Guide To Clinical Management of Idiopathic Pulmonary Fibrosis
Guide To Clinical Management of Idiopathic Pulmonary Fibrosis
Guide To Clinical Management of Idiopathic Pulmonary Fibrosis
Christopher S King
Guide to Clinical
Management of
Idiopathic
Pulmonary
Fibrosis
Guide to Clinical
Management of Idiopathic
Pulmonary Fibrosis
Steven D Nathan • A Whitney Brown
Christopher S King
Guide to Clinical
Management of
Idiopathic Pulmonary
Fibrosis
Steven D Nathan Christopher S King
Inova Fairfax Medical Campus Inova Fairfax Medical Campus
Falls Church Falls Church
Virginia Virginia
USA USA
A Whitney Brown
Inova Fairfax Medical Campus
Falls Church
Virginia
USA
Steven D Nathan, MD
A Whitney Brown, MD
Christopher S King, MD
v
Contents
vii
viii Contents
5-LO 5-Lipoxygenase
6MWT 6-Minute walk test
ABCA3 Adenosine triphosphate-binding cassette
transporter A3
ACS Acute coronary syndrome
AEC Alveolar epithelial cell
AE-IPF Acute exacerbation of idiopathic pulmonary
fibrosis
AIP Acute interstitial pneumonia
ANA Anti-nuclear antibody
ARDS Adult respiratory distress syndrome
ATP Adenosine triphosphate
αvβ6 Alpha v beta 6
BNP Brain natriuretic peptide
bpm Beats per minute
CAD Coronary artery disease
CCP Cyclic citrullinated peptide
CK Creatinine kinase
COP Cryptogenic organizing pneumonia
COPD Chronic obstructive pulmonary disease
CPAP Continuous positive airway pressure
CPFE Combined pulmonary fibrosis/emphysema
CPI Composite physiologic
CTD Connective tissue disease
CTD-ILD Connective tissue disease-associated intersti-
tial lung disease
CTEPH Chronic thromboembolic pulmonary hypertension
CTGF Connective tissue growth factor
CYP Cytochrome P450
xv
xvi Abbreviations
1.1 History
Interstitial lung diseases (ILDs) represent a broad category
of diseases affecting the interstitium of the lung in a diffuse
fashion. Idiopathic pulmonary fibrosis (IPF) is a distinct sub-
type and one of the most common forms of ILD. It is a
chronic fibrosing condition that is limited to the lungs, tends
to be progressive in nature, and results in significant morbid-
ity and mortality. IPF needs to be differentiated from the
many other causes ILDs. Table 1.1 categorizes all the ILDs by
a simple mnemonic of five ‘I’s, a ‘C’, and an ‘N’ [1].
IPF was formerly referred to as cryptogenic fibrosing
alveolitis, but this term has since been abandoned. Indeed,
the definition of IPF has evolved over the past two decades
[2]. IPF used to be a ‘wastebasket’ term that encompassed
many of the idiopathic fibrosing conditions; this group of
disorders is now referred to as the idiopathic interstitial
pneumonias (IIPs) [3] (Table 1.2).
The ‘pneumonia’ or ‘pneumonitis’ aspect of this broad cat-
egory of diseases is somewhat of a misnomer; none of these
entities are infectious in etiology and many lack a significant
inflammatory component. Indeed, it used to be commonly
thought that IPF was an inflammatory disease and that the
fibrosis was the end product of chronic inflammation. This
paradigm evolved from bronchoalveolar lavage studies in the
1.3 Genetics
Familial pulmonary fibrosis (FPF) accounts for 0.5–3.7 % of
IPF cases [16] and is defined by a history of pulmonary fibro-
sis in at least two first degree relatives. The clinical presenta-
tion of FPF is similar to that of sporadic IPF, although the age
of onset tends to be younger [17], therefore FPF should be
suspected in patients who present at an earlier age.
Additionally, any family history of IPF or unknown lung dis-
ease may provide a clue to the familial variant. There are a
number of genetic variants that have been linked to FPF [16, 18–29];
each of which has a variable influence on the development of
pulmonary fibrosis. For example, within families not all per-
sons with the same mutation will develop pulmonary fibrosis,
suggesting that epigenetics are important. In other words,
there seems to be variability in the way in which an individu-
al’s genetic predisposition interacts with the environment
and other factors [18, 23, 26, 30].
In addition to the typical Mendelian inheritance pattern in
the familial variant, potential genetic factors in all afflicted
patients are being investigated. Single nucleotide polymor-
phisms (SNPs) — elucidated through gene-wide association
studies — that have been shown to be important in the pre-
dilection for IPF include:
• MUC5B — a polymorphism in the promoter region of
MUC5B, the mucin gene, has been found in approximately
34 % of patients with FPF versus 9 % of healthy controls [22, 27].
This variant of MUC5B has been found to be associated with
the development of IPF (odds ratios [OR] of developing
disease: 9.0 and 21.8 for heterozygotes and homozygotes,
respectively) [31]. It also holds prognostic information, para-
doxically conferring a twofold survival advantage to patients
with IPF who carry the polymorphism [32].
• TERT and TERC — mutations in TERC and TERT ,
the genes encoding telomerase, may also increase risk
for the development of IPF [ 29 , 33 ]. Telomeres
are regions of repetitive, non-coding nucleotide
sequences at the end of chromosomes, and telomerase
8 Guide to Clinical Management of IPF
Disease
progression Variable onset of symptoms
Diagnosis
Demise
Time
Key Points
• IPF has a typical pattern on HRCT such that lung
biopsy is usually not necessary for diagnosis.
• Other identifiable causes of ILD must be excluded.
• A very small percentage of IPF cases are familial
(defined as 2 or more first degree relatives with pul-
monary fibrosis).
• Mutations in MUC5B and certain telomerase muta-
tions appear to be associated with a higher risk of
IPF, but account for the minority of cases.
