> 0.75 to ≤ 1.5) and poorly controlled (> 1.5); BMI:—normal weight (18.5 to < 25 kg/m2), overweight (25 to < 30 kg/m2), obese (30 to < 35 kg/m2) and extremely obese (35+ kg/m2); FEV1p—normal (> 80%), mild/moderate (50–80%) and severe (< 50%) airway obstruction.

Not-in-Trial Simulations (NITS)

The scenario describing the clinical management of adult patients with moderate–severe asthma in a real-life setting was randomly sampled from the pooled population, including a total of 8000 patients per intervention. Similarly to the procedures used for the CTS, each scenario was repeated 500 times, and patient baseline characteristics were re-sampled for each replicate [16]. The use of baseline data from real patients with moderate–severe asthma ensured accurate representation of the range of values and correlations between demographic and clinical characteristics. As reported by Oosterholt and colleagues [21], treatment was assumed to be independent of baseline characteristics and was assigned prior to the simulations (Fig. 1). All patients started on the same treatment (FP), and symptom control scores over time were generated for individual patients using the longitudinal model. An overview of the model parameter estimates is summarised in Table S3. In this real-life setting scenario, patients who did not achieve control after 3 months on ICS monotherapy were switched to regular maintenance dose with SAL/FP or BUD/FOR for a period of up to 12 months. A responder was defined as a patient achieving symptom control (ACQ-5 < 0.75) at 3 months after treatment initiation, whilst a non-responder was any patient whose ACQ-5 score was ≥ 0.75 at 3 months after treatment initiation. Kaplan–Meier estimates of the simulated exacerbation events were summarised both numerically and graphically per treatment arm.
The statistical significance of the effect of baseline characteristics and treatment choices in each scenario was evaluated using cumulative exacerbation data based on a log-rank test (see Supplementary Material, Table S4 for further details) [24].
All simulation scenarios were implemented in NONMEM version 7.3 (Icon Development Solutions, MD, USA). Graphical summaries and statistical analysis were performed in R version 3.1.1 [25].

Results

An assessment of the baseline risk of exacerbation for individual patients, which considers all factors concurrently (i.e. BMI, sex, smoking status, baseline ACQ-5, season and treatment), requires the use of the time-to-event model. Therefore, a heat map (Fig. 2) describing the contribution of demographic and clinical baseline characteristics to the risk of exacerbation was created to visualise the interaction between different baseline characteristics. The colour gradient clearly shows how risk varies with deteriorating symptom control (ACQ-5), lung function (%FEV1p) and increasing BMI. An overview of the baseline characteristics of the virtual patient cohorts included in each simulation scenario is presented in Tables S5 to S12 (Supplementary Material). Patient randomisation across the different treatment arms reflected the data distribution observed in previous clinical trials.
The influence of interindividual differences in baseline characteristics with the strongest effect on the risk of exacerbation (i.e. symptom control, body mass index, sex, and FEV1) has been assessed for ICS monotherapy and combination therapy with FP/SAL and BUD/FOR after regular maintenance dosing.
The effect of ACQ-5 at baseline is summarised for ICS monotherapy and combination therapy with FP/SAL and BUD/FOR in Fig. 3. Cumulative exacerbation events (%) were fewer in patients with well-controlled and not well-controlled symptoms at baseline, vs. poor control (p < 0.01). In addition, these profiles show that differences between treatment arms are drug-specific and independent from symptom control level. FP/SAL results in significantly lower cumulative incidence of exacerbations at 1 year than regular dosing BUD/FOR (8–13%, p < 0.01), irrespective of baseline symptom control level.
A similar effect was observed for BMI (Fig. 4). A statistically significant increase in the cumulative incidence of exacerbations was found in obese (30 to < 35 kg/m2) and extremely obese (35+ kg/m2) relative those with normal weight (18.5 to < 25 kg/m2) following treatment with BUD/FOR (p < 0.01). In contrast, no significant differences in exacerbation risk were observed between normal, overweight, and obese patients following treatment with FP or FP/SAL. A significant increase in the cumulative incidence of exacerbations was detected only for extremely obese patients (p < 0.01).
The effect of sex and treatment on exacerbation risk is shown in Fig. 5. The cumulative incidence of exacerbations differs significantly between male and female patients (p < 0.01). A comparable pattern was found when assessing the effect of airway obstruction, as measured by predicted FEV1 (%) (Fig. 6), which shows that exacerbation risk is higher in patients with mild/moderate (50–80%) and severe (< 50%) obstruction, irrespective of treatment choice (p < 0.01). On the other hand, the exacerbation risk remains significantly lower following treatment with FP/SAL relative to FP monotherapy or BUD/FOR (p < 0.01). A summary of the results on the effect of smoking status, season and treatment switch is shown in Figs. S1, S2 and S3 (Supplementary Material).
The evaluation of the effect of baseline characteristics and treatment choice on exacerbation risk in a real-world setting reveals that achievement of comparable symptom control across treatment arms does not imply the same effect on exacerbation risk (Fig. 7). As shown by the time course of predicted ACQ-5 scores over the period of 12 months, clinical management of the patients who do not respond to FP monotherapy with FP/SAL resulted in a significantly lower exacerbation risk, as compared to BUD/FOR (p < 0.01). Patients remaining on FP monotherapy generally have lower ACQ-5 baseline values, i.e. patients who have adequate symptom control at baseline are more likely to remain on ICS monotherapy. Further details on the response to treatment of the total cohort population stratified by baseline asthma control level and treatment are presented in Figs. S4 and S5 (Supplementary Material).

