Skip to main content

Main menu

  • Home
  • Current issue
  • ERJ Early View
  • Past issues
  • ERS Guidelines
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • Open access
    • COVID-19 submission information
    • Peer reviewer login
  • Alerts
  • Subscriptions
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

User menu

  • Log in
  • Subscribe
  • Contact Us
  • My Cart

Search

  • Advanced search
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

Login

European Respiratory Society

Advanced Search

  • Home
  • Current issue
  • ERJ Early View
  • Past issues
  • ERS Guidelines
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • Open access
    • COVID-19 submission information
    • Peer reviewer login
  • Alerts
  • Subscriptions

Nintedanib in children and adolescents with fibrosing interstitial lung diseases

Robin Deterding, Lisa R. Young, Emily M. DeBoer, David Warburton, Steven Cunningham, Nicolaus Schwerk, Kevin R. Flaherty, Kevin K. Brown, Mihaela Dumistracel, Elvira Erhardt, Julia Bertulis, Martina Gahlemann, Susanne Stowasser, Matthias Griese for the InPedILD trial investigators
European Respiratory Journal 2023 61: 2201512; DOI: 10.1183/13993003.01512-2022
Robin Deterding
1Section of Pediatric Pulmonary and Sleep Medicine, Department of Pediatrics, University of Colorado Denver, Denver, CO, USA
2The Children's Hospital Colorado, Aurora, CO, USA
15These two authors contributed equally
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lisa R. Young
3Division of Pulmonary and Sleep Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
15These two authors contributed equally
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Emily M. DeBoer
1Section of Pediatric Pulmonary and Sleep Medicine, Department of Pediatrics, University of Colorado Denver, Denver, CO, USA
2The Children's Hospital Colorado, Aurora, CO, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
David Warburton
4Children's Hospital Los Angeles, Los Angeles, CA, USA
5Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Steven Cunningham
6Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Nicolaus Schwerk
7Clinic for Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kevin R. Flaherty
8Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kevin K. Brown
9Department of Medicine, National Jewish Health, Denver, CO, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Mihaela Dumistracel
10Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim am Rhein, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Elvira Erhardt
11Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Elvira Erhardt
Julia Bertulis
11Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Martina Gahlemann
12Boehringer Ingelheim (Schweiz) GmbH, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Susanne Stowasser
13Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Matthias Griese
14Hauner Children's Hospital, Ludwig Maximilians University, German Center for Lung Research (DZL), Munich, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background Childhood interstitial lung disease (ILD) comprises a spectrum of rare ILDs affecting infants, children and adolescents. Nintedanib is a licensed treatment for pulmonary fibrosis in adults. The primary objectives of the InPedILD trial were to determine the dose-exposure and safety of nintedanib in children and adolescents with fibrosing ILD.

Methods Patients aged 6–17 years with fibrosing ILD on high-resolution computed tomography and clinically significant disease were randomised 2:1 to receive nintedanib or placebo for 24 weeks and then open-label nintedanib. Dosing was based on weight-dependent allometric scaling. Co-primary end-points were the area under the plasma concentration–time curve at steady state (AUCτ,ss) at weeks 2 and 26 and the proportion of patients with treatment-emergent adverse events at week 24.

Results 26 patients received nintedanib and 13 patients received placebo. The geometric mean (geometric coefficient of variation) AUCτ,ss for nintedanib was 175 µg·h·L−1 (85.1%) in patients aged 6–11 years and 160 µg·h·L−1 (82.7%) in patients aged 12–17 years. In the double-blind period, adverse events were reported in 84.6% of patients in each treatment group. Two patients discontinued nintedanib due to adverse events. Diarrhoea was reported in 38.5% and 15.4% of the nintedanib and placebo groups, respectively. Adjusted mean±se changes in percentage predicted forced vital capacity at week 24 were 0.3±1.3% in the nintedanib group and −0.9±1.8% in the placebo group.

Conclusions In children and adolescents with fibrosing ILD, a weight-based dosing regimen resulted in exposure to nintedanib similar to adults and an acceptable safety profile. These data provide a scientific basis for the use of nintedanib in this patient population.

Abstract

The results of the randomised placebo-controlled InPedILD trial support a positive benefit–risk assessment for use of nintedanib in children and adolescents with fibrosing ILD https://bit.ly/3qnAhUM

Introduction

Childhood interstitial lung disease (chILD) comprises a spectrum of rare and heterogeneous lung disorders affecting infants, children and adolescents that may be associated with significant morbidity [1, 2]. The pathophysiology of chILD often involves a genetic component, sometimes combined with exposure-related injury or autoimmune dysregulation, although in some cases the aetiology remains unknown [1, 3–6]. Some children with ILD develop pulmonary fibrosis. As in adults, in chILD, pulmonary fibrosis involves tissue damage, excessive deposition of extracellular matrix and aberrant remodelling of the lung, although fibroblastic foci are not seen histologically in children with fibrosis [1, 7]. In the absence of licensed drugs for the treatment of chILD, treatment typically involves immunomodulation [8], but the evidence to support this approach is lacking.