• Prognosis, although heterogeneous, is generally poor,
with an average of survival of 2.5–4 years from the
time of diagnosis.
• The first treatments for IPF, pirfenidone and ninte-
danib, were approved in 2014 and 2015, respectively.
References
1. Wallis A, Spinks K. The diagnosis and management of interstitial
lung diseases. BMJ. 2015;350:h2072.
2. Raghu G, Collard HR, Egan JJ, et al. An official ATS/ERS/JRS/
ALAT statement: idiopathic pulmonary fibrosis: evidence-based
guidelines for diagnosis and management. Am J Respir Crit Care
Med. 2011;183:788–824.
Chapter 1. Overview of Idiopathic Pulmonary Fibrosis 11
29. Armanios MY, Chen JJ, Cogan JD, et al. Telomerase mutations
in families with idiopathic pulmonary fibrosis. N Engl J Med.
2007;356:1317–26.
30. Lawson WE, Loyd JE, Degryse AL. Genetics in pulmonary
fibrosis–familial cases provide clues to the pathogenesis of idio-
pathic pulmonary fibrosis. Am J Med Sci. 2011;341:439–43.
31. Mathai SK, Yang IV, Schwarz MI, Schwartz DA. Incorporating
genetics into the identification and treatment of idiopathic pul-
monary fibrosis. BMC Med. 2015;13:191.
32. Peljto AL, Zhang Y, Fingerlin TE, et al. Association between the
MUC5B promoter polymorphism and survival in patients with
idiopathic pulmonary fibrosis. JAMA. 2013;309:2232–9.
33. Diaz de Leon A, Cronkhite JT, Yilmaz C, et al. Subclinical lung
disease, macrocytosis, and premature graying in kindreds with
telomerase (TERT) mutations. Chest. 2011;140:753–63.
34. Tsakiri KD, Cronkhite JT, Kuan PJ, et al. Adult-onset pulmonary
fibrosis caused by mutations in telomerase. Proc Natl Acad Sci
U S A. 2007;104:7552–7.
35. Diaz de Leon A, Cronkhite JT, Katzenstein AL, et al. Telomere
lengths, pulmonary fibrosis and telomerase (TERT) mutations.
PLoS One. 2010;5:e10680.
36. Young LR, Nogee LM, Barnett B, Panos RJ, Colby TV, Deutsch
GH. Usual interstitial pneumonia in an adolescent with ABCA3
mutations. Chest. 2008;134:192–5.
37. Sgalla G, Biffi A, Richeldi L. Idiopathic pulmonary fibrosis: diag-
nosis, epidemiology and natural history. Respirology. 2015.
doi:10.1111/resp.12683. [Epub ahead of print].
38. Collard HR, Moore BB, Flaherty KR, et al. Acute exacerbations
of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med.
2007;176:636–43.
39. Nathan SD, Shlobin OA, Weir N, et al. Long-term course and
prognosis of idiopathic pulmonary fibrosis in the new millen-
nium. Chest. 2011;140:221–9.
40. King C, Nathan SD. Identification and treatment of comorbidi-
ties in idiopathic pulmonary fibrosis and other fibrotic lung dis-
eases. Curr Opin Pulm Med. 2013;19:466–73.
Chapter 2
Clinical Presentation
and Diagnosis
2.1 Symptoms
Patients typically present with insidious onset of shortness of
breath. This may or may not be accompanied by a chronic dry
cough. Cough is the presenting symptom in approximately
12 % of patients [1]. Patients who are physically more active
and robust in their daily lives or who exercise regularly are
more likely to notice the onset of their symptoms sooner
compared to those patients who are more sedentary. Given
the non-specific symptoms of idiopathic pulmonary fibrosis
(IPF), patients are frequently misdiagnosed initially. Non-
specific symptoms include:
• shortness of breath;
• dry cough;
• tiredness;
• loss of appetite; and
• weight loss.
Some patients may be diagnosed while still asymptomatic,
either based on findings from physical examination or chest
imaging. An astute clinician may hear inspiratory crackles
(present in 96.8 % of symptomatic patients [1]) on routine
auscultation of the lung bases and initiate an evaluation with
imaging. Alternatively, pulmonary fibrosis may be initially
detected as an incidental finding on imaging carried out for
Sclerodactyly
Rheumatoid arthritis
-UIP Joint pains Arthritic changes-hands
-NSIP
-OP
a b
Figure 2.2 Chest X-rays showing (a), early and (b), advanced inter-
stitial changes
a UIP pattern
Subpleural, basal predominance
Reticular abnormality
Honeycombing with or without traction bronchiectasis (red arrow)
Absence of inconsistent features
a b
c d
2.8.1 Bronchoscopy
however, the absence of these does not rule out IPF. Physical
examination may yield signs that are suggestive of conditions
that may mimic IPF, specifically signs of an underlying con-
nective tissue disorder (Table 2.1). Tests that should be con-
sidered to exclude other conditions, primarily connective
tissue disorders, are shown in Table 2.2.
Radiographic imaging, in particular the HRCT, is integral
and necessary to establish the diagnosis of IPF. The HRCT
of the chest should be placed in the context of the clinical
picture. Older age, in particular, increases the pretest
28 Guide to Clinical Management of IPF
Suspected
ILD
Thorough PFTs
Identifiable
history and
cause of PF
physical exam
UIP pattern
Consider
Diagnose Not IPF
VATS
IPF
biopsy
Key Points
• IPF diagnosis requires thorough patient history to
assess for risk factors or alternative diagnoses.
• Diagnosis should include blood-work to assess for
other causes of UIP pattern (such as CTD or hyper-
sensitivity pneumonitis).
• IPF is more common in men than women and
increases with age (>65–70 years of age).
• Typical physical exam findings are dry ‘Velcro®’
crackles at lung bases +/− digital clubbing.
