You are on page 1of 9

Articles

Inhaled treprostinil and forced vital capacity in patients with


interstitial lung disease and associated pulmonary
hypertension: a post-hoc analysis of the INCREASE study
Steven D Nathan, Aaron Waxman, Sudarshan Rajagopal, Amy Case, Shilpa Johri, Hilary DuBrock, David J De La Zerda, Sandeep Sahay,
Christopher King, Lana Melendres-Groves, Peter Smith, Eric Shen, Lisa D Edwards, Andrew Nelsen, Victor F Tapson

Summary
Background INCREASE was a randomised, placebo-controlled, phase 3 trial that evaluated inhaled treprostinil in Lancet Respir Med 2021
patients with interstitial lung disease (ILD) and associated pulmonary hypertension. Treprostinil improved exercise Published Online
capacity from baseline to week 16, assessed with the use of a 6-min walk test, compared with placebo. Improvements June 29, 2021
https://doi.org/10.1016/
in forced vital capacity (FVC) were also reported. The aim of this post-hoc analysis was to further characterise the
S2213-2600(21)00165-X
effects of inhaled treprostinil on FVC in the overall study population and in various subgroups of interest.
See Online/Comment
https://doi.org/10.1016/
Methods In this post-hoc analysis, we evaluated FVC changes in the overall study population and in various subgroups S2213-2600(21)00264-2
defined by cause of disease or baseline clinical parameters. The study population included patients aged 18 years and Advanced Lung Disease and
older who had a diagnosis of ILD based on evidence of diffuse parenchymal lung disease on chest CT done within Transplant Program, Inova
6 months before random assignment (not centrally adjudicated). All analyses were done on the intention-to-treat Fairfax Hospital, Falls Church,
VA, USA (Prof S D Nathan MD,
population, defined as individuals who were randomly assigned and received at least one dose of study drug. The C King MD); Pulmonary and
INCREASE study is registered with ClinicalTrials.gov, NCT02630316. Critical Care Medicine,
Department of Internal
Medicine, Brigham and
Findings Between Feb 3, 2017, and Aug 30, 2019, 326 patients were enrolled in the INCREASE trial. Inhaled treprostinil
Women’s Hospital, Boston, MA,
was associated with a placebo-corrected least squares mean improvement in FVC of 28·5 mL (SE 30·1; 95% CI USA (A Waxman MD); Division
–30·8 to 87·7; p=0·35) at week 8 and 44·4 mL (35·4; –25·2 to 114·0; p=0·21) at week 16, with associated percentage of Cardiology, Department of
of predicted FVC improvements of 1·8% (0·7; 0·4 to 3·2; p=0·014) and 1·8% (0·8; 0·2 to 3·4; p=0·028). Subgroup Medicine, Duke University
Medical Center, Durham, NC,
analysis of patients with idiopathic interstitial pneumonia showed FVC differences of 46·5 mL (SE 39·9; 95% CI
USA (S Rajagopal MD);
–32·5 to 125·5; p=0·25) at week 8 and 108·2 mL (46·9; 15·3 to 201·1; p=0·023) at week 16. Analysis of patients with Piedmont Healthcare, Austell,
idiopathic pulmonary fibrosis showed FVC differences of 84·5 mL (52·7; –20·4 to 189·5; p=0·11) at week 8 and GA, USA (A Case MD);
168·5 mL (64·5; 40·1 to 297·0; p=0·011) at week 16. The most frequent adverse events included cough, headache, Pulmonary Associates of
Richmond, Richmond, VA, USA
dyspnoea, dizziness, nausea, fatigue, and diarrhoea.
(S Johri MD); Department of
Internal Medicine, Division of
Interpretation In patients with ILD and associated pulmonary hypertension, inhaled treprostinil was associated with Pulmonary and Critical Care,
improvements in FVC versus placebo at 16 weeks. This difference was most evident in patients with idiopathic Mayo Clinic, Rochester, MN,
USA (H DuBrock MD); Division
interstitial pneumonia, particularly idiopathic pulmonary fibrosis. Inhaled treprostinil appears to be a promising
of Pulmonary & Critical Care
therapy for idiopathic pulmonary fibrosis that warrants further investigation in a prospective, randomised, placebo- Medicine, University of Miami
controlled study. Health System, Miami, FL, USA
(D J De La Zerda MD); Division of
Pulmonary, Critical Care and
Funding United Therapeutics Corporation. Sleep Medicine, Houston
Methodist Hospital, Houston,
Copyright © 2021 Elsevier Ltd. All rights reserved. TX, USA (S Sahay MD);
Pulmonary & Critical Care
Division, University of New
Introduction fibrosis, and unclassifiable ILD—indicate that both Mexico, 1 University of New
Interstitial lung diseases (ILDs) encompass a broad agents have antifibrotic effects beyond idiopathic Mexico, DoIM MSC10-5550,
range of conditions that are categorised by varying pulmonary fibrosis.5,6,7 These results suggest that once Albuquerque, NM, USA
amounts of inflammation and fibrosis. Idiopathic lung fibrosis supervenes, there are pathways common to (L Melendres-Groves MD);
United Therapeutics
interstitial pneumonias are a category of ILD, of which various forms of ILD that might be targets for therapy. Corporation, Research Triangle
idiopathic pulmonary fibrosis is the most common Treprostinil is a stable analogue of prostacyclin, which Park, NC, USA (P Smith PharmD,
form.1 Clinical trials of idiopathic pulmonary fibrosis promotes vasodilation of pulmonary and systemic E Shen PharmD,
treatments have resulted in approval in many countries arterial vascular beds and inhibits platelet aggregation.8 L D Edwards PhD,
A Nelsen PharmD); Division of
of two so-called antifibrotic agents, pirfenidone and The inhaled formulation of treprostinil is approved in Pulmonary and Critical Care
nintedanib.2,3,4 Both drugs have been shown to slow loss the USA for the treatment of WHO group 1 pulmonary Medicine, Cedars-Sinai Medical
of lung function, as measured by forced vital capacity hypertension.8,9 In addition to its effects on the Center, Los Angeles, CA, USA
(FVC). Results of clinical trials for both drugs in other pulmonary vasculature, there are data to suggest that (Prof V F Tapson MD)

forms of fibrotic lung disease—including scleroderma- treprostinil has antifibrotic properties. Specifically,
associated ILD, ILDs characterised by progressive treprostinil has been shown to affect extracellular matrix

www.thelancet.com/respiratory Published online June 29, 2021 https://doi.org/10.1016/S2213-2600(21)00165-X 1