Discussion

Currently, clinical management of patients with asthma is based on a stepwise approach that relies primarily on symptom control; that is, it involves iterations in which symptoms, risk factors, treatment and response are evaluated. Our analysis shows that long-term response to ICS/LABA, which was assessed in terms of future exacerbation risk, is greatly influenced by patients’ characteristics as well as by the treatment choices. It provides insight into the magnitude of the effect of baseline ACQ-5, BMI, FEV1, smoking status as well as sex and seasonal variation on exacerbation risk. It also allows separation of the treatment effect from these factors. For instance, the cumulative incidence of exacerbations at 1 year was lower for FP/SAL than regular dosing BUD/FOR (8–13%, p < 0.01), where patients with poor control at baseline (ACQ-5 > 1.5) appear to experience the largest effect (Fig. 3). Such a distinction between treatment arms may not be evident in small clinical trials where the sample size affects the precision of the estimated treatment effect and patient baseline symptom control varies across a wide range of ACQ-5 values. Moreover, the use of simulation scenarios indicates that transition to combination therapy with FP/SAL offers a significantly greater reduction in exacerbation risk than higher doses of FP monotherapy. This may be explained by the fact that at therapeutic doses, nearly maximal anti-inflammatory effect is achieved, with further increases in the exposure to ICS having a minor contribution to risk reduction. This is particularly important in those who are overweight or obese, as shown by the difference in the cumulative incidence.
Given that loss of lung function over time in patients with asthma is partially driven by exacerbations, the evidence that differences in BMI, sex and smoking status affect future risk sheds some light onto the ongoing debate about the heterogeneity in individual response to treatment and underlying clinical phenotypes [i.e. type 2 (T2) and non-T2 asthma] [26, 27]. Whilst clinical and demographic factors may not be directly associated with the complex interplay between airway inflammation and airway remodelling, our results suggest that the baseline characteristics identified as covariates on the base hazard parameter describing the risk of exacerbations in moderate to severe asthma could serve as a proxy for airway hyperresponsiveness status.
Even though the benefits of ICS/LABA in this group of patients have been evaluated extensively in different investigations [2830], using simulation scenarios it was possible to show that FP/SAL does not only offer acute symptomatic relief but may also have disease-modifying properties, reducing the incidence of exacerbations. Whilst the definition of disease modification may vary, a disease-modifying treatment can be described as a sustained reduction in symptoms and disease activity beyond the immediate or temporary effects of an intervention [31]. Our results showed that initiation of treatment with FP/SAL in patients with poor symptom control not responding to FP monotherapy resulted in approximately 3.2% reduction in the incidence of exacerbations per year relative to those responding to ICS monotherapy. Such an effect appeared to be drug-specific, as the transition to BUD/FOR did not translate into a reduction in exacerbation risk relative to FP monotherapy. This was corroborated by the effect of FP/SAL on symptom control level. Treatment with FP/SAL led to a larger proportion of patients (42%) achieving symptom improvement (i.e. ACQ-5 > 0.5) compared to BUD/FOR (30%).
Finally, our analysis illustrates that baseline characteristics affect base hazard rate and as such contribute to the instantaneous risk but are not predictive of the overall long-term response to an intervention, which is greatly determined by treatment. Indeed, demographic and clinical baseline characteristics in non-responders to FP do not differ significantly from patients on combination therapy. This implies that individual future risk at the time of diagnosis will be miscalculated if only baseline characteristics are used to predict treatment response.
From a methodological perspective, we recognise that to address the main research question from this investigation one should distinguish the contribution of multiple interacting factors to the instantaneous risk of exacerbation, including baseline covariates, trial design and treatment type. Whilst control or stratification of all these factors may not be feasible in a prospective study, CTS offer an opportunity to stratify and eventually offset the effect of confounding or uncontrolled factors [3134]. Similarly, any attempt to use retrospective data will be fraught with difficulties, as one needs to consider the effect of censoring and other deviations, which cannot be easily accounted for during data analysis.
Our work has some limitations. Consequently, assumptions had to be made regarding the generalisability of the findings from the different simulation scenarios, as similar protocol conditions may not be easily implemented or controlled in a real-life setting. An overview of the main assumptions and limitations is summarised in Table S4. As a result of ethical reasons, data on the incidence of exacerbation following withdrawal of treatment or use of placebo administration over 12 months is not available. Therefore, a reference intervention had to be defined in order to model the effect of the baseline covariates and treatment choices on exacerbation risk. Moreover, as ICS dose was not a covariate in the TTE model; treatment comparisons were performed using the mean and/or mode dose level used during the maintenance phase of treatment. This was based on the underlying dose–response relationships of FP and BUD [21, 3537]. As currently used ICS doses yield nearly maximum pharmacological effect, the impact of varying dose level on basal hazard was assumed to be minor. In fact, here we applied the principles endorsed by Beasley and colleagues [38]. From a statistical perspective, we have also assumed no carryover effect when treatment was switched. In addition, as transition from FP to FP/SAL or BUD/FOR was implemented by design, i.e. switching at a pre-specified time, adjustment or correction in estimates was not deemed necessary [34].
The predicted differences in exacerbation risk reduction following treatment with ICS/LABA combination therapies, which seemed to differ from mean estimates of treatment effect in previously published reports [3941], were also carefully scrutinised. A few points need to be considered to understand such a discrepancy. First, it should be noted that randomisation of patients to different treatment arms does not necessarily provide a balanced distribution of all relevant factors that may affect treatment response (i.e. exacerbation events). In fact, baseline risk is not a criterion for inclusion into a trial. Consequently, it is plausible to have cohorts whose baseline characteristics look comparable but have a different exacerbation risk level at baseline. This is illustrated by the heat maps in Fig. 2, which show that the effect of treatment on risk reduction (i.e. reduction in hazard) is comparable across patients with different baseline characteristics. Second, the short duration and relatively small number of events (i.e. exacerbations) observed in each single study may result in imprecise estimates of the treatment effect. These results are often extrapolated to describe annualised exacerbation rates, which may lead to poor generalisability of the estimates to a larger population. By contrast, the model used for the simulations is based on combined trials that have longer duration, allowing prediction of exacerbation rates at shorter or longer intervals, which are likely to be more precise than the results observed in single, shorter clinical trials. Third, the eventual confounding or selection bias associated with the large, but yet potentially limited pool of studies available for the investigation. This has been substantially dismissed following the evaluation of the predictive performance of the model including additional data from 697 patients receiving BUD/FOR (4.5/160 μg two inhalations bid) and 693 patients receiving FP/SAL (50/250 μg one inhalation bid) who were enrolled into study SAM40040 [42]. This study was not available at the time of model development. This additional data increased the sample size of both treatment groups and corroborated the findings in the original analysis, i.e. that the effect of FP/SAL on exacerbation risk is significantly different from BUD/FOR (p < 0.01). The visual predictive check, including the observed exacerbations over 12 months along with the model-predicted 95% confidence intervals, is shown in Fig. S6. This was complemented by a propensity score matching (PSM) [43, 44]. Propensity scores are often used in observational research to construct an artificial control group, which matches subjects with similar propensity scores in both treatment and control, so that potential confounding is reduced. As it can be seen in Fig. S7, comparison of the survival (observed exacerbation events) in a subset of perfectly matched patients reveals similar or slightly larger differences between treatment arms (FP/SAL vs. BUD/FOR). These results show that the findings obtained with the full dataset are unlikely to be caused by confounding. Moreover, this was corroborated by the calculated E-value, i.e. an alternative approach to sensitivity analyses for unmeasured confounding in observational studies [45], which indicates how strong the unmeasured confounding should be to refute the observed results.

