Download as pdf or txt
Download as pdf or txt
You are on page 1of 69

Interstitial Lung Disease 1st Edition

Edition Harold R Collard And Luca


Richeldi (Auth.) - eBook PDF
Go to download the full and correct content document:
https://ebooksecure.com/download/interstitial-lung-disease-ebook-pdf/
More products digital (pdf, epub, mobi) instant
download maybe you interests ...

3D Lung Models for Regenerating Lung Tissue 1st Edition


- eBook PDF

https://ebooksecure.com/download/3d-lung-models-for-regenerating-
lung-tissue-ebook-pdf/

Developing the Digital Lung: From First Lung CT to


Clinical AI 1st Edition - eBook PDF

https://ebooksecure.com/download/developing-the-digital-lung-
from-first-lung-ct-to-clinical-ai-ebook-pdf/

Developing the Digital Lung: From First Lung CT to


Clinical AI 1st Edition John D. Newell - eBook PDF

https://ebooksecure.com/download/developing-the-digital-lung-
from-first-lung-ct-to-clinical-ai-ebook-pdf-2/

Progress in Heterocyclic Chemistry Volume 29 1st


Edition - eBook PDF

https://ebooksecure.com/download/progress-in-heterocyclic-
chemistry-ebook-pdf/
Microalgae in Health and Disease Prevention 1st Edition
- eBook PDF

https://ebooksecure.com/download/microalgae-in-health-and-
disease-prevention-ebook-pdf/

Dry Eye Disease 1st Edition - eBook PDF

https://ebooksecure.com/download/dry-eye-disease-ebook-pdf/

Chronic Coronary Artery Disease: A Companion to


Braunwald's Heart Disease 1st Edition- eBook PDF

https://ebooksecure.com/download/chronic-coronary-artery-disease-
a-companion-to-braunwalds-heart-disease-ebook-pdf/

The Newborn Lung: Neonatology Questions and


Controversies 3rd Edition Eduardo Bancalari - eBook PDF

https://ebooksecure.com/download/the-newborn-lung-neonatology-
questions-and-controversies-ebook-pdf/

Epigenetics in Cardiovascular Disease 1st edition -


eBook PDF

https://ebooksecure.com/download/epigenetics-in-cardiovascular-
disease-ebook-pdf/
Interstitial Lung
Disease
HAROLD R. COLLARD, MD
Division of Pulmonary and Critical Care Medicine
Department of Medicine
University of California San Francisco
San Francisco, CA, United States

LUCA RICHELDI, MD, PhD


Division of Pulmonary Medicine
Agostino Gemelli University Hospital
Catholic University of the Sacred Heart
Rome, Italy
1600 John F. Kennedy Blvd.
Ste 1800
Philadelphia, PA 19103-2899

INTERSTITIAL LUNG DISEASE ISBN: 978-0-323-48024-6


Copyright © 2018 by Elsevier, Inc. All rights reserved.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic
or mechanical, including photocopying, recording, or any information storage and retrieval system,
without permission in writing from the publisher. Details on how to seek permission, further infor-
mation about the Publisher’s permissions policies and our arrangements with organizations such
as the Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website:
www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the Publisher
(other than as may be noted herein).

Notices

Knowledge and best practice in this field are constantly changing. As new research and experience
broaden our understanding, changes in research methods, professional practices, or medical treat-
ment may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evalu-
ating and using any information, methods, compounds, or experiments described herein. In using
such information or methods they should be mindful of their own safety and the safety of others,
including parties for whom they have a professional responsibility.
With respect to any drug or pharmaceutical products identified, readers are advised to check the
most current information provided (i) on procedures featured or (ii) by the manufacturer of each
product to be administered, to verify the recommended dose or formula, the method and duration
of administration, and contraindications. It is the responsibility of practitioners, relying on their
own experience and knowledge of their patients, to make diagnoses, to determine dosages and the
best treatment for each individual patient, and to take all appropriate safety precautions.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors,
assume any liability for any injury and/or damage to persons or property as a matter of products
liability, negligence or otherwise, or from any use or operation of any methods, products, instruc-
tions, or ideas contained in the material herein.

International Standard Book Number: 978-0-323-48024-6

Content Strategist: Patrick Manley


Content Development Specialist: Meredith Clinton
Design Direction: Renee Duenow

Printed in United States of America

Last digit is the print number: 9 8 7 6 5 4 3 2 1


Contributors

Danielle Antin-Ozerkis, MD Robert J. Homer, MD, PhD


Associate Professor of Medicine Professor of Pathology and Medicine (Pulmonary)
Director, Yale Interstitial Lung Disease Program Yale University School of Medicine
Pulmonary and Critical Care Medicine Section New Haven, CT, United States
Department of Internal Medicine
Yale University School of Medicine Kirk D. Jones, MD
New Haven, CT, United States Professor
Department of Pathology
Jürgen Behr, MD University of California, San Francisco, School of
Department of Internal Medicine V Medicine
University of Munich San Francisco, CA, United States
Asklepios Kliniken München-Gauting Comprehensive
Pneumology Center Dong Soon Kim, MD, PhD
Member, German Center for Lung Research Emeritus Professor
Munich, Germany University of Ulsan
Seoul, South Korea
Stefania Cerri, MD, PhD
Center for Rare Lung Disease Kathleen Oare Lindell, PhD, RN
University Hospital of Modena Research Assistant Professor of Medicine
Modena, Italy Clinical Nurse Specialist
Dorothy P. & Richard P. Simmons Center for
Sonye K. Danoff, MD, PhD Interstitial Lung Disease
Department of Medicine Pulmonary, Allergy and Critical Care Medicine
Pulmonary and Critical Care Medicine The University of Pittsburgh
Johns Hopkins University School of Medicine Pittsburgh, PA, United States
Baltimore, MD, United States
Fabrizio Luppi, MD, PhD
Janine Evans, MD Center for Rare Lung Disease
Associate Professor of Medicine University Hospital of Modena
Rheumatology Section Modena, Italy
Department of Internal Medicine
Yale University School of Medicine Richard A. Matthay, MD
New Haven, CT, United States Boehringer Ingelheim Emeritus Professor of Medicine
and Senior Research Scientist
Charlene D. Fell, MD, MSc, FRCPC Pulmonary and Critical Care Medicine Section
Division of Respirology Department of Internal Medicine
Department of Medicine Yale University School of Medicine
University of Calgary New Haven, CT, United States
Calgary, AB, Canada

Christine Kim Garcia, MD, PhD


McDermott Center for Human Growth and
Development
Department of Internal Medicine, Division of
Pulmonary and Critical Care
University of Texas Southwestern Medical Center
Dallas, TX, United States v
vi CONTRIBUTORS

Amy L. Olson, MD, MSPH Aditi Shah, MD


Assistant Professor Division of Respirology, Critical Care, and Sleep
Department of Medicine Medicine
Division of Pulmonary, Critical Care and Sleep Department of Medicine
Medicine University of Saskatchewan
National Jewish Health Saskatoon, SK, Canada
Denver, CO, United States;
Department of Medicine Leann L. Silhan, MD
Division of Pulmonary Sciences and Critical Care Department of Medicine
Medicine Pulmonary and Critical Care Medicine
University of Colorado, Denver Johns Hopkins University School of Medicine
Aurora, CO, United States Baltimore, MD, United States

Luca Paoletti, MD Paolo Spagnolo, MD, PhD


Assistant Professor Respiratory Disease Unit
Division of Pulmonary and Critical Care Medicine Department of Cardiac, Thoracic, and Vascular
Medical University of South Carolina Sciences
Charleston, SC, United States University of Padua
Padua, Italy
Silvia Puglisi, MD
Regional Referral Centre for Rare Lung Diseases Jeffrey J. Swigris, DO, MS
University of Catania Associate Professor
University-Hospital Policlinico–Vittorio Emanuele Department of Medicine
Catania, Italy Division of Pulmonary, Critical Care and Sleep
Medicine
Luca Richeldi, MD, PhD National Jewish Health
Division of Pulmonary Medicine Denver, CO, United States;
Agostino Gemelli University Hospital Department of Medicine
Catholic University of the Sacred Heart Division of Pulmonary Sciences and Critical Care
Rome, Italy Medicine
University of Colorado, Denver
Ami Rubinowitz, MD Aurora, CO, United States
Associate Professor of Diagnostic Radiology
Chief of Thoracic Imaging Anatoly Urisman, MD, PhD
Department of Diagnostic Radiology and Biomedical Assistant Professor
Imaging Department of Pathology
Yale University School of Medicine University of California, San Francisco, School of
New Haven, CT, United States Medicine
San Francisco, CA, United States
Jay H. Ryu, MD
Division of Pulmonary and Critical Care Medicine Carlo Vancheri, MD, PhD
Mayo Clinic Regional Referral Centre for Rare Lung Diseases
Rochester, MN, United States University of Catania
University-Hospital Policlinico–Vittorio Emanuele
Giacomo Sgalla, MD Catania, Italy
National Institute for Health Research
Respiratory Biomedical Research Unit
University Hospital Southampton
Southampton, United Kingdom
CONTRIBUTORS vii