Nintedanib is an intracellular inhibitor of tyrosine kinases that inhibits processes fundamental to the progression of pulmonary fibrosis [9, 10]. Randomised controlled trials demonstrated that in adults with idiopathic pulmonary fibrosis, other forms of progressive pulmonary fibrosis and fibrosing ILD associated with systemic sclerosis, nintedanib consistently reduced the rate of decline in forced vital capacity (FVC), a surrogate measure of ILD progression in adults [11], compared with placebo, with an adverse event profile characterised predominantly by gastrointestinal events [12–15]. Based on the clinical efficacy of nintedanib in adults, and presumed similarities in the pathophysiology of fibrotic lung remodelling in adults and children, it was postulated that nintedanib may provide similar benefit in a paediatric population with clinically significant or progressive pulmonary fibrosis. As a confirmatory trial of the efficacy of nintedanib in paediatric patients was deemed not feasible, the InPedILD trial (ClinicalTrials.gov: NCT04093024; EU Clinical Trials Register: EudraCT 2018-004530-14) was designed with the primary objectives of evaluating the dose-exposure and safety of nintedanib in children and adolescents with fibrosing ILD. In addition, data on efficacy end-points were collected to support evaluation of the benefit–risk of nintedanib in the paediatric population.

Methods

Patients

The InPedILD trial enrolled children or adolescents aged 6–17 years with fibrosing ILD on a high-resolution computed tomography (HRCT) scan performed ≤12 months before screening, confirmed by central review (supplementary material), FVC ≥25% predicted and clinically significant disease [16]. Clinically significant disease was defined as a Fan score ≥3 [17] or documented evidence of clinical progression over any time frame. The Fan score assesses disease severity in children and adolescents with ILD based on symptoms, oxygen saturation and pulmonary hypertension; scores range from 1 to 5, with higher scores indicating greater disease severity [17]. Evidence of clinical progression was based on a relative decline in FVC ≥10% predicted, a relative decline in FVC of 5–10% predicted with worsening symptoms, worsening fibrosis on HRCT or other measures of clinical worsening attributed to progressive pulmonary fibrosis (e.g. increased oxygen requirement or decreased diffusion capacity). Key exclusion criteria are listed in the supplementary material.

Trial design

The InPedILD trial was a phase 3 trial conducted at 43 sites in 21 countries [16]. After a 4-week screening period, patients were randomised 2:1 to receive nintedanib or placebo using interactive response technology, stratified by age group (6–11 and 12–17 years) (figure 1). Dosing was based on weight-dependent allometric scaling. Starting doses were 50, 75, 100 or 150 mg twice daily and the dose was adjusted during treatment based on the patient's weight (table 1). Dose reductions to the dose below and treatment interruptions were permitted to manage adverse events. The lowest dose was 25 mg twice daily. Dose re-escalation was allowed within 4 weeks of dose reduction for adverse events considered related to the trial drug or within 8 weeks for adverse events not considered related to the trial drug.

FIGURE 1
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1

Trial design. R: randomisation; EoT: end of treatment.

View this table:
  • View inline
  • View popup
TABLE 1

Dosing and dose adjustments

The trial consisted of a placebo-controlled double-blind period of 24 weeks followed by a variable period during which all patients received open-label nintedanib (figure 1). Patients who prematurely discontinued the study drug were asked to attend all visits as originally planned. Once ≥30 patients (including ≥20 patients aged 12–17 years) had completed pharmacokinetic sampling at week 26 or had prematurely discontinued the trial, recruitment was closed. Following confirmation that sufficient pharmacokinetic data had been collected, all patients were scheduled for an end-of-treatment visit, after which they entered a 4-week follow-up period or rolled over into an open-label extension trial in which all patients received nintedanib (InPedILD-ON: ClinicalTrials.gov: NCT05285982; EU Clinical Trials Register: EudraCT 2020-005554-23).

A steering committee provided scientific advice on the design of the trial. A safety monitoring committee reviewed pharmacokinetic and safety data to determine the safety profile and benefit–risk of nintedanib, and to advise on dose modification, additional assessments, and the appropriateness of further enrolment and continuation of the trial. An adjudication committee was established to adjudicate deaths due to cardiac, respiratory or other causes and review adverse events categorised as major adverse cardiovascular events. The trial was carried out in compliance with the protocol, the principles of the Declaration of Helsinki and the Harmonised Tripartite Guideline for Good Clinical Practice from the International Conference on Harmonisation, EU directive 2001/20/EC/EU regulation 536/2014, and other relevant guidelines. The trial was initiated at each site (listed in the supplementary material) following approval by the respective institutional review board/independent ethics committee and competent authority according to national and international regulations. Written informed consent and assent (where applicable) were obtained prior to trial entry.

End-points

The co-primary end-points were the area under the plasma concentration–time curve at steady state (AUCτ,ss) at week 2 of nintedanib treatment (i.e. at week 2 in patients randomised to receive nintedanib and at week 26 in patients randomised to receive placebo for 24 weeks followed by nintedanib) and the proportion of patients with treatment-emergent adverse events at week 24, which were defined as events with onset from the first intake of the trial drug until the day prior to the first intake of open-label nintedanib or the last intake of randomised treatment plus 28 days (whichever occurred first). Adverse events were coded according to the Medical Dictionary for Regulatory Activities (MedDRA) version 25 (www.meddra.org).