Articles

Correspondence to:
Prof Steven D Nathan, Advanced Research in context
Lung Disease and Lung
Transplant Program, Evidence before this study Added value of this study
Falls Church, VA 22042, USA Clinical trials in patients with chronic fibrosing interstitial lung In this post-hoc analysis of the INCREASE trial, inhaled
steven.nathan@inova.org diseases (ILDs) have led to the development of two antifibrotic treprostinil was associated with significant improvements in
agents, nintedanib and pirfenidone. These therapies have been the percentage of predicted FVC at week 8 and week 16.
shown to reduce the rate of decline in lung function and are Improvement in FVC was greatest among patients with
now recommended in clinical practice guidelines for patients idiopathic interstitial pneumonia, particularly those with
with idiopathic pulmonary fibrosis. The INCREASE study was a idiopathic pulmonary fibrosis. These results suggest that
randomised clinical trial of patients with ILD and pulmonary inhaled treprostinil might have independent antifibrotic
hypertension that evaluated the safety and efficacy of inhaled properties beyond traditional vasodilatory effects. Whether this
treprostinil. The study met its primary endpoint of change in the will be substantiated in a study of patients with interstitial lung
peak 6-min walk distance at week 16. Pulmonary function disease, with or without pulmonary hypertension, remains to
testing was obtained as a safety parameter in this study. be determined.
Somewhat unexpectedly, this study also showed that inhaled
Implications of all the available evidence
treprostinil was associated with improvements in forced vital
The present study serves as proof of concept that inhaled
capacity (FVC) over a 16-week period. We searched PubMed on
treprostinil can have a beneficial effect on the loss of lung
June 9, 2021, using the search terms “treprostinil” and “lung
function in patients with ILDs and associated pulmonary
function” for all articles published from database inception up to
hypertension. These findings were most pronounced in
June 9, 2021, with no language restrictions. One retrospective
patients with idiopathic pulmonary fibrosis. Further studies are
study of 22 patients with interstitial lung disease and associated
needed to investigate the clinical benefits of inhaled treprostinil
pulmonary hypertension treated with inhaled treprostinil
in this patient population, with or without associated
assessed lung function. No significant changes were observed in
pulmonary hypertension, and to explore potential mechanisms
percentage predicted FVC but this study was limited by its
of action.
observational, uncontrolled design. The effects of inhaled
treprostinil on lung function in this disease population have
otherwise not been studied.

remodelling and fibrosis in vitro by reducing recruit­ subgroups of interest, including specific disease sub­
ment of fibrocytes to sites of vascular remodelling, as groups and patients stratified by median baseline clinical
well as suppressing profibrotic fibroblast activity and characteristics.
the synthesis and deposition of collagen and fibronectin
in mice.10,11 Methods
The INCREASE study was a 16-week randomised Study design and participants
controlled trial that was designed to evaluate the safety INCREASE was a multicentre, randomised, double-blind,
and efficacy of inhaled treprostinil in patients with ILD placebo-controlled, parallel-group trial. The steering
and pulmonary hypertension documented by right heart committee in collaboration with the sponsor (United
catheterisation. Patients received inhaled treprostinil, up Therapeutics Corporation) designed the study and
to 12 breaths (72 μg) four times daily, or placebo.12 The oversaw its conduct. Detailed study procedures and
dose of drug or placebo was adjusted, with dose escalation results have been described previously.12 The study
(an additional breath, four times daily) occurring as often population included patients aged 18 years and older
as every 3 days, with a target dose of nine breaths with a diagnosis of ILD based on evidence of diffuse
four times daily and a maximum dose of 12 breaths parenchymal lung disease on chest CT done within
four times daily. Investigators adjusted the dose for each 6 months before random assignment (not centrally
individual patient to achieve the maximum tolerated adjudicated). Confirmation of WHO group 3 pulmonary
dose. The study met its primary endpoint of change in hypertension was required, based on right heart
the peak 6-min walk distance, a measure of exercise catheterisation within 1 year before random assignment
capacity, at week 16. Secondary endpoints, including (including pulmonary vascular resistance >3 Wood units,
change in N-terminal pro-brain natriuretic peptide pulmonary capillary wedge pressure ≤15 mm Hg, and
(NT-proBNP) concentration at week 16 and time to mean pulmonary arterial pressure ≥25 mm Hg). Patients
clinical worsening, were also met. Pulmonary function with connective tissue disease-associated ILD were
testing was done as a safety assessment at baseline, additionally required to have a baseline FVC <70%.
week 8, and week 16, and was previously reported.12 Patients were randomly assigned (1:1) to inhaled
The aim of this post-hoc analysis was to further treprostinil (Tyvaso; United Therapeutics Corporation,
characterise the effects of inhaled treprostinil on FVC Research Triangle Park, NC, USA) or placebo in a double-
among the overall study population and in various blind manner. No new antifibrotics or anti-inflammatory

2 www.thelancet.com/respiratory Published online June 29, 2021 https://doi.org/10.1016/S2213-2600(21)00165-X


Articles

Idiopathic interstitial Idiopathic pulmonary Combined pulmonary Connective tissue p value*


pneumonia including fibrosis only (n=92) fibrosis and emphysema disease-associated ILD
idiopathic pulmonary (n=82) (n=72)
fibrosis (n=146)
Sex ·· ·· ·· ·· <0·0001
Female 60 (41%) 31 (34%) 23 (28%) 55 (76%) ··
Male 86 (59%) 61 (66%) 59 (72%) 17 (24%) ··
Age, years 67·8 (11·2) 70·8 (9·1) 71·7 (9·8) 57·2 (11·5) ··
Age group, years ·· ·· ·· ·· <0·0001
<65 41 (28%) 15 (16%) 12 (15%) 53 (74%) ··
65 to <80 93 (64%) 67 (73%) 54 (66%) 19 (26%) ··
≥80 12 (8%) 10 (11%) 16 (20%) 0 ··