Conclusions

Interindividual differences in sex, baseline ACQ-5, BMI, FEV1, and smoking habit (treatable traits), as well as season alter the exacerbation risk, irrespective of treatment choice. Our investigation also shows that regular dosing with FP/SAL yields a significantly lower exacerbation risk relative to FP or BUD/FOR, independently from baseline characteristics. Of note is the exacerbation risk reduction observed in overweight and obese patients receiving FP/SAL. Most importantly, simulation scenarios in a real-world setting indicate that achieving comparable levels of symptom control whilst on treatment does not imply a comparable risk reduction. Such a difference may be associated with corticosteroid-specific properties, which vary between inhaled corticosteroids [22]. Consequently, patients showing clinically comparable levels of immediate relief and symptom control may not achieve the same long-term reduction in exacerbation risk. These factors should be considered in clinical practice as a basis for personalised management of patients with moderate–severe asthma symptoms.

Acknowledgements

We would like to thank the clinical experts for their input into this work. We also would like to acknowledge the support from Dr Shuying Yang (Clinical Pharmacology Modelling and Simulation, GSK, UK) and Prof Li Wei (Department of Policy and Practice, School of Pharmacy, University College London, UK) for their invaluable statistical insights. Dave Singh is supported by the National Institute for Health Research (NIHR) Manchester Biomedical Research Centre (BRC).

Funding

This investigation is part of the MASTER (Modelling Asthma TrEatment Responses) study, which has been funded by GlaxoSmithKline (study no. 215310). GlaxoSmithKline has also funded the Rapid Service and Open Access Fees.

Author Contributions

Sean Oosterholt was involved in the analysis and interpretation of study data, drafting and critical revision of the manuscript; Abhijith PG was involved the design of the study and interpretation of study data, drafting and critical revision of the manuscript; Ian Pavord, Dave Singh, and Gabriel Garcia were involved in the interpretation of study data, drafting and critical revision of the manuscript; Oscar Della Pasqua was involved in the conception/design and interpretation of study data, drafting and critical revision of the manuscript.

Disclosures

Ian Pavord has received honoraria for speaking at sponsored meetings from AstraZeneca, Boehringer Ingelheim, Aerocrine, Almirall, Novartis, Teva, Chiesi, Sanofi/Regeneron, Menarini and GSK, and payments for organising educational events from AstraZeneca, GSK, Sanofi/Regeneron and Teva; he has received honoraria for attending advisory panels with Genentech, Sanofi/Regeneron, AstraZeneca, Boehringer Ingelheim, GSK, Novartis, Teva, Merck, Circassia, Chiesi and Knopp and payments to support FDA approval meetings from GSK; he has received sponsorship to attend international scientific meetings from Boehringer Ingelheim, GSK, AstraZeneca, Teva and Chiesi; he has received a grant from Chiesi to support a phase 2 clinical trial in Oxford; he is co-patent holder of the rights to the Leicester Cough Questionnaire and has received payments for its use in clinical trials from Merck, Bayer and Insmed; and in 2014–2015 he was an expert witness for a patent dispute involving AstraZeneca and Teva; Dave Singh has received personal fees from Aerogen, AstraZeneca, Boehringer Ingelheim, Chiesi, Cipla, CSL Behring, Epiendo, Genentech, GSK, Glenmark, Gossamerbio, Kinaset, Menarini, Novartis, Pulmatrix, Sanofi, Synairgen, Teva, Theravance and Verona, and is supported by the National Institute for Health Research (NIHR) Manchester Biomedical Research Centre (BRC); Gabriel Garcia has participated in advisory boards for GSK, AstraZeneca, Sanofi, Novartis and Boehringer Ingelheim; he has received honoraria for speaking at sponsored meetings from GSK, Boehringer Ingelheim, AstraZeneca, Sanofi, Phoenix, and Novartis; and is a principal investigator in trials sponsored by GSK, Boehringer Ingelheim, AstraZeneca, Novartis, Sanofi, PPD, Zambon, Parexel, Covance, IQVIA, and Chiesi; Sean Oosterholt, Abhijith PG and Oscar Della Pasqua are GSK employees and hold stocks/shares in GSK.

Compliance with Ethics Guideline

This article is based on in silico modelling and simulation and does not contain any new studies with human participants or animals performed by any of the authors. All clinical data used for the development and validation of the time-to-event and longitudinal models, as well as those required for generating baseline characteristics for the virtual cohorts derived from clinical trials, which have been performed according to the Declaration of Helsinki and were approved by the required ethics committee(s) and/or ethics review board(s). Re-use of the data for the purpose of the current investigation is in alignment with the terms of informed consent.