Pathological findings on bone imaging and stunted growth of the dental root on dental imaging were defined as adverse events of special interest. Secondary safety end-points included the proportion of patients with pathological findings of epiphyseal growth plates on imaging and pathological findings on dental examination or imaging at week 24. Follow-up imaging of epiphyseal growth plates was only conducted in patients with open physes. Change from baseline in height-for-age z-score at week 24 was a further end-point.

Secondary efficacy end-points included changes in FVC % pred, peripheral oxygen saturation (SpO2) at rest, 6-min walk test (6MWT) distance and Pediatric Quality of Life (PedsQL) questionnaire scores at week 24. Spirometry was performed using standardised spirometers supplied to each site and according to American Thoracic Society/European Respiratory Society guidelines [18]. FVC % pred values were calculated using equations published by the Global Lung Function Initiative [19]. The PedsQL questionnaire comprised the young child report (5–7 years), the child report (8–12 years) and the teen report (>12 years), as well as a parent report for each age range; each of these scores ranges from 0 to 100, with higher scores indicating better health-related quality of life [20].

Analyses

For the primary pharmacokinetic analysis, AUCτ,ss was calculated using noncompartmental and compartmental analyses and descriptive statistics. In patients randomised to nintedanib, the AUCτ,ss at week 2 was used; if this value was missing, the value at week 26 was used. In patients randomised to placebo, the AUCτ,ss at week 26 (corresponding to week 2 of nintedanib treatment) was used. Pharmacokinetic analyses were conducted in patients who received ≥1 doses of trial medication and provided evaluable data for ≥1 pharmacokinetic end-points without protocol violations relevant to the evaluation of pharmacokinetics. Safety analyses were conducted in patients who received ≥1 doses of trial medication. Adjusted mean changes from baseline in FVC % pred, SpO2, 6MWT distance, PedsQL questionnaire scores and height-for-age z-score at week 24 were analysed using a mixed model for repeated measures, with fixed categorical effects of treatment at each visit and age group and fixed continuous effects of baseline at each visit, and random effect for patient. Adverse events are presented descriptively.

Sample size was calculated based on the evaluation of the primary pharmacokinetic end-point and the need for the clearance parameter to be estimated with adequate precision. Assuming a coefficient of variation (CV) of 70.8% (based on the geometric CV (gCV) of apparent clearance of nintedanib in the plasma at steady state (CL/Fss) following extravascular multiple-dose administration), ≥20 patients with pharmacokinetic measurements per age group (6–11 and 12–17 years) would be needed to achieve ≥80% probability (loosely referred to as power [21]) of having the 95% confidence interval of CL/Fss and AUCτ,ss within 60% and 140% of the geometric mean estimate. Based on the inclusion criteria and a preliminary feasibility assessment, it was anticipated that the study would only enrol enough patients aged ≥12 years to achieve this probability; however, pharmacokinetic data from children aged 6–11 years were also collected and analysed. A sample size of ≥30 patients (≥10 aged 6–11 years and ≥20 aged 12–17 years) was targeted.

Results

Patients

Of 87 patients screened, 39 patients (12 patients aged 6–11 years and 27 patients aged 12–17 years) were randomised and treated (26 patients with nintedanib and 13 patients with placebo) (figure 2 and supplementary table S1). Baseline characteristics were generally balanced between the treatment groups (table 2). The majority of patients were female (61.5%) and White race (79.5%). Mean±sd age was 12.6±3.3 years, weight was 42.2±17.8 kg and FVC was 59.4±21.9% predicted. The most common diagnostic category was surfactant protein deficiency (30.8%). The most frequent criterion met for clinically significant disease was a Fan score ≥3 (59.0%) (supplementary table S2).

FIGURE 2
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2

Disposition of patients during the double-blind period. #: see supplementary table S1; ¶: adverse event (n=2) and other (n=1). ILD: interstitial lung disease; HRCT: high-resolution computed tomography.

View this table:
  • View inline
  • View popup
TABLE 2

Baseline characteristics

Exposure

Mean±sd exposure to trial medication during the double-blind period was 22.3±4.8 weeks in the nintedanib group and 22.6±4.2 weeks in the placebo group (table 3). During this period, four patients (15.4%) in the nintedanib group and one patient (7.7%) in the placebo group had ≥1 dose reductions and three patients (11.5%) in the nintedanib and none in the placebo group had ≥1 treatment interruptions; three patients (11.5%) in the nintedanib group and none in the placebo group discontinued treatment (figure 2). Mean±sd exposure to trial medication over the whole trial was 46.4±22.3 weeks for patients randomised to nintedanib and 49.6±23.6 weeks for patients who received placebo followed by open-label nintedanib (supplementary table S3).

View this table:
  • View inline
  • View popup
TABLE 3

Exposure to trial medication during the double-blind period

Pharmacokinetics

The geometric mean (gCV) AUCτ,ss for nintedanib was 175 µg·h·L−1 (85.1%) in patients aged 6–11 years and 160 µg·h·L−1 (82.7%) in patients aged 12–17 years. Additional pharmacokinetic data are shown in supplementary table S4.

Adverse events

During the double-blind period, adverse events were reported in 22 patients (84.6%) in the nintedanib group and 11 patients (84.6%) in the placebo group (table 4). Diarrhoea was the most frequent adverse event associated with nintedanib, reported in 10 patients (38.5%) in the nintedanib group and two patients (15.4%) in the placebo group. Two patients (7.7%) had adverse events that led to discontinuation of nintedanib (reported as epiphyses premature fusion (not confirmed by central review of imaging) and liver injury). Two patients (7.7%) in the nintedanib group and one patient (7.7%) in the placebo group had serious adverse events (table 4).