Robert Vassallo, MD Zulma X. Yunt, MD


Division of Pulmonary and Critical Care Medicine Assistant Professor
Departments of Medicine, Physiology and Biomedical Department of Medicine
Engineering Division of Pulmonary, Critical Care and Sleep
Mayo Clinic Medicine
Rochester, MN, United States National Jewish Health
Denver, CO, United States;
Timothy P.M. Whelan, MD Department of Medicine
Professor of Medicine Division of Pulmonary Sciences and Critical Care
Division of Pulmonary and Critical Care Medicine Medicine
Medical University of South Carolina University of Colorado, Denver
Charleston, SC, United States Aurora, CO, United States
Preface

The interstitial lung diseases (ILDs) are a large and het- congestive heart failure, pneumothorax, or drugs. How-
erogeneous group of disease entities that differ signifi- ever, in many cases the etiology is not certain despite
cantly with respect to presentation, cause, prevention, rigorous searches. The current state of our understand-
therapy, and prognosis. Many of them have no clear eti- ing of this important manifestation of IPF is discussed.
ology. However, in recent years considerable progress Also, the mechanisms of action of current and potential
has been made in our understanding of these entities. IPF therapies are reviewed, with particular reference to
In Interstitial Lung Disease, we discuss the most impor- current disease understanding and to highlight areas of
tant of the ILDs and update the reader about their IPF disease biology that afford attractive targets for the
management. development of the new treatments. The general man-
Arriving at the correct diagnosis often requires cor- agement of a patient with IPF is described, especially
relation with clinical, radiologic, and pathologic find- the management of important comorbidities (e.g., pul-
ings. The diagnostic strategy that should be employed monary hypertension and gastroesophageal reflux) and
when faced with a patient with ILD is described. symptoms (e.g., dyspnea, exercise limitation, fatigue,
Given the important role of high-resolution computed anxiety, mood disturbance, sleep disorders) that dra-
tomography (HRCT) in the diagnosis of ILD, we intro- matically affect IPF patients’ lives. Furthermore, the
duce pulmonologists and clinicians to the imaging increasingly important role of lung transplantation in
appearances of ILDs by providing a pattern approach the treatment of patients with ILD is discussed, espe-
to the evaluation of HRCT. In addition, a focused dis- cially the evidence suggesting that patients with pul-
cussion of the HRCT findings in the common ILDs is monary fibrosis undergoing lung transplantation have
presented. a favorable long-term survival compared with other
Idiopathic pulmonary fibrosis (IPF) is the most disease indications.
common of the idiopathic interstitial pneumonias. Finally, several chapters review important individual
Over the past decade, our thinking about IPF has been ILD entities, including inherited fibrotic lung diseases,
reshaped and guidelines have been revised using an nonspecific interstitial pneumonia, ILD associated with
evidence-based approach. A number of epidemiologic connective tissue diseases, chronic hypersensitivity
studies have identified potential risk factors for IPF. We pneumonitis, and smoking-related ILDs.
summarize the approach to diagnosing IPF and review
its epidemiology. With improved understanding of IPF, Harold R. Collard, MD
we now recognize that there may also be distinct phe- Division of Pulmonary and Critical Care Medicine
notypes among this group of patients. Furthermore, we Department of Medicine
describe how the identification and management of University of California San Francisco
comorbidities may improve the overall quality of life San Francisco, CA, USA
and well-being of these patients. It has become clear
that the clinical course of individual patients with IPF is Luca Richeldi, MD, PhD
variable and that the sudden deterioration of a patient’s Division of Pulmonary Medicine
respiratory condition during a relatively stable course Agostino Gemelli University Hospital
is possible. This deterioration can result from well- Catholic University of the Sacred Heart
known causes such as infection, pulmonary embolism, Rome, Italy

ix
CHAPTER 1

Genetic Interstitial Lung Disease


CHRISTINE KIM GARCIA

KEY POINTS

• The interstitial lung diseases (ILDs), or diffuse parenchymal lung diseases, are a heterogeneous collection of more
than 100 different pulmonary disorders that affect the tissue and spaces surrounding the alveoli.
• Many of the genes involve pathways that lead to altered surfactant metabolism, increased ER stress signaling, and
telomere shortening.
• Genetics provides a molecular framework for explaining phenotypes and, sometimes, provides information that
directly affects patient care.

The interstitial lung diseases (ILDs) include a wide mechanism of disease. A detailed family history also
variety of relatively uncommon disorders. Technologic provides important information about personal and
advances in sequencing and genotyping have led to an family member phenotypes, providing important
explosion of genetic discoveries, shedding new light on clues that may suggest a certain genetic diagnosis. For
the underlying pathogenesis of ILD. New monogenic example, a personal or family history of bone marrow
syndromes have been described, often with clinically failure, early graying, or liver disease in a patient with
diverse and extreme phenotypes, based on the discov- adult-onset pulmonary fibrosis suggests a short telo-
ery of single gene mutations. In addition, many appar- mere syndrome. Younger and more severely affected
ently disparate clinical presentations have been linked individuals in later generations may reflect genetic
together through the discovery of mutations in the same anticipation, which can also be seen in short telomere
gene or mutations in multiple genes sharing a common syndromes. Some diseases show a predisposition for
pathway. These genetic discoveries have only further affecting a certain gender, for example, affected males
increased the number of discrete ILDs. The genetic eti- and asymptomatic carrier females suggest an X-linked
ology provides a molecular framework for the disease disorder.
and provides patients and their treating physicians All ILDs arise from the infiltration of inflamma-
with an explanation of phenotypes that are often seen tory and fibrotic mediators into the lung parenchyma.
across multiple organs. As more cohorts of patients are Very few cells normally reside within the interstitium,
described with these rare syndromes, more information which is the delicate space between the alveolar epi-
will be gleaned about the natural history of disease and thelial cells and the capillary vascular endothelial
best practices for surveillance and treatment. The goal cells. The filling of the interstitial space with inflam-
of this chapter is to summarize (1) genetic syndromes matory cells, activated fibroblasts, and extracellular
involving multiple organs, in which ILD is one of many matrix causes irreversible architectural distortion and
different phenotypes, and (2) genetic disorders in impairs gas exchange. Most ILDs share similar clinical
which ILD is the dominant phenotype. signs and symptoms, including respiratory distress and
As a rule, manifestations of ILD even within a cough. Pulmonary restriction and a decreased diffusion
single gene syndrome are generally characterized by capacity are frequently found, as well as radiographic
a spectrum of clinical presentations, a wide range evidence of parenchymal abnormalities. However, the
in age of onset, and incomplete penetrance. Thus, a radiographic and histopathologic features of ILDs vary
high level of suspicion is needed for many of these widely. Historically, the type of infiltrating cells, the
disorders. Detecting a pattern of inheritance in large, pattern of infiltration (nodular, reticular, alveolar), the
extended kindreds across multiple generations sepa- nature of extracellular protein deposits (collagen, elas-
rated by time and space strongly supports a genetic tin, periodic acid–Schiff [PAS]-positive), the location