• Diagnosis can be made on HRCT alone if radio-
graphic diagnostic criteria are met.
• If HRCT is inconclusive, VATS lung biopsy may be
advised (if the patient is suitable for surgery).
• Bronchoscopy is not generally recommended as it is
likely to be non-diagnostic.
• The standard of care for diagnosis is a multidisci-
plinary approach with radiologic and pathologic (if
applicable) findings considered in clinical context.
References
1. Nathan SD, Shlobin OA, Weir N, et al. Long-term course and
prognosis of idiopathic pulmonary fibrosis in the new millen-
nium. Chest. 2011;140:221–9.
2. Nathan SD, Basavaraj A, Reichner C, et al. Prevalence and impact
of coronary artery disease in idiopathic pulmonary fibrosis.
Respir Med. 2010;104:1035–41.
3. Shin S, King CS, Brown AW, et al. Pulmonary artery size as a pre-
dictor of pulmonary hypertension and outcomes in patients with
chronic obstructive pulmonary disease. Respir Med. 2014;108:
1626–32.
Chapter 2. Clinical Presentation and Diagnosis 31
a b
a b
c d
Key Points
• NSIP:
• Can be idiopathic or more commonly secondary
to CTD.
• Diagnosis requires lung biopsy.
• Treatment is with immunosuppressive therapy in
most cases.
• CTD-ILD
• ILD can be the presenting symptom in some cases.
• Diagnosis can be made on clinical history and
positive serologies.
• If lung biopsy is obtained, it can show NSIP, UIP,
OP, pleuritis, or any combination thereof.
• Treatment is with immunosuppressive therapy in
most cases.
• Chronic HP
• Can occur in the absence of an identifiable
exposure.
• Chest imaging may show poorly formed small
nodules, ground-glass attenuation, peribronchio-
lar infiltrates, or areas of air trapping.
40 Guide to Clinical Management of IPF
References
1. Katzenstein AL, Fiorelli RF. Nonspecific interstitial pneumonia/
fibrosis. Histologic features and clinical significance. Am J Surg
Pathol. 1994;18:136–47.
2. Kligerman SJ, Groshong S, Brown KK, Lynch DA. Nonspecific
interstitial pneumonia: radiologic, clinical, and pathologic consid-
erations. Radiographics. 2009;29:73–87.
3. Travis WD, Costabel U, Hansell DM, et al. An official American
Thoracic Society/European Respiratory Society statement:
update of the international multidisciplinary classification of the
idiopathic interstitial pneumonias. Am J Respir Crit Care Med.
2013;188:733–48.
4. Kim EJ, Collard HR, King Jr TE. Rheumatoid arthritis-associated
interstitial lung disease: the relevance of histopathologic and
radiographic pattern. Chest. 2009;136:1397–405.
5. Gutsche M, Rosen GD, Swigris JJ. Connective tissue disease-
associated interstitial lung disease: a review. Curr Respir Care
Rep. 2012;21(1):224–32.
6. Travis WD, Hunninghake G, King Jr TE, et al. Idiopathic nonspe-
cific interstitial pneumonia: report of an American Thoracic
Society project. Am J Respir Crit Care Med. 2008;177:1338–47.
7. Latsi PI, du Bois RM, Nicholson AG, et al. Fibrotic idiopathic
interstitial pneumonia: the prognostic value of longitudinal func-
tional trends. Am J Respir Crit Care Med. 2003;168:531–7.
Chapter 3. Diseases that Mimic IPF 41
4.1 Etiology
The etiology of idiopathic pulmonary fibrosis (IPF) remains
incompletely understood. Historically, IPF was thought to be
a condition characterized by inflammation, leading to fibro-
sis. The current understanding is based on the concept of
repetitive injury with an abnormal wound healing response in
a genetically susceptible host [1]. The initial injury appears to
be to the alveolar epithelial cells (AECs), particularly type II
AECs. The current understanding of the pathogenesis of IPF
is depicted in Fig. 4.1 [2].
Alveolar
collapse and
Fibroblast re-epithelialization
Lung
Epithelium injury
Capillary
Epithelial cell Matrix
death or accumulation
Macrophage reprogramming and cross-linking
Immune Alveolus
activation and
polarization
Fibroblast
activation and
Vascular leak and myofibroblast
extravascular differentiation
coagulation
Fibrin Fibroblast
clot recruitment,
Myofibroblast
invasion,
proliferation,
and persistence
• vascular leak;
• extravascular coagulation;
• fibroblast recruitment and activation; and
• activation of the innate immune system.
4.5 Cytokines
There are multiple cytokine derangements that have been
found in patients with IPF. Presently, the role of these cyto-
kines remains investigational and has not been incorporated
into routine clinical practice. The major cytokine culprits
thought to play a pivotal role in the genesis and perpetuation
of IPF include [2]:
• transforming growth factor-beta (TGF-β);
• platelet derived growth factor (PDGF);
• connective tissue growth factor (CTGF);
• vascular endothelial growth factor (VEGF); and
• fibroblast growth factor (FGF).
Other important cytokines include:
• endothelin;
• tumor necrosis factor-alpha (TNF-α); and
• interleukin-13 (IL-13).
Chapter 4. Pathogenesis of Idiopathic Pulmonary Fibrosis 47
4.7 Pathology
Pathologic features that are important in the diagnosis of IPF
include subpleural fibrosis, microscopic honeycombing, nor-
mal lung tissue, and fibroblastic foci [13]. Fibroblasts are typi-
cally seen clumped in groups and are recognized by their
elongated spindle-like appearance. They are frequently noted
adjacent to areas of fresh collagen deposition at the interface
with normal lung. They are felt to represent the ‘leading edge’
of the injurious process.
Other pathologic features that may be seen, that are not
diagnostic features but are permissible in the context of a
lung biopsy that is otherwise diagnostic for IPF, include mild
to moderate inflammation and a few very poorly formed
granulomas. However, the presence of multiple granulomas
always should be an alert to the possible diagnosis of chronic
hypersensitivity pneumonitis (HP).