View this table:
  • View inline
  • View popup
TABLE 4

Adverse events during the double-blind period

Over the whole trial, adverse events were reported in all patients randomised to nintedanib and in 12 patients (92.3%) who received placebo as randomised treatment and then open-label nintedanib; serious adverse events were reported in five patients (19.2%) randomised to nintedanib and in three patients (23.1%) who received placebo followed by open-label nintedanib (supplementary table S5). No patients had adverse events leading to treatment discontinuation during the open-label period. Two patients (7.7%) randomised to nintedanib had aspartate transaminase and/or alanine transaminase ≥3 times the upper limit of the normal range; none met criteria for Hy's law.

Epiphyseal growth plate and dental examinations

At week 24, 20 patients (76.9%) and nine patients (69.2%) in the nintedanib and placebo groups, respectively, had epiphyseal growth plate imaging. Pathological findings were observed in two patients (7.7%) in the nintedanib group and one patient (7.7%) in the placebo group (supplementary table S6).

At week 24, dental examination was performed in 23 patients (88.5%) and 10 patients (76.9%), and dental imaging in 22 patients (84.6%) and 10 patients (76.9%), in the nintedanib and placebo groups, respectively. On dental examination, new pathological findings were reported in five patients (19.2%) treated with nintedanib and one patient (7.7%) who received placebo. On dental imaging, stunted growth of the dental root was reported in six patients (23.1%) treated with nintedanib and none who received placebo (supplementary table S7). Review by the safety monitoring committee and a dental expert determined that in three patients, root development was completed (i.e. stunted growth was implausible) and in three patients, different angulations between assessments had led to an overread. Treatment was reinitiated in all cases. In five of the six patients, no indication of stunted growth of dental root was identified in central review of dental imaging at week 52. Details of these cases are provided in supplementary table S8.

Height-for-age z-score

Adjusted mean±se changes from baseline in height-for-age z-score at week 24 were −0.05±0.03 in the nintedanib group and −0.03±0.04 in the placebo group (difference −0.02 (95% CI −0.12–0.09); nominal p=0.75) (supplementary figure S1).

Forced vital capacity

Adjusted mean±se changes from baseline in FVC % pred at week 24 were 0.3±1.3% in the nintedanib group and −0.9±1.8% in the placebo group (difference 1.2% (95% CI −3.4–5.8%); nominal p=0.60) (figure 3). The treatment effect of nintedanib observed in this trial was within the range of the effect observed in trials conducted in adults with fibrosing ILDs over 24 weeks (supplementary figure S2).

FIGURE 3
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3

Change in percentage predicted forced vital capacity (FVC) over 24 weeks.

SpO2, 6MWT distance and PedsQL questionnaire

Adjusted mean±se changes from baseline in SpO2 at rest at week 24 were 0.07±0.77% in the nintedanib group and −2.25±1.08% in the placebo group (difference 2.31% (95% CI −0.39–5.02%); nominal p=0.09) (figure 4). Adjusted mean±se changes from baseline in 6MWT distance at week 24 were 17.6±16.5 m in the nintedanib group and 10.5±22.9 m in the placebo group (difference 7.2 (95% CI −50.7–65.0) m; nominal p=0.80) (supplementary table S9).

FIGURE 4
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4

Change in peripheral oxygen saturation (SpO2) at rest over 24 weeks.

Adjusted mean±se changes from baseline in the PedsQL patient report score at week 24 were 6.5±1.9 and 5.5±2.7 in the nintedanib and placebo groups, respectively (difference 1.0 (95% CI −5.8–7.9); nominal p=0.76) (supplementary table S8). Adjusted mean±se changes from baseline in the PedsQL parent report score at week 24 were 5.5±2.5 and 5.6±3.5 in the nintedanib and placebo groups, respectively (difference −0.1 (95% CI −9.0–8.7); nominal p=0.98) (supplementary table S10).

Discussion

The main objectives of the InPedILD trial were to determine the dosing of nintedanib that would result in an exposure in children and adolescents comparable to adults, and to assess the safety of nintedanib in children and adolescents with clinically significant fibrosing ILD. The results demonstrated that nintedanib had an acceptable safety profile in this patient population. As in adults [12–14, 22], the most common adverse event associated with nintedanib in the InPedILD trial was diarrhoea, but the proportion of children who experienced diarrhoea was lower than that observed in adults and there were no discontinuations due to diarrhoea during the double-blind period.

Inhibition of vascular endothelial growth factor receptor (VEGFR) results in decreased angiogenesis, which is essential for bone growth and development [23]. Data from animal models have suggested that nintedanib, an inhibitor of VEGFR [24], may have reversible effects on the epiphyseal growth plates of large bones and an impact on tooth development [25]. Thus, close monitoring of growth, bone and tooth development was implemented in the InPedILD trial. Pathological findings on epiphyseal growth plate imaging showed no evidence of premature closure of the physes. Changes in height-for-age z-scores suggested normal linear growth. Although six cases of stunted dental root growth were reported, these could not be confirmed by an expert paediatric dentist taking into account additional clinical and demographic data. Although reassuring, the potential effects of nintedanib on epiphyseal growth plates and dentition will continue to be monitored in the InPedILD-ON open-label extension study.