1
2 Interstitial Lung Disease

of abnormalities (peripheral, alveolar, peribronchio- and extrathoracic lymphatic structures. The hallmark
lar), the pattern of the fibrotic response (fibroblastic feature of the disease is the radiographically apparent
foci, temporal/spatial homogeneity, or heterogeneity), numerous, round, thin-walled cysts, generally between
and the form of lung destruction (cysts, bronchiectasis, 2 and 60 mm in diameter, which are found throughout
honeycombing) have been used to describe different the lung. Extraparenchymal lung abnormalities include
clinical forms of ILD. Now that genetic underpinnings pneumothorax and chylous pleural effusions. Eighty
of some monogenic ILDs are being established, clas- percent of patients develop a pneumothorax at some
sification by genetic etiology may ultimately supplant time during the course of their disease. Pleural effusions
historical classification schemes. This will occur most are characterized by elevated triglyceride levels and an
readily for those disorders in which the genetic clas- abundance of chylomicrons. Obstructive and mixed
sification predicts specific treatments (e.g., sirolimus obstructive-restrictive ventilator defects are observed.
for tuberous sclerosis complex [TSC]–lymphangioleio- Renal angiomyolipomas occur in ∼30% of sporadic
myomatosis [LAM]). A genetic classification of ILD is LAM patients and in >90% of TSC-LAM patients.1
also advisable for those diseases in which the genetic LAM can occur either in patients with sporadic (non-
information provides information relevant to patient inherited) disease or in 1–3% of patients with TSC, an
care. For example, regular screening may lead to ear- autosomal dominant multisystem disorder character-
lier interventions to remove premalignant renal cancers ized by hamartomas in multiple organ systems, includ-
in patients with Birt-Hogg-Dubé syndrome (BHDS). ing the brain, skin, heart, kidneys, and lungs. The ILD
Similarly, knowledge of a monogenic short telomere manifestation of TSC patients is indistinguishable
syndrome provides prognostic information regarding from sporadic LAM.2 TSC patients who develop LAM
the rate of ILD progression and the nature of specific are often women over 30 years of age who have little
post–lung transplant complications. or no mental retardation. Extrapulmonary features
In this chapter, I focus primarily on disorders caused include facial angiofibromas (or adenoma sebaceum),
by rare mutations and include selected common vari- hypomelanotic macules (which are most easily visual-
ants that significantly increase susceptibility to ILD. ized with a Wood light), rough, yellow thickening of
The nomenclature of diseases follows the genetic clas- skin over the lumbosacral area (shagreen patch), ungual
sification system adopted by the Online Mendelian or periungual fibromas, renal angiomyolipomas, renal
Inheritance in Man (http://www.omim.org). Other pul- cysts, subependymal giant cell astrocytomas, brain
monary genetic diseases are not covered. I refer to other cysts, hamartomas, cardiac rhabdomyomas, dental pits,
excellent resources for reviews of alpha-1-antitrypsin epilepsy, learning difficulties, and mental retardation.
deficiency, cystic fibrosis and CFTR-related disorders, TSC is characterized by autosomal dominant inheri-
primary ciliary dyskinesia, pulmonary capillary or veno- tance. However, about two-thirds of patients have de novo
occlusive disease, pulmonary malformation syndromes, mutations. There are two genes associated with this syn-
and disorders that primarily affect the thoracic cage. drome. Mutations in TSC2 occur more frequently, account-
ing for 75–80% of all cases.3 Mutations in TSC1 are less
common.3 There are no mutational hot spots within these
GENETIC DISORDERS AFFECTING MULTIPLE two genes. In general, TSC2 mutations are associated with
ORGANS, INCLUDING THE LUNG more severe disease, more frequent and severe epilepsy,
Table 1.1 lists the defined genetic disorders that are mental retardation, and cortical tubers. Inactivation of
associated with ILDs, with the lung being only one of both alleles of TSC1 or TSC2 is needed for the develop-
the many different affected organs. In this broad cat- ment of tumors. Loss of heterozygosity is frequently found
egory that encompasses many different disorders, ILD in renal angiomyolipomas and supports Knudson’s two-
may be a more severe or life-threatening manifestation hit model tumor suppressor pathogenesis.4
of disease or one of the more common features associ- Most sporadic LAM results from two somatic muta-
ated with the disease. tions in the TSC2 gene, although rare cases are caused
by germline TSC1 mutations.5–7 In contrast with TSC
Lymphangioleiomyomatosis and Tuberous patients in whom germline mutations are present in
Sclerosis Complex tumor and normal tissue, the mutations in LAM patients
Pulmonary LAM is a rare disease that almost exclu- are generally not present in normal tissues. This sug-
sively affects women. It is caused by proliferating gests that two mutations arise in a precursor cell of ori-
smooth muscle–like cells, which can involve small gin and that the mutation-carrying smooth muscle–like
airways, the pulmonary vasculature, and intrathoracic cells spread to various organs (lungs, kidneys).
CHAPTER 1 Genetic Interstitial Lung Disease 3

TABLE 1.1
Genetic Interstitial Lung Disease (ILD): Disorders Affecting Multiple Organs, Including the Lung
Pulmonary ILD
Disease Inheritance Gene Pathogenesis Involvement Presentation
LAM/tuberous Sporadic, TSC1, TSC2 mTOR ∼100% Multiple cysts
sclerosis AD activation
Birt-Hogg-Dubé AD FLCN Loss of ∼90% Multiple cysts
syndrome folliculin
Dyskeratosis XLR, AD, DKC1, TERC, TERT, Telomere Second Inflammatory
congenita AR NOP10, NHP2, shortening most serious infiltrates and
TINF2, WRAP53, complication interstitial fibrosis
RTEL1, ACD, CTC1,
PARN
RIDDLE syndrome AR RNF168 DSB repair defect 50% Pulmonary fibrosis
Hermansky-Pudlak AR HPS1, AP3B1, Cytoplasmic or- Described Fibrosis, foamy
syndrome HPS3, HPS4, HPS5, ganelle defect for those type II
HPS6, DTNBP1, with HPS1, pneumocytes
BLOC1S3, BLOC1S6 HPS2, and
HPS4
NKX2-1 related AD NPX2-1 Transcription fac- ∼50% Respiratory
disorders tor defect distress, NEHI,
pulmonary fibrosis
Neurofibromatosis AD NF1 Loss of tumor ∼10% Pulmonary
suppressor fibrosis, bullae
Poikiloderma with tendon AD FAM111B Unknown >50% UIP
contractures and
pulmonary fibrosis
ILD, nephrotic AR ITGA3 Integrin defect 100% Abnormal
syndrome, and alveolarization
epidermolysis
bullosa
STING-associated AD TMEM173 Increased >50% Alveolitis with
vasculopathy interferon fibrosis
Autoimmune disease AR ITCH E3 ubiquitin ligase ∼90% NSIP
with facial defect
dysmorphism
Autoimmune interstitial AD COPA ER stress ∼100% Lymphocytic
lung, joint, and kidney infiltration
disease
GATA2 deficiency Sporadic, GATA2 Transcription fac- ∼60% PAP
AD tor defect
CVID syndromes Sporadic, ICOS, CD19, CD20, Variable 55–90% Bronchiectasis,
AR, AD CD21, CD81, TN- antibody GLILD, organizing
FRSF13B, TNFRS- deficiencies pneumonia
F13C, LRBA, IL21,
NFKB1, NFKB2,
IKZF1
Agammaglobulinemia X-linked, BTK, IGHM, IGLL1, B cell defect ∼25% Bronchiectasis
AR, AD CD79A, CD79B,
BLNK, TCF3
Continued
4 Interstitial Lung Disease

TABLE 1.1
Genetic Interstitial Lung Disease (ILD): Disorders Affecting Multiple Organs, Including the Lung—cont’d
Pulmonary ILD
Disease Inheritance Gene Pathogenesis Involvement Presentation
Hyper IgE syndrome Sporadic, STAT3 STAT3 defect, ∼70% Bronchiectasis,
AD Lack of Th17 cells pneumatoceles
Activated PI3K-δ AD PIK3CD Activation of PI3K ∼75% Lymphoid
syndrome aggregates
CTLA4 deficiency AD CTLA4 Activated T cells ∼75% GLILD
X-linked reticulate X-linked POLA1 Increased type 1 ∼100% Bronchiectasis
pigmentary interferons
Gaucher disease, type I AR GBA Deficiency of acid ∼65% Gaucher cell
β-glucosidase infiltration
Niemann-Pick disease, AR SMPD1, NPC1, Deficiency of acid Various GGO and septal
types A, B, C1, and C2 NPC2 sphingomyelinase; thickening
defective intracellu-
lar lipid movement
Lysinuric protein AR SLC7A7 Defect of cationic PAP, septal
intolerance amino acid thickening
transport

AD, autosomal dominant; AR, autosomal recessive; DSB, double-stranded DNA break; GGO, ground-glass opacities; GLILD, granulomatous
lymphocytic interstitial lung disease; mTOR, mammalian target of rapamycin; NEHI, neuroendocrine cell hyperplasia of infancy; NSIP, nonspe-
cific interstitial pneumonia; PAP, pulmonary alveolar proteinosis; UIP, usual interstitial pneumonia; XLR, X-linked recessive.

The proteins encoded by these two genes regulate and represent a forme fruste of BHDS.17,18 Nearly all
the mammalian target of rapamycin (mTOR) path- mutations, including the common mutation hot spot
way.8 Inhibition of the mTOR complex corrects the that introduces a frameshift mutation (c.1285insC
specific molecular defects underlying TSC. mTOR and c.1285delC), predict a truncated protein prod-
inhibitors cause shrinkage of renal or retroperito- uct.15 The pulmonary cysts are generally bilateral, are
neal angiomyolipomas and subependymal giant cell usually located in a subpleural distribution in the
astrocytomas in TSC patients.9–11 In addition, siro- mid- and lower-lung zones, and are of varying sizes,
limus stabilizes lung function, reduces respiratory ranging from round, oval, lentiform, to multisep-
symptoms, and improves the quality of life of LAM tated shapes.19,20 Because the skin findings typically
patients.12 appear in the fourth decade and the renal malignancy
can be a late finding (mean age of 48 years), a spon-
Birt-Hogg-Dubé Syndrome taneous pneumothorax in the second or third decade
This disorder, which is also known as Hornstein- of life is often the presenting manifestation.15,18
Knickenberg syndrome,13 is characterized by the The molecular mechanism of lung cyst formation is
autosomal dominant inheritance of multiple benign incompletely understood.
skin tumors, which are usually characterized as fibro- Genetic testing is indicated for all individuals with sus-
folliculomas. Approximately 90% of affected indi- pected BHDS because the presence of a mutation prompts
viduals have evidence of lung cysts,14 and up to 34% screening for renal cancer for which surgical resection may
develop kidney tumors.15 The disease is caused by be curative. As was described in a past case,21 the pheno-
loss-of-function mutations in the folliculin (FLCN) type of familial spontaneous pneumothorax may be the
gene.16 Two different studies, one using whole- presenting feature for the index case that then leads to the
genome linkage analysis and the other a candidate identification of an occult renal malignancy in a family
gene approach, independently found that kindreds member with a FLCN mutation. All first-degree relatives
presenting solely with familial spontaneous pneu- have a 50% chance of having the same germline FLCN
mothorax and/or lung cysts have mutations in FLCN mutation.
CHAPTER 1 Genetic Interstitial Lung Disease 5