Race ·· ·· ·· ·· <0·0001
White 113 (77%) 76 (83%) 69 (84%) 34 (47%) ··
Black 24 (16%) 10 (11%) 12 (15%) 32 (44%) ··
American Indian or Alaska 2 (1%) 2 (2%) 0 1 (1%) ··
Native
Asian 6 (4%) 4 (4%) 1 (1%) 4 (6%) ··
Multiple 0 0 0 1 (1%) ··
Unknown 1 (1%) 0 0 0 ··
Ethnicity† ·· ·· ·· ·· 0·35
Hispanic or Latino 14 (10%) 10 (11%) 4 (5%) 8 (11%) ··
Not Hispanic or Latino 132 (90%) 82 (89%) 77 (95%) 64 (89%) ··
Time since diagnosis, years 0·6 (1·58) 0·5 (1·52) 0·4 (0·63) 0·6 (1·15) 0·59
Use of supplemental oxygen 102 (70%) 68 (74%) 64 (78%) 44 (61%) 0·073
Use of background therapy ·· ·· ·· ·· <0·0001
None 91 (62%) 43 (47%) 69 (84%) 69 (96%) ··
Pirfenidone only 34 (23%) 30 (33%) 6 (7%) 2 (3%) ·
Nintedanib only 21 (14%) 19 (21%) 7 (9%) 1 (1%) ·
Pulmonary function tests
Total lung capacity % 60·7 (16·1) 60·2 (16·0) 77·8 (14·7) 52·9 (13·5) <0·0001
predicted, %
Total lung capacity, L 3·6 (1·2) 3·7 (1·1) 4·8 (1·2) 2·9 (1·2) <0·0001
FVC% predicted, % 59·5 (17·2) 60·8 (17·7) 82·5 (18·8) 49·5 (13·4) <0·0001
FVC, mL 2175·7 (764·6) 2192·1 (724·6) 2992·6 (799·8) 1630·6 (552·4) <0·0001
FEV1% predicted, % 64·3 (19·0) 67·4 (18·8) 77·0 (21·1) 51·0 (15·1) <0·0001
FEV1, mL 1757·0 (585·8) 1793·1 (551·4) 2051·1 (598·6) 1320·1 (456·2) <0·0001
DLCO% predicted, % 30·0 (12·1) 29·1 (11·5) 27·6 (11·3) 28·3 (11·7) 0·35
DLCO, mL/min/mm Hg 8·2 (4·6) 8·0 (4·7) 6·9 (3·0) 7·0 (4·0) 0·058
Data are n (%) or mean (SD). DLCO=diffusing capacity for carbon monoxide. FVC=forced vital capacity. ILD=interstitial lung disease. *p value compares idiopathic interstitial
pneumonia including idiopathic pulmonary fibrosis, combined pulmonary fibrosis and emphysema, and connective tissue disease populations only. †One patient in the
placebo group had a missing response for ethnicity.

Table 1: Baseline characteristics by disease cause

therapies could be started during the study. The protocol each study site. We evaluated pulmonary function testing
was approved by the institutional review board at each for the overall study population and by subgroup based on
participating site; all participants provided written disease cause (idiopathic interstitial pneumonia [including
informed consent. idiopathic pulmonary fibrosis], idiopathic pulmonary
fibrosis alone, combined pulmonary fibrosis and
Procedures and outcomes emphysema, and connective tissue disease-associated
This post-hoc analysis assessed the results of pulmonary ILD) and subgroups stratified by median baseline
function testing, which was prespecified as a safety characteristics (percentage of predicted FVC, percentage
endpoint and done at baseline, week 8, and week 16 (or at of predicted diffusing capacity for carbon monoxide
early termination) after sufficient recovery from the 6-min [DLCO], pulmonary vascular resistance, mean pulmonary
walk tests. Pulmonary function tests were done locally at arterial pressure, NT-proBNP concentration, and 6-min

www.thelancet.com/respiratory Published online June 29, 2021 https://doi.org/10.1016/S2213-2600(21)00165-X 3


Articles

LSM difference: LSM difference: LSM difference:


A 28·5 (SE 30·1) 44·4 (SE 35·4) 108·2 (SE 46·9)
50 95% CI: 95% CI: –25·2 to 114·0 A 95% CI: 15·3 to 201·1)
–30·8 to 87·7 p=0·21 100 Treatment group p=0·023
40
LSM change from baseline in FVC (mL)

p=0·35 80 Inhaled treprostinil

LSM change from baseline in FVC (mL)


30
60 Placebo
20 N=58 N=52
N=129 40
10 N=141 20
0 0
–20
–10
–40
–20 –60 N=70
–30 N=140 –80
–40 N=124 –100 LSM difference:
Treatment group –120 46·5 (SE 39·9)
–50 –140 95% CI: –32·5 to 125·5 N=62
Inhaled treprostinil
–60 Placebo –160 p=0·25
–70 –180
–200
B B LSM difference:
4 LSM difference: 4 2·9 (SE 1·1)
LSM change from baseline in FVC% predicted (%)

LSM difference:

LSM change from baseline in FVC% predicted (%)


LSM difference: 1·8 (SE 0·8) 95% CI: 0·7 to 5·0
3 1·8 (SE 0·7) 2·0 (SE 0·9)
95% CI: 0·2 to 3·4 3 p=0·0096
95% CI: 0·4 to 3·2 95% CI: 0·1 to 3·8
2 p=0·028
p=0·014 2 p=0·037
N=52
1 N=58
N=141 N=129 1
0 0
–1 N=124 –1
N=140
N=70 N=62
–2 –2
–3 –3
–4 –4
0 8 16 0 8 16
Treatment week
Treatment week
Figure 1: Change in FVC at week 8 and week 16 for the overall population
(A) LSM change in FVC (mL) by week for the overall intention-to-treat Figure 2: Change in FVC at week 8 and week 16 for patients with idiopathic
population. (B) LSM change in percentage of predicted FVC by week for the interstitial pneumonia
overall intention-to-treat population. Bars above and below the dots represent (A) LSM change in FVC (mL) by week for the subgroup of patients with
the SE. Mixed model repeated measures methodology was used to compare idiopathic interstitial pneumonia who received treprostinil or placebo.
differences between the inhaled treprostinil and placebo group. FVC=forced vital (B) LSM change in percentage of predicted FVC by week for the subgroup of
capacity. LSM=least squares mean. patients with idiopathic interstitial pneumonia. Bars above and below the dots
represent the SE. Mixed model repeated measures methodology was used to
walk distance). An additional safety endpoint was the compare differences between the inhaled treprostinil and placebo group.
FVC=forced vital capacity. LSM=least squares mean.
incidence of acute exacerbations of disease, determined by
individual site principal investigators. Other prespecified
safety endpoints (not included in the present analysis) interaction as fixed effects, and baseline FVC as a covariate.
were adverse events, oxygenation measured by pulse An unstructured variance or covariance structure shared
oximetry and supplemental oxygen requirement, clinical across treatment groups was used to model the within-
laboratory parameters, vital signs, electrocardiograms, subject errors. Treatment differences, associated 95% CIs,
and cardiopulmonary hospitalisations.12 and p values were calculated. Change in percentage of
predicted FVC was analysed similarly using the MMRM
Statistical analysis model, with the percentage of predicted FVC as the
The analyses reported here were post-hoc and exploratory. dependent variable, with treatment, week, and treatment-
All assessments are summarised by descriptive statistics, by-week interaction as fixed effects, and baseline percentage
as appropriate. Comparisons for categorical assessments of predicted FVC as a covariate. An unstructured variance
were done by Mantel-Haenszel tests and comparisons for or covariance structure shared across treatment groups was
continuous assessments were done by Mann-Whitney used to model the within-subject errors. Changes during
tests. All analyses were done on the intention-to-treat the 16-week study were evaluated for the entire cohort and
population, defined as individuals who were randomly for disease-specific subgroups. Only observed data were
assigned and received at least one dose of study drug. The included in the analysis; no data were imputed. When
change in FVC was analysed using mixed model repeated stratifying by baseline characteristics, median values were
measures (MMRM) methodology. The MMRM model used as a cutoff (eg, below vs above median). The time to
included the change from baseline in FVC as the dependent first acute exacerbation was analysed via Kaplan-Meier
variable, with treatment, week, and treatment-by-week curves and log-rank test. p<0·05 was considered to indicate