During the double-blind period of the InPedILD trial, five patients in the nintedanib group had coronavirus disease 2019 (COVID-19) compared with one patient in the placebo group. Three patients (11.5%) in the nintedanib group and four patients (30.8%) in the placebo group received a COVID-19 vaccine during the double-blind period; a further patient (7.7%) in the placebo group had COVID-19 before the trial. Based on mechanism of action, nonclinical and clinical data, there is no indication that nintedanib would increase the risk of infection or worsen the disease course of COVID-19.

A dose of 150 mg twice daily was investigated in the pivotal trials of nintedanib in adults with pulmonary fibrosis [12–14] and is the recommended dose for adult patients [26]. Studies investigating the impact of exposure on the efficacy and safety of nintedanib in adults support this dose [27, 28]. In the InPedILD trial, dosing was based on weight-dependent allometric scaling. In adults treated with nintedanib 150 mg twice daily, the derived geometric mean (gCV) AUCτ,ss of nintedanib is 203 µg·h·L−1 (67.5%) and the median (5th–95th percentile) AUCτ,ss is 181 (81.5–398) µg·h·L−1 (Boehringer Ingelheim; data on file). Thus, the pharmacokinetic data from the InPedILD trial show that the weight-based dosing regimen achieved exposure in paediatric patients that was comparable to that observed in adults treated with 150 mg twice daily, supporting the use of this dosing regimen in the paediatric population.

FVC was an exploratory end-point in the InPedILD trial. The mean±se change in FVC % pred at week 24 was 0.3±1.3% in the nintedanib group versus −0.9±1.8% in the placebo group. We also observed a stabilisation of SpO2 at rest over 24 weeks in the nintedanib group compared with a decline in the placebo group (mean±se change 0.07±0.77% versus −2.25±1.08%). These observations need to be interpreted with caution given the paucity of data on the natural history of lung function in children with ILD [7, 29, 30] and the short duration of the trial. However, the observed reduction in decline in FVC % pred with nintedanib versus placebo in this paediatric population was within the range observed in adults with fibrosing ILDs treated for 24 weeks.

Conducting clinical trials in children with fibrosing ILD is challenging due to factors including its rarity and a lack of consensus on how to measure the progression of pulmonary fibrosis in children [1]. Other than the InPedILD trial, the only randomised placebo-controlled trial of an intervention in children with fibrosing ILD to have published results is a trial of hydroxychloroquine conducted in Germany, which differed substantially from the InPedILD trial in its design [31]. The InPedILD trial is the first international placebo-controlled trial to have been conducted in children with ILD and as such represents a landmark in the field. However, it also has limitations, including the small sample size and short duration, which limit interpretation of the data. Of note, randomisation of 39 patients in the InPedILD trial required the involvement of dozens of sites, with a failure to meet radiological criteria based on central review the main reason for screen failure.

In conclusion, data from the InPedILD trial demonstrate that nintedanib had an acceptable safety and tolerability profile in children and adolescents with fibrosing ILD, with no new safety signals observed compared with adult patients. Exposure levels achieved with weight-based allometric dosing were within the range observed in adults. These data provide a scientific basis for the use of nintedanib in children and adolescents with fibrosing ILD.

Supplementary material

Supplementary Material

Please note: supplementary material is not edited by the Editorial Office, and is uploaded as it has been supplied by the author.

Supplementary material ERJ-01512-2022.Supplement

Shareable PDF

Supplementary Material

This one-page PDF can be shared freely online.

Shareable PDF ERJ-01512-2022.Shareable

Acknowledgements

We thank the patients who participated in the InPedILD trial and their parents and caregivers. The InPedILD trial was supported by Boehringer Ingelheim International GmbH (BI). The authors meet criteria for authorship as recommended by the International Committee of Medical Journal Editors (ICMJE). The authors did not receive payment for development of this manuscript. Writing assistance was provided by Julie Fleming and Wendy Morris of FleishmanHillard (London, UK), which was contracted and funded by BI. BI was given the opportunity to review the manuscript for medical and scientific accuracy as well as intellectual property considerations.

Footnotes

  • This clinical trial was prospectively registered at ClinicalTrials.gov with identifier number NCT04093024 and EU Clinical Trials Register with EudraCT number 2018-004530-14. To ensure independent interpretation of clinical study results and enable authors to fulfil their role and obligations under the ICMJE criteria, BI grants all external authors access to relevant clinical study data. In adherence with the BI Policy on Transparency and Publication of Clinical Study Data, scientific and medical researchers can request access to clinical study data after publication of the primary manuscript in a journal, regulatory activities are complete and other criteria are met. Researchers should use https://vivli.org to request access and visit https://www.mystudywindow.com/msw/datasharing for further information.