Data Availability

Anonymised individual participant data and study documents can be requested for further research from www.​clinicalstudydat​arequest.​com.
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial 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-nc/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Reddel HK, FitzGerald JM, Bateman ED, et al. A fundamental change in asthma management: treatment of asthma with short-acting bronchodilators alone is no longer recommended for adults and adolescents. Eur Respir J. 2019;2019:53. Reddel HK, FitzGerald JM, Bateman ED, et al. A fundamental change in asthma management: treatment of asthma with short-acting bronchodilators alone is no longer recommended for adults and adolescents. Eur Respir J. 2019;2019:53.
3.
Zurück zum Zitat Beasley R, Bird G, Harper J, et al. The further paradoxes of asthma management: time for a new approach across the spectrum of asthma severity. Eur Respir J. 2018;52:1800694.PubMedCrossRef Beasley R, Bird G, Harper J, et al. The further paradoxes of asthma management: time for a new approach across the spectrum of asthma severity. Eur Respir J. 2018;52:1800694.PubMedCrossRef
4.
Zurück zum Zitat Pavord ID, Barnes PJ, Lemière C, et al. Diagnosis and assessment of the asthmas. J Allergy Clin Immunol Pract. 2023;11:1–8.PubMedCrossRef Pavord ID, Barnes PJ, Lemière C, et al. Diagnosis and assessment of the asthmas. J Allergy Clin Immunol Pract. 2023;11:1–8.PubMedCrossRef
5.
Zurück zum Zitat Blakey JD, Zaidi S, Shaw DE. Defining and managing risk in asthma. Clin Exp Allergy. 2014;44:1023–32.PubMedCrossRef Blakey JD, Zaidi S, Shaw DE. Defining and managing risk in asthma. Clin Exp Allergy. 2014;44:1023–32.PubMedCrossRef
6.
Zurück zum Zitat Couillard S, Shrimanker R, Lemaire-Paquette S, et al. Longitudinal changes in sputum and blood inflammatory mediators during FeNO suppression testing. Thorax. 2022;77:933–8.PubMedCrossRef Couillard S, Shrimanker R, Lemaire-Paquette S, et al. Longitudinal changes in sputum and blood inflammatory mediators during FeNO suppression testing. Thorax. 2022;77:933–8.PubMedCrossRef
7.
Zurück zum Zitat Quezada W, Kwak ES, Reibman J, et al. Predictors of asthma exacerbation among patients with poorly controlled asthma despite inhaled corticosteroid treatment. Ann Allergy Asthma Immunol. 2016;116:112–7.PubMedCrossRef Quezada W, Kwak ES, Reibman J, et al. Predictors of asthma exacerbation among patients with poorly controlled asthma despite inhaled corticosteroid treatment. Ann Allergy Asthma Immunol. 2016;116:112–7.PubMedCrossRef
8.
Zurück zum Zitat Beuther DA, Murphy KR, Zeiger RS, et al. The asthma impairment and risk questionnaire (AIRQ) control level predicts future risk of asthma exacerbations. J Allergy Clin Immunol Pract. 2022;10:3204-3212.e3202.PubMedCrossRef Beuther DA, Murphy KR, Zeiger RS, et al. The asthma impairment and risk questionnaire (AIRQ) control level predicts future risk of asthma exacerbations. J Allergy Clin Immunol Pract. 2022;10:3204-3212.e3202.PubMedCrossRef
9.
Zurück zum Zitat Gibson PG. Inflammatory phenotypes in adult asthma: clinical applications. Clin Respir J. 2009;3:198–206.PubMedCrossRef Gibson PG. Inflammatory phenotypes in adult asthma: clinical applications. Clin Respir J. 2009;3:198–206.PubMedCrossRef
11.
Zurück zum Zitat de Pérez Llano L, Dacal Rivas D, Blanco Cid N, et al. Phenotype-guided asthma therapy: an alternative approach to guidelines. J Asthma Allergy. 2021;14:207–17.CrossRef de Pérez Llano L, Dacal Rivas D, Blanco Cid N, et al. Phenotype-guided asthma therapy: an alternative approach to guidelines. J Asthma Allergy. 2021;14:207–17.CrossRef
12.
Zurück zum Zitat Martin A, Bauer V, Datta A, et al. Development and validation of an asthma exacerbation prediction model using electronic health record (EHR) data. J Asthma. 2020;57:1339–46.PubMedCrossRef Martin A, Bauer V, Datta A, et al. Development and validation of an asthma exacerbation prediction model using electronic health record (EHR) data. J Asthma. 2020;57:1339–46.PubMedCrossRef