Typical histopathologic changes of IPF (usual interstitial
pneumonia pattern) are shown in Fig. 4.2.
Pathologic features suggestive of a diagnosis other than
IPF include [14]:
• multiple poorly formed granulomas — suggestive of
chronic HP, especially if they are seen in a bronchiolocen-
tric distribution;
• lymphoid aggregates — suggestive of an underlying con-
nective tissue disease (CTD), especially rheumatoid arthri-
tis or Sjögren’s syndrome (especially if extensive);
Chapter 4. Pathogenesis of Idiopathic Pulmonary Fibrosis 49
a b
c d
Key Points
• The pathogenesis of IPF is not well elucidated
at present.
• IPF is thought to arise from initial repetitive injury to
AECs and an abnormal wound healing process.
• Fibroblasts appear to be central to the propagation
of disease when they fail to be ‘turned off’ and pro-
liferate in a disinhibited manner.
• Persistently activated fibroblasts deposit an
increased amount of collagen, which provides a scaf-
fold for fibrosis.
• Cytokine imbalances have been found in patients
with IPF, but the clinical relevance has yet to be proven.
• Telomere shortening, increased LOXL2 expression,
and re-activation of the Wnt pathway are factors
linked to IPF development.
References
1. Borensztajn K, Crestani B, Kolb M. Idiopathic pulmonary fibro-
sis: from epithelial injury to biomarkers–insights from the bench
side. Respiration. 2013;86:441–52.
2. Ahluwalia N, Shea BS, Tager AM. New therapeutic targets in
idiopathic pulmonary fibrosis. Aiming to rein in runaway
wound-healing responses. Am J Respir Crit Care Med. 2014;
190:867–78.
3. Günther A, Korfei M, Mahavadi P, von der Beck D, Ruppert C,
Markart P. Unravelling the progressive pathophysiology of idio-
pathic pulmonary fibrosis. Eur Respir Rev. 2012;21:152–60.
4. Phillips RJ, Burdick MD, Hong K, et al. Circulating fibrocytes
traffic to the lungs in response to CXCL12 and mediate fibrosis.
J Clin Invest. 2004;114:438–46.
5. Marmai C, Sutherland RE, Kim KK, et al. Alveolar epithelial
cells express mesenchymal proteins in patients with idiopathic
pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol.
2011;301:L71–8.
Chapter 4. Pathogenesis of Idiopathic Pulmonary Fibrosis 51
5.1 Prognosis
The prognosis of idiopathic pulmonary fibrosis (IPF) is gener-
ally regarded as quite poor with a median survival from the
time of diagnosis of anywhere from 2.5 to 4 years [1]. However,
it is difficult a priori to predict the course of the disease in
individual patients [2]. There are some patients who have a
protracted course and survive 5 years or more, but this disease
phenotype can only be recognized in retrospect. This repre-
sents about 20–25 % of all patients with IPF [3].
5.3.2.1 Distance
5.3.2.2 Desaturation
A
PA
5.3.6 Hospitalization
5.3.7 Biomarkers
Key Points
• The disease progression of IPF is highly variable.
• Lower FVC, DLCO, 6MWT distance, and nadir SpO2
during 6MWT portend decreased survival rates.
• Disease progression is monitored through evaluation
of PFT and 6MWT.
• At this time there are no widely available blood bio-
markers for IPF.
• Presence of PH and need for hospitalization increase
the risk of mortality or lung transplantation.
• Common causes of acute worsening in IPF include
pulmonary embolism, systolic or diastolic heart fail-
ure, cardiac ischemia, or infection.
References
1. Brown AW, Shlobin OA, Weir N, et al. Dynamic patient counsel-
ing: a novel concept in idiopathic pulmonary fibrosis. Chest.
2012;142:1005–10.
2. Nathan SD, Shlobin OA, Weir N, et al. Long-term course and
prognosis of idiopathic pulmonary fibrosis in the new millen-
nium. Chest. 2011;140:221–9.
3. Brown AW, Shlobin OA, Weir N, Albano MC, Ahmad S, Smith M,
Leslie K, Nathan SD. Dynamic patient counseling: a novel con-
cept in idiopathic pulmonary fibrosis. Chest. 2012;142:
1005–10.
4. Ley B, Collard HR, King Jr TE. Clinical course and prediction of
survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care
Med. 2011;183:431–40.
5. Erbes R, Schaberg T, Loddenkemper R. Lung function tests in
patients with idiopathic pulmonary fibrosis. Are they helpful for
predicting outcome? Chest. 1997;111:51–7.
6. Zappala CJ, Latsi PI, Nicholson AG, et al. Marginal decline in
forced vital capacity is associated with a poor outcome in idio-
pathic pulmonary fibrosis. Eur Respir J. 2010;35:830–6.
7. Nathan SD, Meyer KC. IPF clinical trial design and endpoints.
Curr Opin Pulm Med. 2014;20:463–71.
Chapter 5. Prognosis, Clinical Course, and Monitoring 65
6.2 Cardiovascular
6.3 Pulmonary
6.3.1 Pulmonary Hypertension
Fibrosis
Cytokines Neovascularization COPD
Vasculopathy
Ventilation/ ↓ Vascular
perfusion mismatch capacitance Diastolic
dysfunction
Shear stress Hypoxic
vasoconstriction
Coronary artery
Acute and chronic disease
PVOD
pulmonary emboli
IPF associated
pulmonary
hypertension
6.5 Complications
6.5.1 Acute Exacerbations of Idiopathic
Pulmonary Fibrosis
The annual incidence of acute exacerbations of IPF (AE-IPF)
ranges from 1 to 20 % of patients with IPF, depending on the
population studied and the definition used [23]. AE-IPF
accounts for more than 50 % of hospital admissions and up to
40 % of all deaths [23]. The classic definition of an acute exac-
erbation includes [25]:
• the subacute onset of shortness of breath; accompanied by
• new alveolar infiltrates on the background of radiographic
usual interstitial pneumonia; with
• other potential etiologies such as heart failure, pneumonia,
or pulmonary embolism excluded.