  • A plain language summary of these data is available from www.globalmedcomms.com/respiratory/ERS2022/Deterding/plain-language-summary

  • Conflict of interest: R. Deterding reports consultancy fees, fees for participation on the steering committee of the InPedILD trial and support for travel from Boehringer Ingelheim (BI); and consultancy fees from Roche. L.R. Young reports grants paid to her institution from the National Institutes of Health and Orphan Disease Center at the University of Pennsylvania; royalties from UpToDate; consultancy fees and fees for participation on the steering committee of the InPedILD trial from BI; consultancy fees from Roche and Sanofi; and honoraria from NYU Langone Health. E.M. DeBoer reports fees from BI for participation on the steering committee of the InPedILD trial. S. Cunningham reports fees paid to his institution by BI for his participation on the steering committee of the InPedILD trial. D. Warburton reports a grant re: the RECOVER (COVID-19) initiative from the National Heart, Lung, and Blood Institute (NHLBI) and fees from BI for participation on the steering committee of the InPedILD trial. N. Schwerk reports consultancy fees from Novartis; payment for lectures from AbbVie, Allergopharma, Chiesi, Infectopharm, Novartis, Pfizer and Sanofi; fees for participation in data safety monitoring boards or advisory boards for BI and Novartis; and fees from BI for participation on the steering committee of the InPedILD trial; he is a member of the German Society for Pediatric Pneumology, European Respiratory Society, Children's Interstitial Lung Disease Clinical Research Collaboration (chILD-EU), European Cooperation in Science and Technology (COST) network for translational research in children's and adult ILD (ENTeR-chILD), and Secretary of Kinderlungenregister. K.R. Flaherty reports grants paid to his institution from BI; royalties from UpToDate; and consultancy fees from Arrowhead, AstraZeneca, Bellerophon, BI, CSL Behring, Daewoong, DevPro, Dispersol, FibroGen, Horizon, Immunet, Lupin, NeRRe Therapeutics, Pliant, Polarean, Pure Health, PureTech, Respivant, Roche/Genentech, Shionogi, Sun Pharmaceuticals, Trevi and United Therapeutics; he is a steering committee chair for the Pulmonary Fibrosis Foundation and reports fees from BI for participation on the steering committee of the InPedILD trial. K.K. Brown reports grants from NHLBI; consultancy fees, speaker fees, support for travel and/or has served as an advisor or on a data monitoring committee for AbbVie, Biogen, Blade Therapeutics, BI, Bristol Myers Squibb, CSL Behring, DevPro Biopharma, Dispersol, Eleven P15, Galapagos, Galecto, Huitai Biomedcine, Humanetics, the Open Source Imaging Consortium (OSIC), Pliant, Redx Pharma, Sanofi, Third Pole and Translate Bio; and fees from BI for participation on the steering committee of the InPedILD trial; he holds leadership or fiduciary roles with the Fleischner Society and OSIC. M. Dumistracel, E. Erhardt, J. Bertulis, M. Gahlemann and S. Stowasser are employees of BI. M. Griese reports grants from BI for analysis of the chILD-EU register; has received fees from BI for membership of the InPedILD steering committee, an advisory board and an adjudication board, and for a web-based series.

  • Support statement: The InPedILD trial was supported by Boehringer Ingelheim International GmbH (BI). Funding information for this article has been deposited with the Crossref Funder Registry.

  • Received July 29, 2022.
  • Accepted August 25, 2022.
  • Copyright ©The authors 2023.
http://creativecommons.org/licenses/by-nc/4.0/

This version is distributed under the terms of the Creative Commons Attribution Non-Commercial Licence 4.0. For commercial reproduction rights and permissions contact permissions{at}ersnet.org