Dyskeratosis Congenita with the catalytically active RNA and protein compo-
Dyskeratosis congenita (DC) is a rare multisystem dis- nent of telomerase.34 Mutations in both genes encod-
order characterized by the classic triad of a lacy reticular ing telomerase (hTR, encoded by TERC gene and TERT)
pigmentation on the upper chest and neck, nail dystro- have been found in patients with autosomal dominant
phy, and oral leukoplakia. The prevalence is approxi- DC35,36; biallelic mutations in TERT have been found
mately 1 in 1,000,000, with death occurring at a median in patients with autosomal recessive DC.37,38 Germ-
age of 16 years.22 Patients are usually healthy at birth line mutations in TINF2, a component of the shelterin
and then develop different organ dysfunction, includ- complex that protects the telomeric ends of chromo-
ing bone marrow failure, pulmonary fibrosis, and eye, somes, mostly occur de novo and are found in patients
tooth, gastrointestinal, endocrine, skeletal, urologic, and with autosomal dominant DC.39,40 Mutations in the
immunologic abnormalities. Significant developmental gene ACD, encoding another member of the shelterin
delay is found for the more severe clinical variants of DC. complex, TPP1, have been found in two kindreds with
There is a wide variation in the severity and spectrum of DC.41,42 Compound heterozygous mutations in CTC1
clinical findings, which is only partly explained by locus were first described in patients with Coats plus and in
heterogeneity. The mode of inheritance varies by gene the phenotypically similar disorder cranioretinal micro-
mutation. Most patients in a large international registry angiopathy with calcifications and cysts and then were
are male with X-linked recessive inheritance.23 Kindreds later described in patients with autosomal recessive
with autosomal recessive and autosomal dominant pat- DC.43,44 CTC1 is part of the trimeric telomere capping
terns of inheritance are less common. For affected male complex that cooperates with the shelterin complex to
patients, the classic skin and nail findings are present in protect telomeres. Homozygous mutations in NOLA2
∼90%. The abnormal skin findings have been described and NOLA3 account for less than 1% of DC mutations;
as poikiloderma vascularis atrophicans or a latticework these genes encode NHP2 and NOP10, respectively,
hypo- and hyperpigmentation defect found especially which maintain the stability and proper trafficking of
on the neck, upper chest, and proximal limbs.23,24 Bone a complex of H/ACA small nucleolar RNAs, including
marrow failure is very common (>85%) and is the lead- the telomerase RNA, hTR.45–47 Mutations in WRAP53
ing cause of death. have been described linked to DC; this gene encodes
After bone marrow failure, pulmonary fibrosis is TCAB1 that binds telomerase and directs its localiza-
the most serious and life-threatening complication of tion to nuclear Cajal bodies, an important step in telo-
DC. In most cases it presents either after hematopoietic mere maintenance.48 Whole exome sequencing has led
cell transplantation25,26 or as a later manifestation of to the identification of heterozygous or compound het-
disease in those over the age of 30 years.27–29 Patients erozygous mutations in RTEL1.49–51 This gene encodes
with pulmonary disease have rales, digital clubbing, a a DNA helicase that is crucial for telomere maintenance
restrictive pulmonary defect, a reduced diffusion capac- and DNA repair. Homozygous or compound hetero-
ity, and diffuse interstitial markings on high-resolution zygous mutations in PARN have most recently been
computed tomography (HRCT) imaging of the chest. linked to autosomal recessive DC.52–54 The PARN pro-
Lung histopathology generally features a mixture of tein belongs to a family of conserved exoribonucleases
cellular inflammatory infiltrates and interstitial fibro- that shorten the poly(A) tail of messenger RNA through
sis.30,31 Clinical survival of DC patients after the devel- deadenylation. One of the RNA species that is modi-
opment of ILD is poor as pulmonary disease is generally fied by PARN is hTR or the RNA component of telo­
rapidly progressive. Death is usually 12–40 months merase.54 Thus, mutations in PARN lead to defective
after the onset of clinical symptoms.28 hTR and telomerase biogenesis and telomere disease.
Mutations in 11 different genes have been described Genetic anticipation has been seen in DC kindreds
in DC patients. Regardless of the pattern of genetic with TERC and TERT mutations. Inheritance of pro-
inheritance or the individual genetic mutation, all gressively shorter telomere lengths in each subsequent
DC patients exhibit short telomere lengths for their generation of mutation carriers provides a molecular
age.32 Telomeres are specialized structures essential for explanation for the observation of an earlier age of
maintenance of integrity of chromosomal ends com- onset in more severely affected individuals with each
posed of nucleotide repeats (TTAGGG)n and telomere- successive generation.36,55 Siblings who do not inherit
specific accessory proteins. Positional cloning first led the mutated gene can inherit short telomere lengths
to the identification of mutations in the DKC1 gene from the affected parent.56 Thus, DC patients have to
in those with the X-linked recessive form of the dis- inherit both short telomere lengths and a deleterious
ease.33 Dyskerin is a nucleolar protein that copurifies telomerase mutation to demonstrate anticipation.
6 Interstitial Lung Disease

RIDDLE Syndrome a peripheral distribution of HRCT reticulations with a


Patients with this very rare autosomal recessive disorder trend toward increasing involvement of the central por-
have a syndrome of increased sensitivity to radiation, tions of the lungs with progressive severity as a result of
immunodeficiency, learning difficulties, dysmorphic peribronchovascular thickening and bronchiectasis.73
features, short stature, and pulmonary fibrosis in later Surgical lung biopsies demonstrate lung remodeling,
years.57,58 The syndrome is caused by homozygous or numerous chronic inflammatory cells, and distinctive
compound heterozygous mutations in the RNF168 clusters of clear vacuolated type II pneumocytes with
gene, an E3 ubiquitin ligase critical for double-stranded florid foamy swelling and degeneration (“giant lamel-
DNA break (DSB) repair. It promotes ubiquitination of lar body degeneration”).78,79 Despite an early positive
H2A and H2AX, which, in turn, mediates accumula- study,76 a follow-up clinical trial showed no benefit of
tion of 53BP1 and BRACA1 at DSBs to promote DNA pirfenidone for the treatment of Hermansky-Pudlak
repair.59,60 It also has a role in TRF2-mediated protec- pulmonary fibrosis.80
tion of telomeres.61 Pathogenesis of pulmonary fibrosis is incompletely
understood. Studies of a mouse model of disease that is
Hermansky-Pudlak Syndrome homozygous for both HPS1 and HPS2 mutations dem-
Hermansky-Pudlak syndrome (HPS) is an autosomal onstrate additive effects of the genetic defects toward
recessive disease first described in 1959.62 It is character- the development of spontaneous pulmonary fibrosis.81
ized by oculocutaneous albinism, a bleeding diathesis Subpleural reticulations begin at 3 months and exten-
resulting from platelet storage pool deficiency, and in sive fibrosis is seen by 9 months.82 The histology of
some cases, pulmonary fibrosis.63 It is now known that the mouse lung replicates the human phenotype with
HPS is caused by defects of multiple cytoplasmic organ- “giant lamellar body degeneration” of the type II cells
elles, including melanosomes, platelet-dense granules, and demonstrates decreased phospholipid and surfac-
and lysosomes leading to diverse clinical features. tant protein (SP)-B and C secretion.81,83 The underlying
Other clinical features of the disease include granulo- molecular mechanism of murine HPS-associated pul-
matous colitis, neutropenia, immunodeficiency, cuta- monary fibrosis seems related to endoplasmic reticu-
neous malignancies, cardiomyopathy, and renal failure. lum (ER) stress and chronic alveolar epithelial type II
Currently the diagnosis is confirmed by the absence of cell injury.82
dense bodies on whole-mount electron microscopy of
platelets.64 Mutations in nine different genes cause this NKX2-1–Related Disorders
disease. Each of these genes functions in trafficking of These disorders include benign hereditary chorea, neo-
vesicular cargo proteins to cytoplasmic organelles or in natal respiratory distress, and congenital hypothyroid-
organelle biogenesis and maturation.65–70 HPS is the ism; it is also known as brain-lung-thyroid syndrome.
most common single gene disorder in Puerto Rico with Childhood-onset chorea is the hallmark clinical feature
an estimated frequency of about 1 in 1800 and a carrier of this disorder. Pulmonary disease is the second most
frequency of 1 in 21.71 Mutations in HPS1 and HPS3 common presentation, with nearly half of the patients
are found in 75% and 25% of Puerto Rican patients, having some type of pulmonary manifestation.84 These
respectively. include neonatal respiratory distress with or without
ILD generally causes symptoms in the 30s and, pulmonary hypertension, neuroendocrine cell hyper-
when it occurs, is generally fatal within a decade. plasia, ILD in children between the ages of 4 months
Patients have progressive restrictive disease with a to 7 years, and pulmonary fibrosis in older individu-
highly variable course.63,72 Pulmonary fibrosis has been als.85–87 Defective surfactant homeostasis and recurrent
described most frequently in patients with HPS1 muta- respiratory infections have been found to be a promi-
tions, and more rarely in patients with HPS2 or HPS4 nent feature in some subjects.86,88 Mutations include
mutations.72–77 In this regard, molecular subtyping is missense, nonsense, and deletions of the NKX2-1 gene.
important to assess the risk of developing pulmonary
fibrosis. In one of the largest studies, the mean age of Neurofibromatosis
onset was 35 years with a range of 15–53 years.72 The Neurofibromatosis (NF1) is an autosomal dominant
variability of pulmonary findings was generally not disorder that affects all ethnic groups. Type I, von
attributable to prior environmental exposures. Over Recklinghausen disease or classic NF1, is character-
80% of patients had abnormalities on CT scans, which ized by multiple (>6) café au lait spots, axillary and
were generally predictive of the degree of physiologic inguinal freckling, multiple cutaneous neurofibromas,
impairment and mortality. Most patients demonstrate and iris Lisch nodules. Pulmonary manifestations
CHAPTER 1 Genetic Interstitial Lung Disease 7