74 Guide to Clinical Management of IPF
6.5.2 Pneumothorax
6.5.3 Aspergilloma
Key Points
• Comorbidities are common and contribute to the
morbidity and mortality of IPF.
• GERD may be occult and has an association with
IPF, perhaps as a cause of lung injury.
• Patients with IPF have a higher incidence of pulmonary
embolism and lung cancer than age-matched controls.
• CAD and heart failure are common in this aging
population.
• Pulmonary hypertension may develop and is associ-
ated with worse outcomes.
• Depression and anxiety are prevalent and likely
related to impaired quality of life and poor prognosis.
• AEs generally do not respond well to treatment and
result in high mortality (50–90 %).
Chapter 6. Comorbidities and Complications 77
References
1. Raghu G, Freudenberger TD, Yang S, et al. High prevalence of
abnormal acid gastro-oesophageal reflux in idiopathic pulmo-
nary fibrosis. Eur Respir J. 2006;27:136–42.
2. Tobin RW, Pope 2nd CE, Pellegrini CA, Emond MJ, Sillery J,
Raghu G. Increased prevalence of gastroesophageal reflux in
patients with idiopathic pulmonary fibrosis. Am J Respir Crit
Care Med. 1998;158:1804–8.
3. Lee JS, Ryu JH, Elicker BM, et al. Gastroesophageal reflux
therapy is associated with longer survival in patients with idio-
pathic pulmonary fibrosis. Am J Respir Crit Care Med. 2011;
184:1390–4.
4. Hoppo T, Jarido V, Pennathur A, et al. Antireflux surgery pre-
serves lung function in patients with gastroesophageal reflux
disease and end-stage lung disease before and after lung trans-
plantation. Arch Surg. 2011;146:1041–7.
5. Raghu G, Yang ST, Spada C, Hayes J, Pellegrini CA. Sole treat-
ment of acid gastroesophageal reflux in idiopathic pulmonary
fibrosis: a case series. Chest. 2006;129:794–800.
6. Raghu G, Rochwerg B, Zhang Y, et al. An official ATS/ERS/JRS/
ALAT clinical practice guideline: treatment of idiopathic pulmo-
nary fibrosis. An update of the 2011 clinical practice guideline.
Am J Respir Crit Care Med. 2015;192:e3–19.
7. King C, Nathan SD. Identification and treatment of comorbidi-
ties in idiopathic pulmonary fibrosis and other fibrotic lung dis-
eases. Curr Opin Pulm Med. 2013;19:466–73.
8. Nathan SD, Weir N, Shlobin OA, et al. The value of computed
tomography scanning for the detection of coronary artery dis-
ease in patients with idiopathic pulmonary fibrosis. Respirology.
2011;16:481–6.
9. Nathan SD, Basavaraj A, Reichner C, et al. Prevalence and
impact of coronary artery disease in idiopathic pulmonary fibro-
sis. Respir Med. 2010;104:1035–41.
10. Panos RJ, Mortenson RL, Niccoli SA, King Jr TE. Clinical dete-
rioration in patients with idiopathic pulmonary fibrosis: causes
and assessment. Am J Med. 1990;88:396–404.
11. Nathan SD, Shlobin OA, Ahmad S, Urbanek S, Barnett
SD. Pulmonary hypertension and pulmonary function testing in
idiopathic pulmonary fibrosis. Chest. 2007;131:657–63.
78 Guide to Clinical Management of IPF
7.1 Pirfenidone
Pirfenidone is an orally bioavailable synthetic compound that
has anti-fibrotic, anti-inflammatory, and antioxidant effects.
7.1.1 History
While the first study was positive, the other failed to show
a difference in FVC between the treatment and placebo
groups. Although the combined data was positive, the US
Food and Drug Administration (FDA) initially elected
not to approve the drug, while the European Medicines
Agency (EMA) did issue an approval. A third ‘tiebreaker’
randomized controlled trial (RCT), the ASCEND study
(NCT01366209), was therefore undertaken in the US. The
52-week trial again studied change in the FVC as the
primary endpoint [4]. ASCEND was a positive study and
resulted in the approval of pirfenidone in October of 2014.
The data from the three pirfenidone Phase III RCTs are
shown in Table 7.1.