13.
Zurück zum Zitat Ulrik CS, Vijverberg S, Hanania NA, et al. Precision medicine and treatable traits in chronic airway diseases—where do we stand? Curr Opin Pulm Med. 2020;26:33–9.PubMedCrossRef Ulrik CS, Vijverberg S, Hanania NA, et al. Precision medicine and treatable traits in chronic airway diseases—where do we stand? Curr Opin Pulm Med. 2020;26:33–9.PubMedCrossRef
14.
Zurück zum Zitat Blakeston S, Harper G, Zabala MJ. Identifying the drivers of patients’ reliance on short-acting β2-agonists in asthma. J Asthma. 2021;58:1094–101.PubMedCrossRef Blakeston S, Harper G, Zabala MJ. Identifying the drivers of patients’ reliance on short-acting β2-agonists in asthma. J Asthma. 2021;58:1094–101.PubMedCrossRef
15.
Zurück zum Zitat Vrijens B, De Geest S, Hughes DA, et al. A new taxonomy for describing and defining adherence to medications. Br J Clin Pharmacol. 2012;73:691–705.PubMedPubMedCentralCrossRef Vrijens B, De Geest S, Hughes DA, et al. A new taxonomy for describing and defining adherence to medications. Br J Clin Pharmacol. 2012;73:691–705.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Chain AS, Dieleman JP, van Noord C, et al. Not-in-trial simulation I: bridging cardiovascular risk from clinical trials to real-life conditions. Br J Clin Pharmacol. 2013;76:964–72.PubMedPubMedCentralCrossRef Chain AS, Dieleman JP, van Noord C, et al. Not-in-trial simulation I: bridging cardiovascular risk from clinical trials to real-life conditions. Br J Clin Pharmacol. 2013;76:964–72.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat D’Agate S, Chavan C, Manyak M, et al. Model-based meta-analysis of the time to first acute urinary retention or benign prostatic hyperplasia-related surgery in patients with moderate or severe symptoms. Br J Clin Pharmacol. 2021;87:2777–89.PubMedCrossRef D’Agate S, Chavan C, Manyak M, et al. Model-based meta-analysis of the time to first acute urinary retention or benign prostatic hyperplasia-related surgery in patients with moderate or severe symptoms. Br J Clin Pharmacol. 2021;87:2777–89.PubMedCrossRef
18.
Zurück zum Zitat Santen G, Horrigan J, Danhof M, et al. From trial and error to trial simulation. Part 2: an appraisal of current beliefs in the design and analysis of clinical trials for antidepressant drugs. Clin Pharmacol Ther. 2009;86:255–62.PubMedCrossRef Santen G, Horrigan J, Danhof M, et al. From trial and error to trial simulation. Part 2: an appraisal of current beliefs in the design and analysis of clinical trials for antidepressant drugs. Clin Pharmacol Ther. 2009;86:255–62.PubMedCrossRef
19.
Zurück zum Zitat Musuamba FT, Teutonico D, Maas HJ, et al. Prediction of disease progression, treatment response and dropout in chronic obstructive pulmonary disease (COPD). Pharm Res. 2015;32:617–27.PubMedCrossRef Musuamba FT, Teutonico D, Maas HJ, et al. Prediction of disease progression, treatment response and dropout in chronic obstructive pulmonary disease (COPD). Pharm Res. 2015;32:617–27.PubMedCrossRef
20.
Zurück zum Zitat Singh D, Garcia G, Maneechotesuwan K, et al. New versus old: the impact of changing patterns of inhaled corticosteroid prescribing and dosing regimens in asthma management. Adv Ther. 2022;39:1895–914.PubMedPubMedCentralCrossRef Singh D, Garcia G, Maneechotesuwan K, et al. New versus old: the impact of changing patterns of inhaled corticosteroid prescribing and dosing regimens in asthma management. Adv Ther. 2022;39:1895–914.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Oosterholt S, Pavord ID, Brusselle G, et al. Modelling Asthma TrEatment Responses (MASTER): Effect of individual patient characteristics on the risk of exacerbation in moderate or severe asthma: a time-to-event analysis of randomised clinical trials. Brit J Clin Pharmacol. 2023. https://doi.org/10.1111/bcp.15801.CrossRef Oosterholt S, Pavord ID, Brusselle G, et al. Modelling Asthma TrEatment Responses (MASTER): Effect of individual patient characteristics on the risk of exacerbation in moderate or severe asthma: a time-to-event analysis of randomised clinical trials. Brit J Clin Pharmacol. 2023. https://​doi.​org/​10.​1111/​bcp.​15801.CrossRef