4 www.thelancet.com/respiratory Published online June 29, 2021 https://doi.org/10.1016/S2213-2600(21)00165-X


Articles

a statistically significant difference and no adjustments LSM difference:


LSM difference:
were made for multiplicity because this was an unplanned, A 84·5 (SE 52·7) 168·5 (SE 64·5)
post-hoc analysis. Analyses were done with SAS version 95% CI: –20·4 to 189·5 95% CI: 40·1 to 297·0
110
p=0·11 p=0·011
9.4. The INCREASE study is registered with ClinicalTrials. 90

LSM change from baseline in FVC (mL)


70
gov, NCT02630316. 50 N=31 N=28
30
Role of the funding source 10
–10
In collaboration with the authors, the funder of the study
–30 N=46
participated in study design, data collection, data analysis, –50
data interpretation, and writing of the report. –70
–90
–110 Treatment group
Results –130 Inhaled treprostinil
N=41

Between Feb 3, 2017, and Aug 30, 2019, 326 patients were –150 Placebo
enrolled in the INCREASE trial. Reasons for screen –170

failures, baseline characteristics by treatment assign­ B


ment, and study discontinuations have been described 5 LSM difference:
LSM change from baseline in FVC% predicted (%)

previously.12 146 (45%) of 326 patients had idiopathic LSM difference:


2·5 (SE 1·2) 3·5 (SE 1·4)
4
interstitial pneumonia, of whom 92 (28%) had idiopathic 95% CI: 0·1 to 4·9 95% CI: 0·7 to 6·3)
3 p=0·015
pulmonary fibrosis, 37 (11%) had non-specific interstitial p=0·038
pneumonia, and 13 (4%) had unclassified idiopathic 2 N=28
interstitial pneumonia. 82 (25%) of 326 patients had N=31
1
combined pulmonary fibrosis and emphysema, and
72 (22%) patients had connective tissue disease- 0

associated ILD. 19 (6%) of 326 patients had chronic –1


N=46
hypersensitivity pneumonitis, six (2%) had occupational –2 N=41
lung disease, and one (<1%) had other idiopathic
interstitial pneumonia. Demographic details of the –3
0 8 16
four largest groups—idiopathic interstitial pneumonia Treatment week
(including idiopathic pulmonary fibrosis), idiopathic
pulmonary fibrosis alone, combined pulmonary fibrosis Figure 3: Change in FVC at week 8 and week 16 for patients with idiopathic
pulmonary fibrosis
and emphysema, and connective tissue disease- (A) LSM change in FVC (mL) by week for the subgroup of patients with
associated ILD—are shown in table 1. Notable differences idiopathic pulmonary fibrosis who received treprostinil or placebo.
included variable baseline pulmonary function tests (B) LSM change in percentage of predicted FVC by week for the subgroup of
between disease subtypes and younger age of patients patients with idiopathic pulmonary fibrosis. Bars above and below the dots
represent the SE. Mixed model repeated measures methodology was used to
with connective tissue disease-associated ILD. Anti­ compare differences between the inhaled treprostinil and placebo groups.
fibrotic therapy was most prevalent in patients with FVC=forced vital capacity. LSM=least squares mean.
idiopathic pulmonary fibrosis, with 19 (21%) of
92 patients on nintedanib and 30 (33%) patients on difference between inhaled treprostinil and placebo) least
pirfenidone. Baseline pulmonary function tests, haemo­ squares mean FVC of 28·5 mL (SE 30·1; 95% CI
dynamics, 6-min walk distance, NT-proBNP concen­ –30·8 to 87·7; p=0·35) at week 8 and 44·4 mL (35·4;
tration, and St George’s Respiratory Questionnaire data –25·2 to 114·0; p=0·21) at week 16 (figure 1A). The
by disease group and treatment assignment are shown in percentage of predicted FVC differences at week 8
the appendix (p 2). (least squares mean 1·8%; SE 0·7; 95% CI 0·4 to 3·2; See Online for appendix
In the inhaled treprostinil group, a median of ten breaths p=0·014) and week 16 (1·8%; 0·8; 0·2 to 3·4; p=0·028)
(IQR 8·5–12) and 12 breaths (9–12), corresponding to were statistically significant, indicating potential benefit of
66 μg and 72 μg per session, was achieved at week 8 and inhaled treprostinil compared with placebo (figure 1B).
week 16, respectively, with 67 (51%) of 132 patients at A subgroup analysis of patients with idiopathic interstitial
week 8 and 67 (58%) of 116 patients at week 16 achieving pneumonia showed significant FVC differences at week 16
10–12 breaths (60–72 μg). 40 (25%) of 163 patients in the (108·2 mL; SE 46·9; 95% CI 15·3 to 201·1; p=0·023)
inhaled treprostinil group and 38 (23%) of 163 patients in although not at week 8 (46·5 mL; 39·9; –32·5 to 125·5;
the placebo group discontinued study drug prematurely p=0·25; figure 2A), and significant differences in
but were encouraged to remain in the study and complete percentage of predicted FVC at week 8 (2·0%; SE 0·9;
study assessments up to week 16. 33 patients assigned 95% CI 0·1 to 3·8; p=0·037) and week 16 (2·9%; 1·1;
to inhaled treprostinil and 35 assigned to placebo 0·7 to 5·0; p=0·0096) favouring inhaled treprostinil
discontinued study participation.12 (figure 2B). Further analysis of patients with idiopathic
Inhaled treprostinil was associated with a non- pulmonary fibrosis showed non-significant FVC
significant increase in placebo-corrected (the treatment differences of 84·5 mL (SE 52·7; 95% CI –20·4 to 189·5;

www.thelancet.com/respiratory Published online June 29, 2021 https://doi.org/10.1016/S2213-2600(21)00165-X 5