7.1.3 Dose
Taniguchi 2010 275 Phase III IPF per ATS/ERS Vital capacity Progression free
et al. [2] double-blind, guideline decline at survival (p = 0.028)
placebo- Age 20–75 years week 52:
controlled O2 desaturation ≥5 % Placebo
on 6MET (−0.16 L)
SpO2 ≥ 85 % during High dose
6MET (−0.09 L)
p = 0.042
CAPACITY 2011 435 Phase III Definite IPF by CT or Change in % Progression free
study double-blind, biopsy proven predicted FVC survival (p = 0.023)
004 [3] placebo- Age 40–80 years at week 72:
controlled FVC ≥50 % Placebo
DLco ≥35 % (−12.4 %)
Either FVC or DLco High dose
≤90 % (−8.0 %)
6MWT ≥150 m p = 0.001
Treatment of Idiopathic Pulmonary Fibrosis
(continued)
83
Table 7.1 (continued)
84
Number Primary
Study Year of patients Study design Inclusion criteria outcome Secondary outcomes
CAPACITY 2011 344 Phase III Definite IPF by CT or Change in % Mean change in
study 006 [3] double-blind, biopsy proven predicted FVC 6MWT distance:
placebo- Age 40–80 years at week 72: Placebo (−76.9 m)
controlled FVC ≥50 % Placebo Pirfenidone
DLco ≥35 % (−9.6 %) (−45.1 m)
Either FVC or DLco Pirfenidone p = 0.0009
≤90 % (−9.0 %)
6MWT ≥150 m p = 0.501
ASCEND 2014 555 Phase III Definite IPF by CT or Change in % 6MWT change at
[4] double-blind, biopsy proven predicted FVC week 52:
placebo- Age 40–80 years at week 52: p = 0.04
controlled FVC 50–90 % p < 0.001 Progression free
Guide to Clinical Management of IPF
7.1.4 Pharmacokinetics
7.1.5 Side-Effects
7.1.6 Administration
7.2 Nintedanib
7.2.1 History
7.2.3 Dose
7.2.4 Pharmacokinetics
7.2.5 Side-Effects
7.2.6 Administration
(continued)
Table 7.3 (continued)
90
Number
of Study Inclusion
Study Year patients design criteria Primary outcomes Secondary outcomes
INPULSIS-2 2014 548 Phase III IPF Annual rate of Time to acute
[7] double- adjudicated decline in FVC: exacerbation:
blind, prior to Placebo HR = 0.38
placebo- enrollment (−207.3 mL/year) (p = 0.005)
controlled FVC ≥50 % Nintedanib
predicted (−113.6 mL/year)
DLco 30–79 % p < 0.001
predicted
ATS American Thoracic Society, DLco diffusing capacity of the lungs for carbon monoxide, ERS European Respiratory
Society, FVC forced vital capacity, HR hazard ratio, IPF idiopathic pulmonary fibrosis, L liters, PaO2 partial pressure
arterial oxygen
Guide to Clinical Management of IPF
Chapter 7. Treatment of Idiopathic Pulmonary Fibrosis 91
Since both agents only slow the rate of loss of lung function, a
cogent argument can be made for starting the drug as soon as
possible after the diagnosis, even in the context of patients who
are asymptomatic and/or who have normal lung function.
In the clinical trials of both agents, patients with FVC
50–90 % with DLco >30 % were studied [2–4, 7, 8], although
therapy may be beneficial outside of that lung function range.
92 Guide to Clinical Management of IPF
7.4.2 Warfarin
7.4.3 Ambrisentan
• Beware that more harm than good could result from treat-
ment with pulmonary vasodilator therapy.
– Worsening ventilation perfusion mismatching with
worsening oxygenation.
– Pulmonary veno-occlusive-like lesions have been
described in IPF [17]. Pulmonary veno-occlusive dis-
ease is a contraindication for PH therapies as they
may precipitate pulmonary edema.
– Occult heart failure, specifically heart failure with
preserved ejection fraction is a common comorbidity
(9–16 %) in IPF [18, 19]. This may be worsened by
PH therapies.
• Further prospective randomized, placebo-controlled stud-
ies of pulmonary vasodilator therapy in IPF are warranted
and ongoing.
Key Points
• Two anti-fibrotic agents are currently approved to
treat IPF; nintedanib and pirfenidone.
• These drugs slow down rate of loss of lung function,
but do not reverse or cure disease.
• The older treatment paradigm of treating inflam-
mation with the combination of azathioprine, corti-
costeroids, and N-acetylcysteine has been shown to
be harmful.
• Several drugs are in development and future thera-
pies are likely to be combination in nature.
• There are no approved agents to treat PH associated
with IPF, but this is a target of current clinical trials.
References
1. Azuma A, Nukiwa T, Tsuboi E, et al. Double-blind, placebo-
controlled trial of pirfenidone in patients with idiopathic pulmo-
nary fibrosis. Am J Respir Crit Care Med. 2005;171:1040–7.
96 Guide to Clinical Management of IPF
15. Raghu G, Behr J, Brown KK, Egan JJ, et al. Treatment of idio-
pathic pulmonary fibrosis with ambrisentan: a parallel, random-
ized trial. Ann Intern Med. 2013;158:641–9.
16. Tzouvelekis A, Bonella F, Spagnolo P. Update on therapeutic
management of idiopathic pulmonary fibrosis. Ther Clin Risk
Manag. 2015;11:359–70.
17. Sherner J, Collen J, King CS, Nathan SD. Pulmonary hyperten-
sion in idiopathic pulmonary fibrosis: epidemiology, diagnosis,
and therapeutic implications. Curr Respir Care Rep.
2012;1:233–42.
18. Nathan SD, Shlobin OA, Ahmad S, Urbanek S, Barnett SD.
Pulmonary hypertension and pulmonary function testing in idio-
pathic pulmonary fibrosis. Chest. 2007;131:657–63.
19. Raghu G, Nathan SD, Behr J, et al. Pulmonary hypertension in
idiopathic pulmonary fibrosis with mild-to-moderate restriction.
Eur Respir J. 2015;46:1370–7.