22.
Zurück zum Zitat Daley-Yates P, Brealey N, Thomas S, et al. Therapeutic index of inhaled corticosteroids in asthma: a dose-response comparison on airway hyperresponsiveness and adrenal axis suppression. Br J Clin Pharmacol. 2021;87:483–93.PubMedCrossRef Daley-Yates P, Brealey N, Thomas S, et al. Therapeutic index of inhaled corticosteroids in asthma: a dose-response comparison on airway hyperresponsiveness and adrenal axis suppression. Br J Clin Pharmacol. 2021;87:483–93.PubMedCrossRef
23.
Zurück zum Zitat Kwon JW, Jung H, Kim SH, et al. High ACT score is not sufficient to reduce the risk of asthma exacerbations in asthma with low lung function. Respir Med. 2019;150:38–44.PubMedCrossRef Kwon JW, Jung H, Kim SH, et al. High ACT score is not sufficient to reduce the risk of asthma exacerbations in asthma with low lung function. Respir Med. 2019;150:38–44.PubMedCrossRef
24.
Zurück zum Zitat Fine JP, Gray RJ. A proportional hazards model for the subdistribution of a competing risk. J Am Stat Assoc. 1999;94:496–509.CrossRef Fine JP, Gray RJ. A proportional hazards model for the subdistribution of a competing risk. J Am Stat Assoc. 1999;94:496–509.CrossRef
27.
Zurück zum Zitat Robinson D, Humbert M, Buhl R, et al. Revisiting type 2-high and type 2-low airway inflammation in asthma: current knowledge and therapeutic implications. Clin Exp Allergy. 2017;47:161–75.PubMedCrossRef Robinson D, Humbert M, Buhl R, et al. Revisiting type 2-high and type 2-low airway inflammation in asthma: current knowledge and therapeutic implications. Clin Exp Allergy. 2017;47:161–75.PubMedCrossRef
28.
Zurück zum Zitat Aalbers R, Vogelmeier C, Kuna P. Achieving asthma control with ICS/LABA: a review of strategies for asthma management and prevention. Respir Med. 2016;111:1–7.PubMedCrossRef Aalbers R, Vogelmeier C, Kuna P. Achieving asthma control with ICS/LABA: a review of strategies for asthma management and prevention. Respir Med. 2016;111:1–7.PubMedCrossRef
29.
30.
Zurück zum Zitat Vähätalo I, Ilmarinen P, Tuomisto LE, et al. Inhaled corticosteroids and asthma control in adult-onset asthma: 12-year follow-up study. Respir Med. 2018;137:70–6.PubMedCrossRef Vähätalo I, Ilmarinen P, Tuomisto LE, et al. Inhaled corticosteroids and asthma control in adult-onset asthma: 12-year follow-up study. Respir Med. 2018;137:70–6.PubMedCrossRef
32.
33.
Zurück zum Zitat Della Pasqua OE. PKPD and disease modeling: concepts and applications to oncology. Clinical trial simulations. New York: Springer; 2010. p. 281–306. Della Pasqua OE. PKPD and disease modeling: concepts and applications to oncology. Clinical trial simulations. New York: Springer; 2010. p. 281–306.
34.
Zurück zum Zitat Branson M, Whitehead J. Estimating a treatment effect in survival studies in which patients switch treatment. Stat Med. 2002;21:2449–63.PubMedCrossRef Branson M, Whitehead J. Estimating a treatment effect in survival studies in which patients switch treatment. Stat Med. 2002;21:2449–63.PubMedCrossRef
35.
Zurück zum Zitat Borella E, Oosterholt S, Magni P, et al. Characterisation of individual ferritin response in patients receiving chelation therapy. Br J Clin Pharmacol. 2022;88:3683–94.PubMedCrossRef Borella E, Oosterholt S, Magni P, et al. Characterisation of individual ferritin response in patients receiving chelation therapy. Br J Clin Pharmacol. 2022;88:3683–94.PubMedCrossRef
37.
Zurück zum Zitat Hübner M, Hochhaus G, Derendorf H. Comparative pharmacology, bioavailability, pharmacokinetics, and pharmacodynamics of inhaled glucocorticosteroids. Immunol Allergy Clin North Am. 2005;25:469–88.PubMedCrossRef Hübner M, Hochhaus G, Derendorf H. Comparative pharmacology, bioavailability, pharmacokinetics, and pharmacodynamics of inhaled glucocorticosteroids. Immunol Allergy Clin North Am. 2005;25:469–88.PubMedCrossRef