References

  1. ↵
    1. Deterding RR,
    2. DeBoer EM,
    3. Cidon MJ, et al.
    Approaching clinical trials in childhood interstitial lung disease and pediatric pulmonary fibrosis. Am J Respir Crit Care Med 2019; 200: 1219–1227. doi:10.1164/rccm.201903-0544CI
    OpenUrlCrossRef
  2. ↵
    1. Griese M
    . Etiologic classification of diffuse parenchymal (interstitial) lung diseases. J Clin Med 2022; 11: 1747. doi:10.3390/jcm11061747
    OpenUrl
  3. ↵
    1. Hartl D,
    2. Griese M
    . Interstitial lung disease in children – genetic background and associated phenotypes. Respir Res 2005; 6: 32. doi:10.1186/1465-9921-6-32
    OpenUrlCrossRefPubMed
    1. Glasser SW,
    2. Hardie WD,
    3. Hagood JS
    . Pathogenesis of interstitial lung disease in children and adults. Pediatr Allergy Immunol Pulmonol 2010; 23: 9–14. doi:10.1089/ped.2010.0004
    OpenUrlCrossRefPubMed
    1. Cidon M,
    2. Bansal M,
    3. Hartl D
    . Pulmonary manifestations of rheumatologic diseases. Curr Opin Pediatr 2017; 29: 311–319. doi:10.1097/MOP.0000000000000496
    OpenUrl
  4. ↵
    1. Cunningham S,
    2. Jaffe A,
    3. Young LR
    . Children's interstitial and diffuse lung disease. Lancet Child Adolesc Health 2019; 3: 568–577. doi:10.1016/S2352-4642(19)30117-8
    OpenUrl
  5. ↵
    1. Nathan N,
    2. Thouvenin G,
    3. Fauroux B, et al.
    Interstitial lung disease: physiopathology in the context of lung growth. Paediatr Respir Rev 2011; 12: 216–222. doi:10.1016/j.prrv.2011.04.003
    OpenUrlCrossRefPubMed
  6. ↵
    1. Bush A,
    2. Cunningham S,
    3. de Blic J, et al.
    European protocols for the diagnosis and initial treatment of interstitial lung disease in children. Thorax 2015; 70: 1078–1084. doi:10.1136/thoraxjnl-2015-207349
    OpenUrlAbstract/FREE Full Text
  7. ↵
    1. Wollin L,
    2. Wex E,
    3. Pautsch A, et al.
    Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J 2015; 45: 1434–1445. doi:10.1183/09031936.00174914
    OpenUrlAbstract/FREE Full Text
  8. ↵
    1. Wollin L,
    2. Distler JHW,
    3. Redente EF, et al.
    Potential of nintedanib in treatment of progressive fibrosing interstitial lung diseases. Eur Respir J 2019; 54: 1900161. doi:10.1183/13993003.00161-2019
    OpenUrlPubMed
  9. ↵
    1. Paterniti MO,
    2. Bi Y,
    3. Rekić D, et al.
    Acute exacerbation and decline in forced vital capacity are associated with increased mortality in idiopathic pulmonary fibrosis. Ann Am Thorac Soc 2017; 14: 1395–1402. doi:10.1513/AnnalsATS.201606-458OC
    OpenUrlPubMed
  10. ↵
    1. Richeldi L,
    2. du Bois RM,
    3. Raghu G, et al.
    Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med 2014; 370: 2071–2082. doi:10.1056/NEJMoa1402584
    OpenUrlCrossRefPubMedWeb of Science
    1. Distler O,
    2. Highland KB,
    3. Gahlemann M, et al.
    Nintedanib for systemic sclerosis-associated interstitial lung disease. N Engl J Med 2019; 380: 2518–2528. doi:10.1056/NEJMoa1903076
    OpenUrlCrossRefPubMed
  11. ↵
    1. Flaherty KR,
    2. Wells AU,
    3. Cottin V, et al.
    Nintedanib in progressive fibrosing interstitial lung diseases. N Engl J Med 2019; 381: 1718–1727. doi:10.1056/NEJMoa1908681
    OpenUrlCrossRefPubMed
  12. ↵
    1. Bonella F,
    2. Cottin V,
    3. Valenzuela C, et al.
    Meta-analysis of effect of nintedanib on reducing FVC decline across interstitial lung diseases. Adv Ther 2022; 39: 3392–3402. doi:10.1007/s12325-022-02145-x
    OpenUrl
  13. ↵
    1. Deterding R,
    2. Griese M,
    3. Deutsch G, et al.
    Study design of a randomised, placebo-controlled trial of nintedanib in children and adolescents with fibrosing interstitial lung disease. ERJ Open Res 2021; 7: 00805-2020. doi:10.1183/23120541.00805-2020
    OpenUrlAbstract/FREE Full Text
  14. ↵
    1. Fan LL,
    2. Kozinetz CA
    . Factors influencing survival in children with chronic interstitial lung disease. Am J Respir Crit Care Med 1997; 156: 939–942. doi:10.1164/ajrccm.156.3.9703051
    OpenUrlPubMedWeb of Science
  15. ↵
    1. Miller MR,
    2. Hankinson J,
    3. Brusasco V, et al.
    Standardisation of spirometry. Eur Respir J 2005; 26: 319–338. doi:10.1183/09031936.05.00034805
    OpenUrlAbstract/FREE Full Text
  16. ↵
    1. Quanjer PH,
    2. Stanojevic S,
    3. Cole TJ, et al.
    Multi-ethnic reference values for spirometry for the 3–95-yr age range: the global lung function 2012 equations. Eur Respir J 2012; 40: 1324–1343. doi:10.1183/09031936.00080312
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Niemitz M,
    2. Schwerk N,
    3. Goldbeck L, et al.
    Development and validation of a health-related quality of life questionnaire for pediatric patients with interstitial lung disease. Pediatr Pulmonol 2018; 53: 954–963. doi:10.1002/ppul.24018
    OpenUrlPubMed
  18. ↵
    1. Wang Y,
    2. Jadhav PR,
    3. Lala M, et al.
    Clarification on precision criteria to derive sample size when designing pediatric pharmacokinetic studies. J Clin Pharmacol 2012; 52: 1601–1606. doi:10.1177/0091270011422812
    OpenUrlCrossRefPubMed
  19. ↵
    1. Lancaster L,
    2. Crestani B,
    3. Hernandez P, et al.
    Safety and survival data in patients with idiopathic pulmonary fibrosis treated with nintedanib: pooled data from six clinical trials. BMJ Open Respir Res 2019; 6: e000397. doi:10.1136/bmjresp-2018-000397
    OpenUrlAbstract/FREE Full Text
  20. ↵
    1. Hu K,
    2. Olsen BR
    . The roles of vascular endothelial growth factor in bone repair and regeneration. Bone 2016; 91: 30–38. doi:10.1016/j.bone.2016.06.013
    OpenUrl
  21. ↵
    1. Hilberg F,
    2. Roth GJ,
    3. Krssak M, et al.
    BIBF 1120: triple angiokinase inhibitor with sustained receptor blockade and good antitumor efficacy. Cancer Res 2008; 68: 4774–4782. doi:10.1158/0008-5472.CAN-07-6307
    OpenUrlAbstract/FREE Full Text
  22. ↵
    1. Food and Drug Administration
    . OFEV (nintedanib) pharmacology/toxicology NDA/BLA review and evaluation. 2014. www.accessdata.fda.gov/drugsatfda_docs/nda/2014/205832Orig1s000PharmR.pdf Date last accessed: 23 June 2022.
  23. ↵
    1. Boehringer Ingelheim International GmbH
    . OFEV (nintedanib) prescribing information. 2022. https://docs.boehringer-ingelheim.com/Prescribing%20Information/PIs/Ofev/ofev.pdf Date last accessed: 23 June 2022.
  24. ↵
    1. Schmid U,
    2. Weber B,
    3. Magnusson MO, et al.
    Exposure-efficacy analyses of nintedanib in patients with chronic fibrosing interstitial lung disease. Respir Med 2021; 180: 106369. doi:10.1016/j.rmed.2021.106369
    OpenUrl
  25. ↵
    1. Schmid U,
    2. Weber B,
    3. Sarr C, et al.
    Exposure–safety analyses of nintedanib in patients with chronic fibrosing interstitial lung disease. BMC Pulm Med 2021; 21: 244. doi:10.1186/s12890-021-01598-0
    OpenUrl
  26. ↵
    1. Ring AM,
    2. Carlens J,
    3. Bush A, et al.
    Pulmonary function testing in children's interstitial lung disease. Eur Respir Rev 2020; 29: 200019. doi:10.1183/16000617.0019-2020
    OpenUrlAbstract/FREE Full Text
  27. ↵
    1. DeBoer EM,
    2. Liptzin DR,
    3. Humphries SM, et al.
    Ground glass and fibrotic change in children with surfactant protein C dysfunction mutations. Pediatr Pulmonol 2021; 56: 2223–2231. doi:10.1002/ppul.25356
    OpenUrl
  28. ↵
    1. Griese M,
    2. Kappler M,
    3. Stehling F, et al.
    Randomized controlled phase 2 trial of hydroxychloroquine in childhood interstitial lung disease. Orphanet J Rare Dis 2022; 17: 289. doi:10.1186/s13023-022-02399-2
    OpenUrl
PreviousNext
Back to top
View this article with LENS
Vol 61 Issue 2 Table of Contents
European Respiratory Journal: 61 (2)
  • Table of Contents
  • Index by author
Email