Articles

concentration, and median baseline 6-min walk distance


Inhaled Placebo (n) Placebo-corrected p value
treprostinil (n) difference in week-16 (table 2). We identified no responder or non-responder
FVC, mL subgroup based on analyses of percentage of predicted
FVC% predicted FVC (<60% or ≥60%), percentage of predicted DLCO
<60 62 50 27·5 0·61 (<27% or ≥27%), or mean pulmonary arterial pressure
(53·3; –77·4 to 132·4) (<35 mm Hg or ≥35 mm Hg). However, a subgroup
≥60 67 74 59·2 0·22 analysis of 114 patients with pulmonary vascular
(47·8; –34·9 to 153·4) resistance of 5·275 Wood units or greater showed a
DLCO% predicted significant difference favouring inhaled treprostinil
<27 52 61 35·4 0·49 versus placebo, with a mean placebo-corrected difference
(51·6; –66·2 to 137·1) in FVC of 112·5 mL at 16 weeks (SE 52·6; 95% CI
≥27 70 60 33·3 0·49 9·0 to 215·9; p=0·033; table 2). Similarly, a subgroup
(48·5; –62·3 to 128·9)
analysis of 110 patients with NT-proBNP concentration of
Pulmonary vascular resistance, Wood units
503·85 pg/mL or greater showed a significant difference
<5·275 64 75 –1·6 0·97
(47·9; –95·9 to 92·8)
favouring inhaled treprostinil compared with placebo,
≥5·275 65 49 112·5 0·033
with a placebo-corrected difference in FVC of 94·4 mL at
(52·6; 9·0 to 215·9) 16 weeks (SE 47·4; 95% CI 0·7 to 188·2; p=0·048; table 2).
Mean pulmonary arterial pressure, mm Hg 14 patients in the inhaled treprostinil group were on
<35 63 63 63·4 0·21 pirfenidone versus 18 patients in the placebo group.
(50·2; –35·4 to 162·1) There was no significant placebo-corrected difference
≥35 66 61 25·6 0·61 favouring inhaled treprostinil at 16 weeks (91·5 mL;
(50·2; –73·2 to 124·4) SE 110·2; 95% CI –134·2 to 317·1; p=0·41) in the
NT-proBNP, pg/mL pirfenidone-treated subgroup. Seven patients in the
<503·85 62 75 19·9 0·71 inhaled treprostinil group were on nintedanib versus
(53·7; –86·3 to 126·1)
15 patients in the placebo group. The placebo-corrected
≥503·85 63 47 94·4 0·048
(47·4; 0·7 to 188·2)
difference for inhaled treprostinil was 113·0 mL at
6-min walk distance, m
16 weeks (SE 77·1; 95% CI –46·9 to 272·8; p=0·16) in
the nintedanib-treated subgroup. When patients on
<259 63 56 64·5 0·21
(51·7; –37·3 to 166·4) pirfenidone or nintedanib were evaluated as a group,
≥259 66 68 27·6 0·57 those receiving inhaled treprostinil had no statistically
(48·7; –68·4 to 123·5) significant difference in placebo-corrected FVC (81·5 mL;
Data are mean (SE; 95% CI) unless otherwise indicated. DLCO=diffusing capacity SE 71·0; 95% CI –60·9 to 223·8; p=0·26) or percentage of
for carbon monoxide. FVC=forced vital capacity. NT-proBNP=N-terminal predicted FVC (1·2%; 1·5; –1·8 to 4·2; p=0·43) at week 16.
pro-brain natriuretic peptide. The most frequent adverse events, which have been
Table 2: Placebo-corrected difference in week-16 FVC stratified by reported previously, included cough, headache, dyspnoea,
median baseline clinical characteristics dizziness, nausea, fatigue, and diarrhoea.12 We found no
difference in the frequency of these adverse events
according to disease subtype (table 3). We also observed
p=0·11) at week 8 and significant differences of 168·5 mL no difference in exacerbations between disease subtypes
(64·5; 40·1 to 297·0; p=0·011) at week 16 (figure 3A), and (appendix p 3).
differences in percentage of predicted FVC at week 8 and Regarding safety, no significant treatment-related
week 16 of 2·5% (SE 1·2; 95% CI 0·1 to 4·9; p=0·038) changes associated with inhaled treprostinil in pulse
and 3·5% (1·4; 0·7 to 6·3; p=0·015), respectively, oximetry or supplemental oxygen use were reported
compared with placebo (figure 3B). Of note, 49 (53%) of during the study. However, there were significantly
92 patients with idiopathic pulmonary fibrosis were on fewer lung disease exacerbations in the treprostinil
antifibrotic therapy at baseline. FVC changes for patients group than the placebo group. Specifically, 43 (26%) of
with connective tissue disease-associated ILD and 163 patients in the treatment group had an exacerbation
combined pulmonary fibrosis and emphysema are shown of underlying lung disease compared with 63 (39%) of
in the appendix (pp 4–5). Histograms showing FVC 163 patients in the placebo group (p=0·02 by Fisher’s
categorical changes in 5% increments for the overall study exact test).12 Time to investigator-reported exacerbations
population and the four disease categories are provided in in the two groups is shown in the appendix (p 8), with
the appendix (pp 6–8). inhaled treprostinil associated with a significant
We stratified change in FVC over 16 weeks by baseline treatment effect (log-rank p=0·040).
lung function (median percentage of predicted FVC and
median percentage of predicted DLCO), haemo­dynamics Discussion
(median pulmonary vascular resistance and mean The INCREASE study was designed to evaluate the
pulmonary arterial pressure), median baseline NT-proBNP effects of inhaled treprostinil in patients with various

6 www.thelancet.com/respiratory Published online June 29, 2021 https://doi.org/10.1016/S2213-2600(21)00165-X