are less common. The incidence of ILD in NF1 has Stimulator of Interferon Genes–Associated
been estimated at 6–12%.89–91 It is characterized by Vasculopathy, Infantile-Onset
lower lobe–predominant diffuse interstitial fibrosis Patients with this autosomal dominant disorder have
and honeycombing. Thin-walled bullae are present in an autoimmune vasculopathy that causes severe skin
almost all patients with ILD or may be seen in isola- lesions affecting the face, ears, nose, and digits, result-
tion; they are large, asymmetric, and typically involve ing in ulceration and necrosis beginning in infancy.
the upper lobes.91 Histologic evidence of an alveoli- Over 85% have an ILD apparent on HRCT chest imag-
tis and interstitial fibrosis has been found in patients ing. Lung biopsy samples demonstrate a scattered
with normal chest X-rays or those with only apical mix of lymphocytic inflammatory infiltrate, follicular
bullae.90 Although there is near complete penetrance hyperplasia and B-cell germinal centers, interstitial
of the disease after childhood, the ILD is not observed fibrosis, and emphysematous changes.97,98 Patients can
until adulthood, typically in patients older than have livedo reticularis, Raynaud phenomenon, myosi-
40 years of age.90 The disease is often progressive and tis, joint involvement, immune complex deposition,
may lead to pulmonary hypertension and right-sided hypergammaglobulinemia, leukopenia, and autoan-
heart failure.92 The pathogenesis of NF-associated ILD tibodies (antinuclear, antiphospholipid, anticardio-
is unknown. lipin). All cases are caused by heterozygous, missense,
gain-of-function mutations in the TMEM173 gene,
Poikiloderma, With Tendon Contractures, which encodes the STING (stimulator of interferon
Myopathy, and Pulmonary Fibrosis genes).97,98 Mutations lead to activation of STAT1.97
This rare disorder usually affects individuals from
early childhood; presenting features include thin hair, Autoimmune Disease With Facial
telangiectasias and pigmentary abnormalities on sun- Dysmorphism
exposed areas, and tendon contractures frequently This autosomal recessive disease has been described
involving the ankles and feet.93 There is incomplete only in children with features of failure to thrive, devel-
penetrance of pulmonary fibrosis; >50% of cases have opmental delay, dysmorphic features, autoantibodies,
lung involvement. When it is present, the pulmonary and inflammatory cell infiltration of the lungs, liver,
fibrosis generally develops during the second decade of and gut. Chronic lung disease was found in 9 of 10 chil-
life, is progressive, and can lead to death. The histopa- dren. Three had a chronic oxygen requirement and died
thology for at least one case was consistent with usual of respiratory failure. Surgical lung biopsy from one
interstitial pneumonia.93 Different missense mutations case showed a cellular, nonspecific interstitial pneumo-
in the FAM111B gene have been found in unrelated nitis (NSIP). All affected children in one large Amish
cases of different ancestry.94 The function of this gene kindred were homozygous for a frameshift mutation in
is unclear. the ITCH gene that is predicted to cause a truncation
of an E3 ubiquitin ligase.99 Of note, mice with a small
Interstitial Lung Disease, Nephrotic genomic inversion that disrupts the Itch and agouti
Syndrome, and Epidermolysis Bullosa genes develop a similar spectrum of immunologic dis-
Infants with this congenital autosomal recessive disor- ease of the spleen, lymph nodes, stomach, and skin,
der present with disease in multiple organs. The pul- as well as chronic pulmonary interstitial inflammation
monary and renal features cause significant morbidity and alveolar proteinosis.100,101
and mortality, although dermatologic features are
common (epidermolysis bullosa, thin hair, dystrophic Autoimmune Interstitial Lung, Joint, and
nails, onycholysis). The lung involvement is character- Kidney Disease
ized radiographically by diffuse ground-glass opacity This rare syndrome was recently elucidating by whole
and interlobular septal thickening. Histology demon- exome sequencing of patients from five unrelated
strates abnormal alveolarization with poorly septated families with autoimmune interstitial lung, joint, and
(“simplified”) alveolar spaces.95,96 All mutations are kidney disease.102 All patients had high-titer autoanti-
homozygous deletion/frameshift or missense muta- bodies, ILD, and inflammatory arthritis. The average age
tions in ITGA3, which encode an integrin alpha chain at presentation was 3.5 years, with a range of 6 months
belonging to a family of cell surface adhesion proteins. to 22 years. Some patients presented with pulmonary
One of the missense mutations prevents proper post- hemorrhage that required immunosuppression. Lung
translational modification of the integrin alpha chain biopsies showed lymphocytic interstitial infiltration
with defective cell surface expression. with lung-infiltrating CD4+ T cells and CD20+ B cells
8 Interstitial Lung Disease

within germinal centers. All mutations affect the same This genetically heterogeneous group of diseases
functional domain of the COPA protein, impair intra- includes those with variable antibody and B-cell,
cellular ER-Golgi transport, and lead to ER stress and T-cell, and NK-cell deficiencies, as well as defective
upregulation of cytokines. responses to vaccination. Mutations in multiple genes,
including ICOS,113 TNFRSF13B (encoding the trans-
GATA2 Deficiency membrane activator and CAML interactor TACI),114,115
Haploinsufficiency of the hematopoietic transcrip- TNFRSF13C (encoding the B-cell activating factor),116
tion factor GATA2 leads to a wide spectrum of diseases CD19,117 CD20,118 CD81,119 CD21,120 LRBA,121,122
including primary immunodeficiency syndromes such IL21,123 NFKB1,124 NFKB2,125 and IKZF1 (encoding the
as dendritic cell, monocyte, B lymphocyte, and NK hematopoietic zinc finger transcription factor),126,127
lymphocyte deficiency (DCML) and monocytopenia lead to sporadic, autosomal recessive, and autosomal
and mycobacterial infection syndrome (MONOMAC); dominant patterns of disease.
primary lymphedema with myelodysplasia (Emberger X-linked agammaglobulinemia (XLA) is an immuno-
syndrome); and susceptibility to myelodysplastic syn- deficiency disorder that occurs almost exclusively in
drome and acute myeloid leukemia. Besides immu- males from a failure of B-cell maturation. Although
nodeficiency (with predisposition especially to severe there is a high prevalence of pneumonia in XLA, there
viral, disseminated nontuberculous mycobacterial, and is a lower prevalence of chronic lung disease with 47%
invasive fungal infections), pulmonary disease, vascu- and 25% demonstrating abnormal pulmonary function
lar/lymphatic dysfunction, and hearing loss are com- studies and bronchiectasis, respectively.109 Although
mon.103–107 Most individuals have some manifestation the degree of immune dysregulation may be lower,109
of pulmonary disease; 79% have a diffusion defect, 63% the rate of progression of lung function decline is
with abnormal pulmonary function tests, 18% with greater for those with XLA.128 Between 85% and 90% of
biopsy-proved pulmonary alveolar proteinosis (PAP), cases are caused by mutations in BTK, the gene encod-
and 9% with pulmonary arterial hypertension.107 All ing Bruton tyrosine kinase and a key regulator in B-cell
lack anti–GM-CSF antibodies and are resistant to sub- development.129 The remaining cases include a geneti-
cutaneous or inhaled granulocyte-macrophage colony– cally heterogeneous group of patients with mutations
stimulating factor (GM-CSF) therapy. Chest CT findings in IGHM (encoding the mu heavy-chain),130 IGLL1
include reticulonodular opacities, “crazy paving,” and (encoding immunoglobulin lambda-like-1),130,131
paraseptal emphysema. Median age at first presentation CD79A,132 BLNK,133 CD79B,134 and TCF3.135
is 20 years, but is highly variable (5 months to 78 years). Autosomal dominant hyper-IgE syndrome is an immune
Most mutations are predicted to cause a loss of function deficiency disorder characterized by the triad of recur-
of the mutated allele, leading to haploinsufficiency and rent staphylococcal skin abscesses, pneumonias, and
autosomal dominant inheritance.106,108 Hematopoietic elevations of IgE (usually >2000 IU/mL). Survival is
stem cell transplantation is indicated for severe disease. typically into adulthood, but death is usually secondary
to infections. Diagnosis requires a high level of suspi-
Immunodeficiency Syndromes cion due to variability of phenotypic features. A clini-
Common variable immunodeficiency (CVID) syndromes cal scoring system has been developed that combines
are all associated with an increased susceptibility to immunologic (elevated IgE, eosinophilia >700/μL,
recurrent infections, especially sinopulmonary infec- decreased T-helper [Th]17 cells, recurrent infections)
tions. There is a high variability of clinical features. and nonimmune features (retained primary teeth, sco-
Respiratory complications are frequently responsible liosis, joint hyperextensibility, bone fractures following
for patient morbidity and mortality and include acute minimal trauma, a typical facial appearance, and vascu-
infections, sequelae of infections (bronchiectasis), lar abnormalities). Pneumatoceles and bronchiectasis,
noninfectious immune-mediated infiltrates such as which result from aberrant healing of the pneumonias,
granulomatous lymphocytic interstitial lung disease are seen in ∼70% of patients and lead to significant
(GLILD) and lymphoma, and progressive respiratory mortality.136,137 Patients have heterozygous missense,
failure.109,110 When severe, bronchiectasis is gener- splice site, or small deletions in the STAT3 gene.138 Most
ally associated with obstructive lung disease; GLILD mutations cluster in the DNA-binding domain of this
is typically associated with restrictive physiology and transcription factor, which regulates responses to many
decreased diffusion capacity.111 Lung disease is fre- different cytokines. Patients’ purified native T cells are
quently found in patients with CVID, as >90% have unable to differentiate in vitro into IL-17–producing
radiographic abnormalities apparent on chest CT.112 Th17 cells, which play a critical role in the clearance of
CHAPTER 1 Genetic Interstitial Lung Disease 9