Thank you for your interest in spreading the word on European Respiratory Society .

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Nintedanib in children and adolescents with fibrosing interstitial lung diseases
(Your Name) has sent you a message from European Respiratory Society
(Your Name) thought you would like to see the European Respiratory Society web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
Citation Tools
Nintedanib in children and adolescents with fibrosing interstitial lung diseases
Robin Deterding, Lisa R. Young, Emily M. DeBoer, David Warburton, Steven Cunningham, Nicolaus Schwerk, Kevin R. Flaherty, Kevin K. Brown, Mihaela Dumistracel, Elvira Erhardt, Julia Bertulis, Martina Gahlemann, Susanne Stowasser, Matthias Griese
European Respiratory Journal Feb 2023, 61 (2) 2201512; DOI: 10.1183/13993003.01512-2022

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Nintedanib in children and adolescents with fibrosing interstitial lung diseases
Robin Deterding, Lisa R. Young, Emily M. DeBoer, David Warburton, Steven Cunningham, Nicolaus Schwerk, Kevin R. Flaherty, Kevin K. Brown, Mihaela Dumistracel, Elvira Erhardt, Julia Bertulis, Martina Gahlemann, Susanne Stowasser, Matthias Griese
European Respiratory Journal Feb 2023, 61 (2) 2201512; DOI: 10.1183/13993003.01512-2022
Reddit logo Technorati logo Twitter logo Connotea logo Facebook logo Mendeley logo
Full Text (PDF)

Jump To

  • Article
    • Abstract
    • Abstract
    • Introduction
    • Methods
    • Results
    • Discussion
    • Supplementary material
    • Shareable PDF
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Subjects

  • Interstitial and orphan lung disease
  • Tweet Widget
  • Facebook Like
  • Google Plus One

More in this TOC Section

  • Tool to detect small airways dysfunction in asthma clinical practice
  • Cytokines as prognostic biomarkers in PAH
  • Airway inflammation in subjects with respiratory symptoms and normal spirometry
Show more Original Research Articles

Related Articles

Navigate

  • Home
  • Current issue
  • Archive

About the ERJ

  • Journal information
  • Editorial board
  • Press
  • Permissions and reprints
  • Advertising

The European Respiratory Society

  • Society home
  • myERS
  • Privacy policy
  • Accessibility

ERS publications

  • European Respiratory Journal
  • ERJ Open Research
  • European Respiratory Review
  • Breathe
  • ERS books online
  • ERS Bookshop

Help

  • Feedback

For authors

  • Instructions for authors
  • Publication ethics and malpractice
  • Submit a manuscript

For readers

  • Alerts
  • Subjects
  • Podcasts
  • RSS

Subscriptions

  • Accessing the ERS publications

Contact us

European Respiratory Society
442 Glossop Road
Sheffield S10 2PX
United Kingdom
Tel: +44 114 2672860
Email: journals@ersnet.org

ISSN

Print ISSN:  0903-1936
Online ISSN: 1399-3003

Copyright © 2023 by the European Respiratory Society