Articles

ILDs and associated pulmonary hypertension. The study


Idiopathic interstitial Idiopathic Combined Connective tissue
met its primary endpoint: compared with placebo, pneumonia including pulmonary pulmonary fibrosis disease-associated
treprostinil improved exercise capacity, assessed by idiopathic pulmonary fibrosis only and emphysema ILD (n=72)
6-min walk distance, from baseline at 16 weeks.12 The fibrosis (n=146) (n=92) (n=82)
study also met several secondary endpoints, including Number of subjects 115 (79%) 71 (77%) 63 (77%) 49 (68%)
time to clinical worsening and change in concentration with adverse event of
interest
of NT-proBNP, a marker of cardiac dysfunction that
Cough 55 (38%) 30 (33%) 32 (39%) 31 (43%)
likely reflects right ventricular strain in patients with
Dyspnoea 44 (30%) 24 (26%) 28 (34%) 17 (24%)
pulmonary hypertension due to interstitial lung disease.
Headache 35 (24%) 19 (21%) 22 (27%) 15 (21%)
Lung function, specifically spirometry, was measured at
baseline and at week 8 and week 16 as a safety measure to Dizziness 29 (20%) 17 (18%) 15 (18%) 5 (7%)

ensure that there were no deleterious effects of inhaled Nausea 28 (19%) 18 (20%) 11 (13%) 9 (13%)
treprostinil. We felt that investigation of any deleterious Fatigue 20 (14%) 12 (13%) 13 (16%) 11 (15%)
effects was important because of the patients’ pre- Diarrhoea 23 (16%) 13 (14%) 9 (11%) 6 (8%)
existing lung disease. A noteworthy finding previously Productive cough 4 (3%) 3 (3%) 2 (2%) 5 (7%)
reported in the primary publication12 was that, rather Dyspnoea on exertion 4 (3%) 2 (2%) 2 (2%) 1 (1%)
than causing any decrement, inhaled treprostinil Upper-airway cough 3 (2%) 1 (1%) 1 (1%) 2 (3%)
syndrome
was unexpectedly associated with a placebo-corrected
Sinus headache 2 (1%) 1 (1%) 1 (1%) 0
improvement in FVC. Further subgroup analyses
revealed the FVC difference between the inhaled Muscle fatigue 1 (1%) 1 (1%) 1 (1%) 0

treprostinil group and placebo group to be larger in the Data are n (%). ILD=interstitial lung disease.
idiopathic interstitial pneumonia subgroup, and even Table 3: Adverse events by disease cause
more so in the idiopathic pulmonary fibrosis
subpopulation.
FVC is widely accepted as the standard efficacy medication approved in the USA and Europe for the
endpoint for clinical trials in idiopathic pulmonary treatment of pulmonary arterial hypertension, in patients
fibrosis and other fibrotic lung disorders.13 Indeed, with idiopathic pulmonary fibrosis on background
clinical trials evaluating change in FVC over 52 weeks nintedanib.15 This observation lends cre­ dence to the
have resulted in the approval of both pirfenidone and possibility that the pulmonary vasculature itself might be
nintedanib for the treatment of idiopathic pulmonary involved in the perpetuation of the fibrotic process or
fibrosis.3,4 Nintedanib has since been approved for other that pulmonary hypertension and fibrosis have shared
fibrotic disorders, including scleroderma-associated ILD pathways. However, this synergy was not replicated when
and a broader group of ILDs characterised by a sildenafil was combined with pirfenidone.16
progressive fibrotic phenotype.5,6 The disease groups Two of our subgroup analyses showed a significant
included in a previous cohort study6 of nintedanib were difference in FVC change between the inhaled treprostinil
very similar to those of the INCREASE study.12 Our group and the placebo group. These analyses were in
findings lend support to the study of different forms of patients with higher pulmonary vascular resistance and
fibrotic disease as a group, although the idiopathic higher NT-proBNP concentration. Therefore, pulmonary
pulmonary fibrosis subgroup did show the strongest vessels themselves might contribute to the restrictive
efficacy signal. physiology, perhaps through vascular compliance, which
No drug has been shown to have an effect on fibrosis is ameliorated by inhaled treprostinil. An alternative,
when administered via inhalation. However, there is speculative concept is that the presence of more severe
inherent attractiveness to this mode of delivery because pulmonary hypertension might hasten the progression of
of greater deposition of drug at the site of disease, fibrosis. A further hypothesis is that these FVC findings
resulting in fewer systemic side-effects and preservation, are related to improved right ventricular function or
or possibly improvement, of ventilation–perfusion cardiac output in those receiving inhaled treprostinil, as
matching. Fibrotic lung diseases, and idiopathic evidenced by the greater treatment effect in those with
pulmonary fibrosis in particular, are characterised by higher baseline pulmonary vascular resistance and
areas of vascular ablation and perivascular fibrosis, NT-proBNP concentration, but less striking differences
which raises the issue of adequate systemic drug delivery when stratifying by baseline mean pulmonary arterial
to high-value target cells, including fibroblasts and pressure. Decreased right ventricular size and function
myofibroblasts.14 The finding of a placebo-corrected and improved cardiac output in those with more severe
improvement in FVC indicates that inhaled treprostinil pulmonary hypertension could lead to improvement in
might have distinct antifibrotic properties. Indeed, respiratory muscle function and thus FVC.
animal studies and biological pathways support this It is interesting to compare the findings in our
concept.10,11 Amelioration of loss of lung function has idiopathic pulmonary fibrosis population with those of
been described previously with sildenadil, another the pivotal phase 3 studies of pirfenidone and

www.thelancet.com/respiratory Published online June 29, 2021 https://doi.org/10.1016/S2213-2600(21)00165-X 7