Chapter 8
Non-pharmacologic
Management of Idiopathic
Pulmonary Fibrosis
a c
Figure 8.1 Higher flow oxygen devices. (a) The oxymizer pendant,
(b) the oxymizer ‘moustache’, (c) high humidity high flow nasal can-
nula system with capacity to control liter flow and FiO2 of oxygen
IPF
Breathlessness
Deconditioning Exercise
Educate
Easy fatigue
Empower
Improve QOL
Social isolation
Recondition
Anxiety
Depression
↓ Emotional
well-being Pulmonary rehabilitation
Number 6MWT
First author, of change
year patients Design Population Duration (meters) Notes
Jastrzebski et al. 31 Prospective ILD (67.7 % 6 weeks NR Improved QOL and
(2006) [5] IPF) dyspnea
Nishiyama et al. 28 RCT IPF 10 weeks 46.3a Improved QOL
(2008) [6]
Holland et al. 57 RCT ILD (59.6 % 8 weeks 35a Improved exercise
(2008) [7] IPF) capacity and
symptoms
Ferreira et al. 99 Retrospective ILD (~50 % 6–8 weeks 56a Improved dyspnea
(2009) [8] IPF)
Guide to Clinical Management of IPF
hospitalization if MRC
dyspnea 2 or 3
Swigris et al. 21 Prospective IPF 6 weeks 61.6a Improved fatigue
(2011) [13]
Huppman et al. 402 Observational ILD (50 % 4 weeks 46a Improved QOL
(2013) [14] IPF)
Jackson et al. 21 RCT IPF 3 month No Increase in exercise
(2014) [15] change time
Vainshelboim 32 RCT IPF 12 weeks 81a Improved dyspnea and
et al. (2014) [16] QOL
6MWT 6 min walk test, COPD chronic obstructive pulmonary disease, ILD interstitial lung disease, IPF idiopathic pulmo-
Non-pharmacologic Management
nary fibrosis, MRC medical research council, NR not reported, QOL quality of life, RCT randomized controlled trial
a
Denotes statistically significant results
103
104 Guide to Clinical Management of IPF
Table 8.2 Guidelines for referral and listing of patients with idio-
pathic pulmonary fibrosis for lung transplantation
Guidelines for referral of patients with idiopathic pulmonary
fibrosis for lung transplant evaluation
• Histopathologic or radiographic evidence of usual interstitial
pneumonitis irrespective of lung function
• Abnormal lung function:
◦ FVC <80 % predicted or DLco <40 % predicted
• Any dyspnea or functional limitation attributable to lung
disease
• Any oxygen requirement, even if only during exertion
Guidelines for listing of patients with idiopathic pulmonary
fibrosis for lung transplantation
• 10 % or greater drop in FVC during 6 months of follow-up
• 15 % or greater drop in DLco during 6 months of follow-up
• Desaturation to SpO2 < 88 % or distance <250 m on 6MWT or
>50 m decline in 6 min walk distance over a 6 month period
• Pulmonary hypertension on right heart catheterization or
echocardiogram
• Hospitalization due to respiratory decline, pneumothorax, or
acute exacerbation
Adapted from Weill et al. [18]
6MWT 6 min walking test, DLCO diffusing capacity of the lungs for
carbon monoxide, FVC forced vital capacity, SpO2 arterial oxygen
saturation
lung transplants have been performed for IPF, with the most
recent statistics of 60.2 % versus 39.8 %, respectively [19].
Both types of lung transplant have positives and negatives
and which procedure is optimal for patients with IPF remains
controversial in the context of individual demographics,
108 Guide to Clinical Management of IPF
Key Points
• Oxygen therapy can improve symptoms and quality
of life for patients with IPF who experience rest,
nocturnal, or exertional desaturation, especially
towards later stages of disease.
• As disease progresses, symptoms can lead to a
vicious cycle of deconditioning that has downstream
consequences of increased dyspnea, easy fatigue,
psychological imbalances (eg, anxiety and depres-
sion), skeletal muscle deconditioning, social isola-
tion, and reduced emotional well-being.
• Pulmonary rehabilitation is a supervised exercise
program designed to reverse this cycle of events and
improve symptoms, whilst providing education and
psychological support.
• Palliative care is often ignored or considered late in
disease progression, ideally palliation of symptoms
should be considered early on.
• Palliative care or hospice consultation should be con-
sidered longitudinally as quality of life and prognosis
are discussed and re-visited over time.
• Lung transplantation may improve quality and quan-
tity of life in selected patients with little to no
comorbidities.
• Early referral for lung transplantation (even at time
of diagnosis) is advisable given the unpredictable
nature of disease.
Chapter 8. Non-pharmacologic Management 109
References
1. Hallstrand TS, Boitano LJ, Johnson WC, Spada CA, Hayes JG,
Raghu G. The timed walk test as a measure of severity and survival
in idiopathic pulmonary fibrosis. Eur Respir J. 2005;25:96–103.
2. Morrison DA, Stovall JR. Increased exercise capacity in hypox-
emic patients after long-term oxygen therapy. Chest.
1992;102:542–50.
3. Ryerson CJ, Berkeley J, Carrieri-Kohlman VL, Pantilat SZ,
Landefeld CS, Collard HR. Depression and functional status are
strongly associated with dyspnea in interstitial lung disease.
Chest. 2011;139:609–16.
4. Swigris JJ, Brown KK, Make BJ, Wamboldt FS. Pulmonary reha-
bilitation in idiopathic pulmonary fibrosis: a call for continued
investigation. Respir Med. 2008;102:1675–80.
5. Jastrzebski D, Gumola A, Gawlik R, Kozielski J. Dyspnea and
quality of life in patients with pulmonary fibrosis after six weeks
of respiratory rehabilitation. J Physiol Pharmacol. 2006;57 Suppl
4:139–48.
6. Nishiyama O, Kondoh Y, Kimura T, et al. Effects of pulmonary
rehabilitation in patients with idiopathic pulmonary fibrosis.
Respirology. 2008;13:394–9.
7. Holland AE, Hill CJ, Conron M, Munro P, McDonald CF. Short
term improvement in exercise capacity and symptoms following
exercise training in interstitial lung disease. Thorax. 2008;63:549–54.
8. Ferreira A, Garvey C, Connors GL, et al. Pulmonary rehabilita-
tion in interstitial lung disease: benefits and predictors of
response. Chest. 2009;135:442–7.
9. Ozalevli S, Karaali HK, Ilgin D, Ucan ES. Effect of home-based
pulmonary rehabilitation in patients with idiopathic pulmonary
fibrosis. Multidiscip Respir Med. 2010;5:31–7.
10. Rammaert B, Leroy S, Cavestri B, Wallaert B, Grosbois JM.
Home-based pulmonary rehabilitation in idiopathic pulmonary
fibrosis. Rev Mal Respir. 2011;28:e52–7.