fungal and extracellular bacterial infections of the lung and bone manifestations including osteopenia, lytic
and skin.139 Most patients represent sporadic cases, but lesions, bone crisis, and skeletal deformities. Infiltra-
autosomal dominant transmission has been seen in tion of Gaucher cells into the alveoli, interstitium,
some kindreds. and pulmonary capillaries can lead to lung involve-
Activated PI3K-delta syndrome is a monogenic auto- ment. Over 65% of patients with type I disease have
somal dominant disease that leads to overactivation pulmonary function abnormalities, but only a fraction
of the PI3K signaling pathway and lymphoprolifera- (<5%) have diffuse lower lobe linear infiltrates, restric-
tion. This autosomal dominant disease is character- tive physiologic impairment, and a reduced diffusion
ized by recurrent sinopulmonary infections, reduced capacity consistent with ILD.148,149 Enzyme replace-
IgG2, increased serum IgM, and impaired vaccine ment therapy can reduce organ volumes and improve
responses. In one series, 75% of patients had CT evi- the hematologic parameters and bone pain but is usu-
dence of bronchiectasis or mosaic attenuation.140 The ally poorly effective in treating the lung manifestations
lymphoid aggregates within the lung and can lead to of this disease.150 Other therapeutic treatments, such
the compression of nearby bronchi.110 Heterozygous as substrate reduction therapy as well as others, are in
missense mutations in PIK3CD result in increased development.151
phosphorylation of AKT, consistent with a gain of Niemann–Pick disease types A and B are caused by
function.140,141 an inherited deficiency of acid sphingomyelinase activ-
CTLA4 deficiency syndrome is an autosomal dom- ity. Type C is caused by defective movement of lipids,
inant immunodeficiency characterized by T-cell including cholesterol, from endosomes and lysosomes.
immune dysregulation. Pulmonary manifestations Patients demonstrate a range of ages and can have
mirror CVID with recurrent respiratory infections and diverse symptoms affecting the lung, liver, spleen, bone
GLILD. Lymphocytic infiltration is found for other marrow, skeleton, brain, muscle, mental ability, and
organs, including the bone marrow, kidney, brain, movement. The pulmonary manifestations include ILD
liver, spleen, and lymph nodes. Heterozygous loss- and recurrent lung infections. In some patients the ILD is
of-function mutations in CTLA4 have been recently predominantly in the bases with thickened interlobular
described.142,143 septa, interlobular lines, and ground-glass opacities.153
X-linked reticulate pigmentary disorder is a rare Lysinuric protein intolerance is an autosomal reces-
immunodeficiency disorder with features of recurrent sive disease caused by an inherited defect of cationic
infections and systemic sterile inflammation. Affected amino acid transport. There is excess urinary clear-
patients typically develop recurrent pneumonias in ance of these amino acids and deficient intestinal
the first few months of life. By childhood, affected absorption, which leads to depleted body pools.
males develop diffuse skin hyperpigmentation with Most patients present in infancy with failure to thrive,
a distinctive reticulate pattern, bronchiectasis, hypo- growth retardation, protein aversion, muscular hypo-
hidrosis, corneal scarring, enterocolitis, and recur- tonia, hepatosplenomegaly, and osteoporosis. In one
rent urethral strictures.144–146 A recurrent intronic study, all patients who developed a fatal respiratory
mutation has been found that disrupts the expression insufficiency, usually PAP, were children less than
of the catalytic subunit of DNA polymerase-alpha 15 years of age.154 Most adult patients have evidence of
(POLA1) and leads to increased production of type I an ILD on CT scans with interlobular and intralobular
interferons.147 septal thickening and subpleural cysts, but only a few
are symptomatic.154
Inborn Errors of Metabolism
Gaucher disease is an autosomal recessive lysosomal
storage disease characterized by the accumulation of GENETIC DISEASES IN WHICH
the glycolipid glucosylceramide due to the deficiency INTERSTITIAL LUNG DISEASE IS THE
of the enzyme acid-beta glucosidase. Diagnosis relies DOMINANT PHENOTYPE
on the demonstration of deficient enzyme activity in Table 1.2 lists the genetic disorders in which an ILD is
cells or the identification of two disease-causing muta- the dominant clinical feature. Included in this group
tions in the GBA gene. Patients can display a large are the surfactant disorders. Pulmonary surfactant is
variety of symptoms, ranging from patients who are a mixture of phospholipids and associated proteins
completely asymptomatic to those who present with that covers the alveolar surface at the gas-alveolus
perinatal lethality. The usual clinical findings include interface, where it functions to reduce surface tension
hepatosplenomegaly, anemia, thrombocytopenia, and prevent atelectasis. A group of genetic disorders
10 Interstitial Lung Disease

TABLE 1.2
Genetic Interstitial Lung Disease (ILD): Disorders in Which ILD Is the Dominant Phenotype
Disease Inheritance Gene Pathogenesis Presentation
Surfactant metabolism AR SFTPB Absent SP-B Neonatal respiratory failure
dysfunction 1
Surfactant metabolism AD SFTPC Lack of SP-C and Neonatal respiratory
dysfunction 2 ER stress distress, NSIP, UIP
Surfactant metabolism AR ABCA3 Phospholipid Neonatal respiratory
dysfunction 3 transport defect distress, PAP
Surfactant metabolism AR CSF2RA GM-CSF signaling PAP
dysfunction 4 CSF2RB defect
Interstitial lung and liver AR MARS Methionine tRNA PAP
disease ligase defect
Pulmonary fibrosis, AD TERT Short telomere Pulmonary fibrosis
telomere related, type 1 length
Pulmonary fibrosis, AD TERC Short telomere Pulmonary fibrosis
telomere related, type 2 length
Pulmonary fibrosis, AD RTEL1 Short telomere Pulmonary fibrosis
telomere related, type 3 length
Pulmonary fibrosis, AD PARN Short telomere Pulmonary fibrosis
telomere related, type 4 length
Pulmonary fibrosis, Various DKC1, TINF2, NAF1 Short telomere Pulmonary fibrosis
telomere related length
Pulmonary fibrosis and AD SFTPA1, SFTPA2 ER stress Pulmonary fibrosis,
adenocarcinoma adenocarcinoma, respiratory
distress in infancy
Pulmonary fibrosis Sporadic MUC5B ER stress? UIP, ILAs
susceptibility
Pulmonary fibrosis Sporadic TERT, TERC, OBFC1 Short telomere UIP
susceptibility length
Pulmonary alveolar AR SLC34A2 Phosphate trans- Microliths
microlithiasis port defect

ER, endoplasmic reticulum; GM-CSF, granulocyte-macrophage colony-stimulating factor; ILA, interstitial lung abnormalities; NSIP, nonspe-
cific interstitial pneumonia; PAP, pulmonary alveolar proteinosis; SP, surfactant protein; UIP, usual interstitial pneumonia.