Articles

nintedanib.2–4,17,18 First, INCREASE was only a 16-week exacerbations was significantly less in the treprostinil
study compared with 52-week and 72-week studies for group, allaying any concerns that this inhaled medication
these other agents. Whether our results will dissipate, be might induce acute exacerbations. Therefore, it is possible
sustained, or improve up to 52 weeks is open to that inhaled treprostinil might actually ameliorate acute
speculation given how unpredictable disease behaviour exacerbations of the underlying ILD.
can be over time. The magnitude of the difference was Our analysis has its limitations. First, this was a post-
very similar with inhaled treprostinil at 16 weeks hoc analysis of a parameter that was initially intended as a
(168·5 mL) compared with the combined pirfenidone safety endpoint, and the study was not powered to detect
(148 mL per year) and nintedanib (110·9 ml per year) differences in FVC among the different subgroups in the
datasets at 52 weeks.17,18 However, in addition to the analysis. The study was of relatively short duration, with
different follow-up periods, there were varying imputation 21% of patients discontinuing prematurely before
methods and nuanced differences that limit meaningful week 16.12 The number of patients was relatively small for
comparisons between studies.18,19 The difference in the the subgroup analyses. There was no efficacy signal from
FVC change in INCREASE was partly driven by a the connective tissue disease-associated ILD group or the
numerical FVC increase in the treatment group, which combined pulmonary fibrosis and emphysema group,
contrasts with the pirfenidone and nintedanib studies, which raises questions about global antifibrotic properties
where the difference lay in the rate of decrement in both and whether our findings are relevant for only certain
groups (although there were some patients with subgroups of fibrotic disorders. Also, the relatively rapid
numerical increases in FVC).18,19 This fact raises the rate of FVC decrement in the placebo group over a short
question of whether inhaled treprostinil can ameliorate period of time was unexpected. Aside from patients with
fibrosis; this is conceivable perhaps for areas of fresh more severe disease being at higher risk of progression,
collagen deposition or through so-called switching off of an interesting hypothesis is that perhaps the pulmonary
myofibroblasts that might contribute to restrictive vasculature is mechanistically linked to the progression of
physiology through their contractile properties.20–22 An fibrosis.23 Finally, the finding of fewer acute exacerbations
alternative explanation is that treprostinil improved vessel in the inhaled treprostinil group should be regarded with
capacitance and stiffness, affecting the measured FVC. caution, since these acute exacerbations were investigator-
Although we observed a significant difference in the determined and not centrally adjudicated. The unusually
change in percentage of predicted FVC over the 16-week high rate of acute exacerbations in both study groups is
study period for the cohort as a whole, the difference was noteworthy. It is possible that patients with more advanced
not significant when evaluated FVC by change in mL. disease who have any worsening are more likely to be
This apparent discrepancy could be due to the narrower increasingly symptomatic, with the default labelling of an
CIs when FVC is expressed as a percentage of the acute exacerbation, without necessarily fulfilling all the
predicted value. Many patients with idiopathic pulmonary criteria. We cannot rule out the possibility that any inhaled
fibrosis were already on antifibrotic therapy; however, the therapy, be it placebo or treatment, might increase the
number of patients on nintedanib or pirfenidone in propensity for acute exacerbations.
combination with inhaled treprostinil was too small to In conclusion, inhaled treprostinil appears to have a
draw efficacy conclusions. The smaller numerical salutary effect on loss of lung function in patients with
difference when combining the two antifibrotics ILD and associated pulmonary hypertension. This
(81·5 mL) versus each individually (113·0 mL for patients finding, although intriguing and hypothesis generating,
on nintedanib and 91·5 mL for patients on pirfenidone) warrants further validation in a prospective, randomised,
is a function of MMRM statistical methodology. placebo-controlled study. A clinical trial of inhaled
Nonetheless, the numerical differences show the potential treprostinil is underway in patients with idiopathic
role of combination therapy for idiopathic pulmonary pulmonary fibrosis, with or without associated pul­
fibrosis and targeting the disease concomitantly via the monary hypertension (NCT04708782). This study will
systemic and inhaled routes. use a primary endpoint of change in FVC over 52 weeks
FVC was measured as a safety endpoint in the context and will support the findings of our post-hoc analysis.
of the INCREASE study. Other pulmonary function tests, Contributors
including DLCO, were also measured as safety endpoints SDN, AW, PS, ES, LDE, AN, and VFT made substantial contributions to
but did not show a difference between groups. A the conception and design of the work. SDN, AW, SR, AC, SJ, HD,
DJDLZ, SS, CK, LM-G, and VFT contributed to the acquisition of the
longer-term study of inhaled treprostinil in patients with study data. Data were analysed by SDN, AW, PS, ES, LDE, AN, and VFT.
ILD and pulmonary hypertension is ongoing and will LDE was responsible for the statistical analysis. SDN, AW, and VFT
provide information on gas exchange (NCT02633293). participated on the INCREASE steering committee during the study.
Other safety endpoints, including pulse oximetry SDN, AW, SR, AC, SJ, HD, DJDLZ, SS, CK, LM-G, PS, ES, AN, and VFT
contributed to data interpretation, and to drafting and revision of the
and change in supplemental oxygen needs, did not manuscript. SDN, ES, and LDE have accessed and verified the data.
show any deleterious effects, mitigating concerns about All authors agree to be accountable for all aspects of the work, ensuring
potential worsening of ventilation–perfusion matching. that questions related to the accuracy or integrity of any part of the work
Additionally, the incidence of investigator-reported acute are appropriately investigated and resolved. All authors have read and

8 www.thelancet.com/respiratory Published online June 29, 2021 https://doi.org/10.1016/S2213-2600(21)00165-X