11. Kozu R, Senjyu H, Jenkins SC, Mukae H, Sakamoto N, Kohno S.
Differences in response to pulmonary rehabilitation in idio-
pathic pulmonary fibrosis and chronic obstructive pulmonary
disease. Respiration. 2011;81:196–205.
12. Kozu R, Jenkins S, Senjyu H. Effect of disability level on
response to pulmonary rehabilitation in patients with idiopathic
pulmonary fibrosis. Respirology. 2011;16:1196–202.
110 Guide to Clinical Management of IPF
Typically, any drug that meets the bar for regulatory approval
has to go through three phases of testing:
• Phase I trials in healthy volunteers (primarily for safety
and pharmacokinetics);
• Phase II trials in patients with the disease for further
safety, efficacy, and dosing evaluation; and
• Phase III trials that are the pivotal studies to demonstrate
efficacy prior to approval.
Currently, there are no novel IPF drugs that are in Phase
III clinical trials. Therefore, it is likely that it will be a few
years before any additional novel agents are approved
for IPF. However, there are some agents that are avail-
able and approved for other conditions that are of inter-
est as potential therapies for IPF. Ultimately, it is likely
that management of this complex disorder will include
multimodality therapy targeting different pathways and
consequences of the disease. The sections below outline
some of the therapies being studied as possible treat-
ments for IPF.
9.1.1.1 Cotrimoxazole
9.1.2.2 Azithromycin
9.1.3.3 Omeprazole
9.1.4.4 Riociguat
9.1.5.5 Sirolimus
9.1.6.6 Rituximab
9.1.1.1 FG-3019
9.1.3.3 BMS-986020
9.1.4.4 SAR156597
9.1.5.5 PRM-151
9.1.6.6 BG00011
9.1.7.7 Lebrikizumab
9.2.3 Cryobiopsy
9.2.6 Biomarkers
9.3 Conclusion
Considerable advancements have been made in recent years
in the treatment of IPF. Improved understanding of the patho-
genesis of the disease has led to the development of two phar-
macologic treatments for IPF with demonstrated efficacy in
RCTs. Despite this huge leap forward, IPF remains a deadly
disease and there is much work to be done. It is hoped that
continued research into the genetic basis and pathophysio-
logic mechanisms of IPF will lead to improved techniques for
earlier diagnosis and more effective treatment. Until that
time, therapy will rely on the use of currently approved medi-
cations in combination with supportive care including oxygen,
pulmonary rehabilitation, symptom and comorbidity manage-
ment, and lung transplantation when appropriate.
Key Points
• Agents that are approved for other conditions are cur-
rently being explored as potential therapies for IPF.
• There are several novel treatments in the early drug
development stages for IPF.
• The future therapeutic paradigm is likely one of
combination drug therapy.
• There are no drugs approved for PH in any IIP, how-
ever, riociguat is currently in Phase II studies for
comorbid PH and IPF.
• Future developments in pharmacogenomics might
enable the course of disease in individual patients to
be determined, biomarker development (therefore
non-invasive diagnosis of IPF), and tailored thera-
pies for IPF and other forms of IIP.
120 Guide to Clinical Management of IPF
References
1. Han MK, Zhou Y, Murray S, et al. Lung microbiome and disease
progression in idiopathic pulmonary fibrosis: an analysis of the
COMET study. Lancet Respir Med. 2014;2:548–56.
2. Varney VA, Parnell HM, Salisbury DT, Ratnatheepan S,
Tayar RB. A double blind randomised placebo controlled pilot
study of oral co-trimoxazole in advanced fibrotic lung disease.
Pulm Pharmacol Ther. 2008;21:178–87.
3. Shulgina L, Cahn AP, Chilvers ER, et al. Treating idiopathic pul-
monary fibrosis with the addition of co-trimoxazole: a ran-
domised controlled trial. Thorax. 2013;68:155–62.
4. Fundación Pública Andaluza para la gestión de la Investigación
en Sevilla; Junta de Andalucia. Pilot study Phase III to evaluate
the efficacy and safety of trimethoprim-sulfamethoxazole in the
treatment of idiopathic pulmonary fibrosis. In: ClinicalTrials.gov
[Internet]. Bethesda (MD): National Library of Medicine (US).
2000- [cited 18 Feb 2016]. Available from: https://www.clinicaltri-
als.gov/show/NCT01777737.
5. Wuyts WA, Willems S, Vos R, Vanaudenaerde BM, et al.
Azithromycin reduces pulmonary fibrosis in a bleomycin mouse
model. Exp Lung Res. 2010;36:602–14.
6. University Hospital Inselspital Berne. Azithromycin for the
treatment of cough in idiopathic pulmonary fibrosis- a clinical
trial. In: ClinicalTrials.gov [Internet]. Bethesda: National Library
of Medicine (US). 2000- [cited 18 Feb 2016 ]. Available from:
https://clinicaltrials.gov/show/NCT02173145.
7. Lee JS, Ryu JH, Elicker BM, et al. Gastroesophageal reflux
therapy is associated with longer survival in patients with idio-
pathic pulmonary fibrosis. Am J Respir Crit Care Med.
2011;184:1390–4.
8. Lee JS, Collard HR, Anstrom KJ, et al. Anti-acid treatment and
disease progression in idiopathic pulmonary fibrosis: an analysis
of data from three randomised controlled trials. Lancet Respir
Med. 2013;1:369–76.
9. Newcastle-upon-Tyne Hospitals NHS Trust. A randomised,
placebo-controlled trial of omeprazole in idiopathic pulmo-
nary fibrosis (IPF). In: ClinicalTrials.gov [Internet].
Bethesda: National Library of Medicine (US). 2000- [cited
18 Feb 2016]. Available from: https://clinicaltrials.gov/show/
NCT02085018.
Chapter 9. The Future for Idiopathic Pulmonary Fibrosis 121