involving the production, processing, and clearance surfactant dysfunction disorders, patients with muta-
of surfactant has been recognized as an important tions in SFTPC display the broadest range in age of
cause of neonatal and pediatric respiratory illness. onset, with affected neonates to older adults.
This group, collectively referred to as surfactant dys-
function disorders,155 encompasses a variety of muta- Surfactant Metabolism Dysfunction, Type 1:
tions involving the genes that encode SP-B (SFTPB), SFTPB Mutations
SP-C (SFTPC), the ATP-binding cassette transporter SP-B deficiency is inherited in an autosomal reces-
A3 (ABCA3), and the receptor for granulocyte-mac- sive manner, and the majority of affected patients
rophage colony–stimulating factor (CSF2RA and develop respiratory failure in the neonatal period
CSF2RB). ABCA3 deficiency is the most prevalent for with rapid progression of disease and death at
the surfactant disorders that present in the neonatal 3–6 months.156–161 A few mutations that seem to con-
period with severe respiratory distress. Mutations in fer a milder phenotype have been found. Children
SFTPB, SFTPC, and NKX2-1 are more rare. Of all the with these mutations have partial expression of the
CHAPTER 1 Genetic Interstitial Lung Disease 11

SP-B protein, survive longer, and go on to develop a in ABCA3 are the most common inherited defects in
chronic ILD.162,163 surfactant metabolism presenting either with severe
neonatal disease or as diffuse lung disease in infancy
Surfactant Metabolism Dysfunction, Type 2: or childhood.155,174,179,180 ABCA3 is an ATP-binding
SFTPC Mutations cassette transporter that localizes to the limiting mem-
Autosomal dominant lung disease due to mutations in brane of lamellar bodies where it functions in translo-
the gene encoding SP-C, SFTPC, was first described in cating phospholipids, primarily phosphatidylcholine,
2001 by Nogee and colleagues.164 The index case was a into these organelles for assembly and storage of surfac-
full-term infant who developed respiratory symptoms tant in type II AECs.181,182 The functional consequences
at 6 weeks of age. An open lung biopsy revealed his- of the ABCA3 mutations include decreased expression
tologic features of NSIP. The child’s mother had been of the protein, abnormal localization of the protein to
diagnosed with desquamative interstitial pneumonitis the lamellar membrane, production of surfactant that
at 1 year of age, and the child’s maternal grandfather is deficient in phosphatidylcholine, and increased alve-
had died from lifelong respiratory illness of unknown olar surface tension.183–185
cause. Both the mother and her infant had a hetero-
zygous mutation at the splice donor site in intron 4 Surfactant Metabolism Dysfunction, Types
(c.460+1G > A), which resulted in skipping of exon 4 4 and 5: CSF2RA and CSF3RB Mutations
and deletion of 37 amino acids.164 In the next year, a Protein alveolar proteinosis (PAP) is a rare form of
large five-generation kindred was described with 14 lung disease characterized by intraalveolar accumula-
affected family members.165 The age at diagnosis for the tion of surfactant, which results in respiratory insuffi-
affected individuals ranged from 4 months to 57 years ciency. Histopathology specimens from affected patients
and included four adults with surgical lung biopsy evi- demonstrate distal airspaces filled with foamy alveolar
dence of usual interstitial pneumonitis and three chil- macrophages and a granular, eosinophilic material that
dren with NSIP. Genomic sequencing revealed a rare stains positively with PAS reagent. In general, the under-
heterozygous missense SP-C mutation (L188Q) in all lying lung architecture is normal unless infection is pres-
analyzed individuals. Human type II alveolar epithelial ent.186 When the accumulated surfactant is removed, as
cells from one of the patients and mouse lung epithe- is done with whole-lung lavage, the gas exchange proper-
lial cells producing the mutant SP-C protein showed ties of the lung improve.186 Approximately 90% of PAP
abnormal lamellar bodies. Since these two studies, over cases are acquired and are referred to as primary PAP.187–
30 different mutations in SFTPC have been identified in 189 Circulating autoantibodies to GM-CSF can be found

children and adults. A missense mutation (c.218T > C) in over 92% of patients with primary PAP.190–193 These
that changes a conserved isoleucine at position 73 to antibodies block GM-CSF signaling in vivo, reduce
a threonine is one of the more common and accounts alveolar macrophage surfactant catabolism, and impair
for 25–35% of abnormal alleles.155,166–171 There is surfactant clearance.186,193–196 Secondary PAP occurs in
incomplete penetrance of the ILD phenotype and phe- several different clinical settings, such as in association
notypic heterogeneity. The disease ranges from severe with hematologic malignancies, immunosuppression,
respiratory distress in infants to IPF in older adults. inhalation of inorganic dusts, and certain infections.
Mutations of SFTPC are rare in individuals without a The GM-CSF receptor is composed of an α- and a
family history of pulmonary fibrosis.171–173 At present, β-chain, encoded by the CSF2RA and CSF2RB genes,
the disease phenotype is thought to be caused by aber- respectively.197 Mutations in both CSF2RA and CSF2RB
rant protein-folding, which elicits the unfolded protein have been identified in children with PAP.198,199 In
response, ER stress, and apoptosis of alveolar epithelial 2011, a case of adult-onset hereditary PAP was reported
cells.155,174–178 in Japan.200 Here, a 36-year-old woman with PAP had
elevated circulating levels of GM-CSF, no measurable
Surfactant Metabolism Dysfunction, Type 3: anti-GM-CSF antibodies, and reduced expression of the
ABCA3 Mutations GM-CSF receptor β-chain. Genetic sequencing revealed
There is a significant amount of phenotypic overlap a homozygous, single-base deletion at nucleotide 631 in
between patients with SFTPB mutations and those with exon 6 of CSF2RB. Both of the patient’s parents were het-
defects in ABCA3. Like SP-B deficiency, the disease is erozygous for the mutation. Hereditary PAP follows an
inherited in an autosomal recessive manner. Since autosomal recessive pattern. Mice with deletion of the
the first mutation was discovered in 2004, more than gene encoding GM-CSF or its receptor demonstrate a phe-
200 different mutations have been found. Mutations notype very similar to adults with primary PAP.186,201–204
12 Interstitial Lung Disease

Interstitial Lung and Liver Disease biopsies.210,218 While approximately half of patients
A specific and severe type of PAP has been described carry a diagnosis of idiopathic pulmonary fibrosis (IPF),
affecting infants and children on Réunion Island.205 others have diagnoses of another idiopathic interstitial
Since 1970, 34 children have been diagnosed and pneumonia, unclassifiable lung fibrosis, chronic hyper-
treated, giving rise to an incidence of disease of 1 in at sensitivity pneumonitis, pleuroparenchymal fibroelas-
least 10,000 newborns. The lung disease progresses to tosis, idiopathic fibrosis with autoimmune features, or,
lung fibrosis despite regular whole-lung lavage. It has rarely, a connective tissue disease–associated ILD.218
recently been found that patients have biallelic muta- Many TERT mutation carriers with pulmonary fibro-
tions in MARS, the gene encoding methionine tRNA sis report past cigarette smoking or an exposure to a
ligase.205 Compound heterozygous mutations in the fibrogenic environmental insult, suggesting that injuri-
same gene have been found in one patient with mul- ous environmental exposures in conjunction with the
tiorgan disease, predominated by liver failure.206 The underlying inherited genetic predisposition lead to the
increased prevalence of disease on Réunion Island and lung disease. Disease progression is inexorable, with a
in nearby Tunisia and France is due to founder muta- mean transplant-free survival of <3 years after diagno-
tions. A potential benefit of high-dose methionine sis. TERT mutation patients undergoing lung transplant
supplementation has not yet been studied in these generally have a higher risk of cytopenias and other
patients. extrapulmonary complications.219–221
Genetic anticipation is seen for TERT mutation
Pulmonary Fibrosis, Telomere Related, patients, with an earlier age of diagnosis of lung fibrosis
Type 1 in subsequent generations.218 Similar to DC, progres-
Heterozygous mutations in the gene encoding the pro- sively shorter telomere lengths are found in subse-
tein component (TERT) of telomerase have been found quent generations of TERT mutation carriers, and these
in ∼15% of patients with autosomal dominant famil- shorter telomere lengths explain genetic anticipation
ial pulmonary fibrosis.207,208 The frequency of TERT seen in kindreds.36,210,218
mutations in patients with sporadic adult-onset ILD is
less common.208,209 Telomerase is a multimeric ribo- Pulmonary Fibrosis, Telomere Related,
nucleoprotein enzyme that catalyzes the addition of a Type 2
repetitive DNA sequence to the ends of chromosomes Heterozygous mutations in the gene encoding the RNA
known as telomeres. Each of the mutations is rare and component (TERC) of telomerase have been found in
associated with decreased in vitro activity of telomer- ∼3% of patients with autosomal dominant familial pul-
ase and short leukocyte telomere lengths. TERT muta- monary fibrosis.207,208 Each of the mutations is rare and
tions are the most common genetic mutations found associated with decreased in vitro activity of telomerase
in adult patients with ILD.210 In addition, adult-onset and short leukocyte telomere lengths. A wide range of
pulmonary fibrosis is the most common manifestation progressive adult-onset pulmonary fibrosis subtypes
of TERT mutations.211 Other clinical manifestations of are seen in patients with TERC mutations.218 Patients
TERT mutations include DC (see earlier discussion), with TERC mutations are generally diagnosed with ILD
aplastic anemia and other forms of bone marrow fail- at an earlier age (51 years) than TERT mutation carri-
ure,212 liver disease including liver cirrhosis,213,214 early ers (58 years). In addition, they have a higher incidence
graying of hair,215 and an increased risk for myelodys- of hematologic comorbidities, especially leukopenia,
plastic syndrome and acute myeloid leukemia.216,217 thrombocytopenia, aplastic anemia, or myelodysplas-
Altogether, disparate clinical presentations and dis- tic syndrome.
eases linked together by defects in telomere-related
genes and characterized by short telomere lengths are Pulmonary Fibrosis, Telomere Related,
known as monogenic short telomere syndromes or Type 3
telomeropathies. Autosomal dominant pulmonary fibrosis with incom-
The penetrance of pulmonary fibrosis in TERT plete penetrance has been linked to rare mutations
mutation carriers is age and gender dependent.210 The in RTEL1.222–224 RTEL1 surpassed the threshold for
penetrance of pulmonary fibrosis for men aged 40–49, genomewide significance when comparing the num-
50–59, and >60 years is 14%, 38%, and 60%, respec- ber of observed versus expected novel damaging or
tively. Women show the same age-dependent increase conserved missense mutations in familial pulmonary
in penetrance. Microscopic honeycombing and fibro- fibrosis cases and controls.223 Across independent
blastic foci are commonly found in surgical lung studies, RTEL1 mutations are rare and associated with
CHAPTER 1 Genetic Interstitial Lung Disease 13