38.
Zurück zum Zitat Beasley R, Harper J, Bird G, et al. Inhaled corticosteroid therapy in adult asthma. Time for a new therapeutic dose terminology. Am J Respir Crit Care Med. 2019;199:1471–7.PubMedCrossRef Beasley R, Harper J, Bird G, et al. Inhaled corticosteroid therapy in adult asthma. Time for a new therapeutic dose terminology. Am J Respir Crit Care Med. 2019;199:1471–7.PubMedCrossRef
39.
Zurück zum Zitat Bousquet J, Boulet LP, Peters MJ, et al. Budesonide/formoterol for maintenance and relief in uncontrolled asthma vs. high-dose salmeterol/fluticasone. Respir Med. 2007;101(12):2437–46.PubMedCrossRef Bousquet J, Boulet LP, Peters MJ, et al. Budesonide/formoterol for maintenance and relief in uncontrolled asthma vs. high-dose salmeterol/fluticasone. Respir Med. 2007;101(12):2437–46.PubMedCrossRef
40.
Zurück zum Zitat Kuna P, Peters MJ, Manjra AI, et al. Effect of budesonide/formoterol maintenance and reliever therapy on asthma exacerbations. Int J Clin Pract. 2007;61(5):725–36.PubMedCrossRef Kuna P, Peters MJ, Manjra AI, et al. Effect of budesonide/formoterol maintenance and reliever therapy on asthma exacerbations. Int J Clin Pract. 2007;61(5):725–36.PubMedCrossRef
41.
Zurück zum Zitat Vogelmeier C, D’Urzo A, Pauwels R, et al. Budesonide/formoterol maintenance and reliever therapy: an effective asthma treatment option? Eur Respir J. 2005;26(5):819–28 (Erratum in: Eur Respir J. 2005; 26(6):1191. Erratum in: Eur Respir J. 2011; 38(2):491).PubMedCrossRef Vogelmeier C, D’Urzo A, Pauwels R, et al. Budesonide/formoterol maintenance and reliever therapy: an effective asthma treatment option? Eur Respir J. 2005;26(5):819–28 (Erratum in: Eur Respir J. 2005; 26(6):1191. Erratum in: Eur Respir J. 2011; 38(2):491).PubMedCrossRef
42.
Zurück zum Zitat Dahl R, Chuchalin A, Gor D, et al. EXCEL: a randomised trial comparing salmeterol/fluticasone propionate and formoterol/budesonide combinations in adults with persistent asthma. Respir Med. 2006;100(7):1152–62.PubMedCrossRef Dahl R, Chuchalin A, Gor D, et al. EXCEL: a randomised trial comparing salmeterol/fluticasone propionate and formoterol/budesonide combinations in adults with persistent asthma. Respir Med. 2006;100(7):1152–62.PubMedCrossRef
43.
Zurück zum Zitat Inacio MC, Chen Y, Paxton EW, et al. Statistics in brief: an introduction to the use of propensity scores. Clin Orthop Relat Res. 2015;473(8):2722–6.PubMedPubMedCentralCrossRef Inacio MC, Chen Y, Paxton EW, et al. Statistics in brief: an introduction to the use of propensity scores. Clin Orthop Relat Res. 2015;473(8):2722–6.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Ho D, Imai K, King G, Stuart E. MatchIt: nonparametric preprocessing for parametric causal inference. J Stat Software. 2011;42(8):1–28.CrossRef Ho D, Imai K, King G, Stuart E. MatchIt: nonparametric preprocessing for parametric causal inference. J Stat Software. 2011;42(8):1–28.CrossRef
45.
Zurück zum Zitat Haneuse S, VanderWeele TJ, Arterburn D. Using the E-value to assess the potential effect of unmeasured confounding in observational studies. JAMA. 2019;321(6):602.PubMedCrossRef Haneuse S, VanderWeele TJ, Arterburn D. Using the E-value to assess the potential effect of unmeasured confounding in observational studies. JAMA. 2019;321(6):602.PubMedCrossRef
Metadaten
Titel
Understanding the Clinical Implications of Individual Patient Characteristics and Treatment Choice on the Risk of Exacerbation in Asthma Patients with Moderate–Severe Symptoms
verfasst von
Dave Singh
Sean Oosterholt
Ian Pavord
Gabriel Garcia
Abhijith PG
Oscar Della Pasqua
Publikationsdatum
17.08.2023
Verlag
Springer Healthcare
Erschienen in
Advances in Therapy / Ausgabe 10/2023
Print ISSN: 0741-238X
Elektronische ISSN: 1865-8652
DOI
https://doi.org/10.1007/s12325-023-02590-2