Articles

approved the manuscript. The corresponding author and all co-authors 6 Flaherty KR, Wells AU, Cottin V, et al. Nintedanib in progressive
had full access to all the data in the study and had final responsibility for fibrosing interstitial lung diseases. N Engl J Med 2019;
the decision to submit for publication. 381: 1718–27.
7 Maher TM, Corte TJ, Fischer A, et al. Pirfenidone in patients with
Declaration of interests unclassifiable progressive fibrosing interstitial lung disease:
SDN has received research funding and consulting fees from United a double-blind, randomised, placebo-controlled, phase 2 trial.
Therapeutics, and consulting fees from Boehringer Ingelheim, Lancet Respir Med 2020; 8: 147–57.
Roche-Genentech, and Galapagos; he is on the speaker’s bureau for 8 Whittle BJ, Silverstein AM, Mottola DM, Clapp LH. Binding and
Boehringer Ingelheim and Roche-Genentech. AW reports grants from activity of the prostacyclin receptor (IP) agonists, treprostinil and
United Therapeutics, during the conduct of the study. SR reports iloprost, at human prostanoid receptors: treprostinil is a potent DP1
grants from United Therapeutics, during the conduct of the study; and EP2 agonist. Biochem Pharmacol 2012; 84: 68–75.
grants and personal fees from United Therapeutics and Janssen 9 McLaughlin VV, Benza RL, Rubin LJ, et al. Addition of inhaled
Pharmaceuticals; and personal fees from Altavant Sciences, Liquidia treprostinil to oral therapy for pulmonary arterial hypertension:
Technologies, Insmed, and Bayer Pharmaceuticals, outside the a randomized controlled clinical trial. J Am Coll Cardiol 2010;
submitted work. AC reports grants from United Therapeutics, during 55: 1915–22.
the conduct of the study. SJ reports grants from United Therapeutics, 10 Lambers C, Roth M, Jaksch P, et al. Treprostinil inhibits
during the conduct of the study; grants and personal fees from Bayer proliferation and extracellular matrix deposition by fibroblasts
Pharmaceuticals and Janssen Research & Development; and grants through cAMP activation. Sci Rep 2018; 8: 1087.
from Bellerophon Therapeutics, outside the submitted work. 11 Nikitopoulou I, Manitsopoulos N, Kotanidou A, et al. Orotracheal
HD reports grants from United Therapeutics, during the conduct of treprostinil administration attenuates bleomycin-induced lung
the study; and grants and personal fees from Actelion Pharmaceuticals, injury, vascular remodeling, and fibrosis in mice. Pulm Circ 2019;
9: 2045894019881954.
outside the submitted work. DJDLZ reports grants from United
Therapeutics, during the conduct of the study. SS reports grants from 12 Waxman A, Restrepo-Jaramillo R, Thenappan T, et al. Inhaled
treprostinil in pulmonary hypertension due to interstitial lung
United Therapeutics, during the conduct of the study; personal fees
disease. N Engl J Med 2021; 384: 325–34.
and non-financial support from Bayer Pharmaceuticals, United
13 Paterniti MO, Bi Y, Rekić D, Wang Y, Karimi-Shah BA,
Therapeutics, and Actelion Pharmaceuticals; personal fees from
Chowdhury BA. Acute exacerbation and decline in forced vital
Liquidia, Altavant Sciences, GSK, and Boehringer Ingelheim; capacity are associated with increased mortality in idiopathic
and grants from ACCP CHEST ILD, outside the submitted work. pulmonary fibrosis. Ann Am Thorac Soc 2017; 14: 1395–402.
CK reports grants from United Therapeutics, during the conduct of the 14 Nathan SD, Martinez FJ. Pitfalls in developing new compounds for
study; and personal fees from United Therapeutics, Actelion, idiopathic pulmonary fibrosis. Curr Opin Pulm Med 2017;
Boehringer Ingelheim, and Genentech, outside the submitted work. 23: 426–31.
LM-G reports grants and personal fees from United Therapeutics, 15 Kolb M, Raghu G, Wells AU, et al. Nintedanib plus sildenafil in
during the conduct of the study; and personal fees from United patients with idiopathic pulmonary fibrosis. N Engl J Med 2018;
Therapeutics, Janssen Pharmaceuticals, and Bayer Pharmaceuticals, 379: 1722–31.
outside the submitted work. PS, ES, LDE, and AN report personal fees 16 Behr J, Nathan SD, Wuyts WA, et al. Efficacy and safety of sildenafil
from United Therapeutics, during the conduct of the study; added to pirfenidone in patients with advanced idiopathic
and personal fees from United Therapeutics, outside the submitted pulmonary fibrosis and risk of pulmonary hypertension: a double-
work. VFT reports grants from United Therapeutics, during the blind, randomised, placebo-controlled, phase 2b trial.
conduct of the study; and personal fees from United Therapeutics, Lancet Respir Med 2021; 9: 85–95.
outside the submitted work. 17 Richeldi L, Cottin V, du Bois RM, et al. Nintedanib in patients
with idiopathic pulmonary fibrosis: Combined evidence from the
Data sharing TOMORROW and INPULSIS(®) trials. Respir Med 2016;
Qualified researchers who provide methodologically sound, genuine 113: 74–79.
research proposals can submit data requests to United Therapeutics 18 Noble PW, Albera C, Bradford WZ, et al. Pirfenidone for To submit data requests see
Corporation to obtain specific de-identified clinical trial data. idiopathic pulmonary fibrosis: analysis of pooled data from www.utcrequests.com
three multinational phase 3 trials. Eur Respir J 2016; 47: 243–53.
Acknowledgments
We thank all patients who volunteered for the study, and the clinical and 19 Flaherty KR, Kolb M, Vancheri C, Tang W, Conoscenti CS,
Richeldi L. Stability or improvement in forced vital capacity with
research teams at the INCREASE centres.
nintedanib in patients with idiopathic pulmonary fibrosis.
References Eur Respir J 2018; 52: 1702593.
1 Travis WD, Costabel U, Hansell DM, et al. An official American 20 Lehtonen ST, Veijola A, Karvonen H, et al. Pirfenidone and
Thoracic Society/European Respiratory Society statement: update of nintedanib modulate properties of fibroblasts and myofibroblasts in
the international multidisciplinary classification of the idiopathic idiopathic pulmonary fibrosis. Respir Res 2016; 17: 14.
interstitial pneumonias. Am J Respir Crit Care Med 2013; 188: 733–48. 21 Hostettler KE, Zhong J, Papakonstantinou E, et al. Anti-fibrotic
2 Noble PW, Albera C, Bradford WZ, et al. Pirfenidone in patients effects of nintedanib in lung fibroblasts derived from patients with
with idiopathic pulmonary fibrosis (CAPACITY): two randomised idiopathic pulmonary fibrosis. Respir Res 2014; 15: 157.
trials. Lancet 2011; 377: 1760–69. 22 Ackermann M, Kim YO, Wagner WL, et al. Effects of nintedanib on
3 King TE Jr, Bradford WZ, Castro-Bernardini S, et al. A phase 3 trial the microvascular architecture in a lung fibrosis model. Angiogenesis
of pirfenidone in patients with idiopathic pulmonary fibrosis. 2017; 20: 359–72.
N Engl J Med 2014; 370: 2083–92. 23 Jacob J, Nicholson AG, Wells AU, Hansell DM. Impact of
4 Richeldi L, du Bois RM, Raghu G, et al. Efficacy and safety of pulmonary vascular volume on mortality in IPF: is it time to
nintedanib in idiopathic pulmonary fibrosis. N Engl J Med 2014; reconsider the role of vasculature in disease pathogenesis and
370: 2071–82. progression? Eur Respir J 2017; 49: 1602524.
5 Distler O, Highland KB, Gahlemann M, et al. Nintedanib for
systemic sclerosis-associated interstitial lung disease. N Engl J Med
2019; 380: 2518–28.

www.thelancet.com/respiratory Published online June 29, 2021 https://doi.org/10.1016/S2213-2600(21)00165-X 9

You might also like