Weitere Artikel der Ausgabe 10/2023

Advances in Therapy 10/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

„Überwältigende“ Evidenz für Tripeltherapie beim metastasierten Prostata-Ca.

22.05.2024 Prostatakarzinom Nachrichten

Patienten mit metastasiertem hormonsensitivem Prostatakarzinom sollten nicht mehr mit einer alleinigen Androgendeprivationstherapie (ADT) behandelt werden, mahnt ein US-Team nach Sichtung der aktuellen Datenlage. Mit einer Tripeltherapie haben die Betroffenen offenbar die besten Überlebenschancen.

So sicher sind Tattoos: Neue Daten zur Risikobewertung

22.05.2024 Melanom Nachrichten

Das größte medizinische Problem bei Tattoos bleiben allergische Reaktionen. Melanome werden dadurch offensichtlich nicht gefördert, die Farbpigmente könnten aber andere Tumoren begünstigen.

CAR-M-Zellen: Warten auf das große Fressen

22.05.2024 Onkologische Immuntherapie Nachrichten

Auch myeloide Immunzellen lassen sich mit chimären Antigenrezeptoren gegen Tumoren ausstatten. Solche CAR-Fresszell-Therapien werden jetzt für solide Tumoren entwickelt. Künftig soll dieser Prozess nicht mehr ex vivo, sondern per mRNA im Körper der Betroffenen erfolgen.

Frühzeitige HbA1c-Kontrolle macht sich lebenslang bemerkbar

22.05.2024 Typ-2-Diabetes Nachrichten

Menschen mit Typ-2-Diabetes von Anfang an intensiv BZ-senkend zu behandeln, wirkt sich positiv auf Komplikationen und Mortalität aus – und das offenbar lebenslang, wie eine weitere Nachfolgeuntersuchung der UKPD-Studie nahelegt.

Update Innere Medizin

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