short leukocyte telomere lengths. This gene encodes Pulmonary Fibrosis, Other Telomere Related
the regulator of telomere elongation helicase 1 and has Genes found to be mutated in DC patients are candidate
a known role in telomere maintenance. Mutations in genes for patients with adult-onset pulmonary fibrosis and
RTEL1 were previously shown to cause Hoyeraal-Hre- short telomere lengths. Similar to the other short telomere
idarsson syndrome, a severe variant of DC (see earlier syndromes mentioned earlier, many individuals have clin-
discussion), in which affected children generally have ical features that overlap with DC, including bone marrow
very short telomere lengths and biallelic mutations. In abnormalities and skin changes. A missense mutation in
contrast, those affected with adult-onset pulmonary the DKC1, encoding dyskerin, has been reported in a kin-
fibrosis are heterozygous for rare missense or loss-of- dred with two affected older males with interstitial fibro-
function RTEL1 mutations. Similar to patients with sis, hyperpigmented skin changes, dyskeratotic nails, and
TERT mutations, ∼50% of heterozygous RTEL1 muta- macrocytic anemia.225 Blood leukocyte telomere lengths
tion carriers have hematologic manifestations such as were short. The expression of hTR in a lymphoblastoid
anemia or macrocytosis.218 A wide range of progressive cell line derived from the proband was lower than that
adult-onset pulmonary fibrosis subtypes are seen in of controls, even though expression of DKC1 was not
patients with RTEL1 mutations. Genetic anticipation is reduced. Female mutation carriers were unaffected.
also seen in RTEL1 kindreds. TINF2 encodes one of the shelterin complex pro-
teins that functions to protect the telomeric ends of
Pulmonary Fibrosis, Telomere Related, Type 4 chromosomes. Exome sequencing revealed a complex
Exome sequencing has linked autosomal dominant spice acceptor site and missense mutation on the same
pulmonary fibrosis with incomplete penetrance to TINF2 allele, with predominant expression of the mis-
mutations in PARN, the gene encoding a polyadenyl- sense mutation in lung-derived DNA from a female
ate-specific ribonuclease.223 PARN surpassed the thresh- patient with pulmonary fibrosis and infertility.226
old for genomewide significance when comparing the Heterozygous loss of function mutations in NAF1
number of observed versus expected novel damaging have most recently been linked to pulmonary fibrosis-
mutations in cases and controls.223 Most mutations are emphysema, short telomere lengths, bone marrow fail-
predicted to cause protein loss of function (splice site, ure, and liver disease.226a NAF1 has been shown to be
nonsense, and frameshift). One large kindred was iden- essential for the biogenesis of telomerase RNA.
tified by linking together two smaller and previously
unknowingly related kindreds with familial pulmonary Pulmonary Fibrosis and Adenocarcinoma,
fibrosis and an identical splice-site PARN mutation. The Related to Surfactant A Mutations
kinship of the two families was confirmed by demon- Novel missense mutations in the gene encoding SP-A2
strating that the two probands share ∼6% of their over- (SFTPA2) were discovered by genomewide linkage
all genome, including the genomic segment on which followed by sequencing candidate genes within the
the PARN mutation is located. As a further test for the linked region.227 Affected individuals have evidence of
relevance of the PARN mutations, the cosegregation of pulmonary fibrosis and/or lung adenocarcinoma with
the mutations with pulmonary fibrosis was compared features of bronchoalveolar cell carcinoma. A second
across extended kindreds. The overall backward LOD study reported three additional rare SFTPA2 mutations
score across all informative PARN kindreds was 3.6, in patients with adult-onset pulmonary fibrosis and
reflecting an odds ratio of 1:4096 in favor of linkage. a personal or family history of adenocarcinoma.228 A
Other clinical manifestations of PARN mutations third study has recently described the cosegregation
include DC (see earlier discussion), in which biallelic of ILD and lung adenocarcinoma in an extended kin-
mutations in the gene are frequently found.52–54 Patient dred with a germline mutation in SFTPA1 that is pre-
cells show deficiencies in trimming of small nucleolar dicted to change a conserved tryptophan at position
RNAs, including abnormally adenylated hTR.54 In general, 211 (Trp211Arg).229 While most of the affected indi-
leukocyte telomere lengths of PARN mutation patients are viduals in the SFTPA1 kindred were affected with an
shorter than in controls, but longer than in subjects with ILD after age 30 (range 31–69 years at diagnosis), one
TERT, TERC, or RTEL1 mutations.223 Patients with PARN was diagnosed at 7 months of age and died of respira-
mutations are generally diagnosed with ILD at an older tory failure 2 months later.229 Thus, a wide range of age
age (64 years) than TERT mutation carriers (58 years).218 at the time of diagnosis is observed, from respiratory
However, once pulmonary fibrosis is diagnosed, the respi- insufficiency in infancy to lung fibrosis and adenocar-
ratory disease progresses and patients have a mean trans- cinoma in adulthood. All known mutations in the sur-
plant-free survival of 5.7 years from diagnosis. factant A genes affect highly conserved residues in the
14 Interstitial Lung Disease

carbohydrate recognition domain of the proteins. Two lead to increased susceptibility to lung fibrosis through
of the mutations are predicted to alter the same glycine its effects on increased mucin expression and ER stress.
at amino acid position 231 (Gly231Val, Gly231Arg). The GWAS study confirmed the association of IPF
Cells expressing mutant proteins fail to secrete with variants in the TERT237 and TERC genes, with meta-
mature protein into the culture media.227,229 In addi- analysis p values of 1.7 × 10−19 and 4.5 × 10−8, respec-
tion, expression of these mutant proteins led to fewer tively.232 Overall, common variants in three telomere
intracellular oligomers, greater protein instability, and length–related genes (TERT, TERC, and OBFC1)238–240
increased markers of ER stress.230 Family members were found to be associated with lung fibrosis in this
with these heterozygous mutations secrete comparable study. The GWAS study also found seven new loci that
amounts of total SP-A into the alveolar space, as com- reached genomewide statistical significance.232 These
pared with control family members, suggesting that the loci are located near genes involved with host defense,
pathogenic mechanism may be related to ER stress of cell-cell adhesion, and DNA repair.
the resident epithelial alveolar cells.230
Pulmonary Alveolar Microlithiasis
Susceptibility to Pulmonary Fibrosis, Pulmonary alveolar microlithiasis (PAM) is a rare dis-
Common Variants order characterized by laminated calcium phosphate
Using a genomewide screen, a common variant located concretions within the alveoli. PAM is inherited in an
3 kb upstream of the MUC5B transcription start site autosomal recessive manner and is particularly prevalent
(rs35705950) was found to be present at a higher fre- in Turkey, Italy, the United States, and Japan.241 The chest
quency in IPF patients. This variant is found in ∼9% of radiographs of patients with PAM show diffuse, bilateral,
controls (an overall allele frequency of 0.10) and 38% of micronodular opacities, which obscure the heart border,
patients with IPF. The odds ratio for IPF disease among mediastinum, and diaphragmatic surfaces. Despite the
subjects who are heterozygous or homozygous for the “sandstormlike” appearance, the clinical presentation
minor allele of this single nucleotide polymorphism of PAM is variable.242–245 Early in the disease course,
(SNP) was 9.0 (CI 6.2–13.1) and 21.8 (5.1–93.5), patients are often asymptomatic, but over time develop
respectively.231 The variant allele was associated with cough, dyspnea, and a restrictive defect with reduced
upregulation of MUC5B expression (up to 37.4 times diffusion. In 2006, Corut et al. reported the discovery
as high when compared with those homozygous for the of several mutations in the gene encoding the type IIb
wild-type allele) in lung tissue of unaffected subjects.231 sodium-phosphate cotransporter protein (SCL34A2)
A genomewide association study (GWAS) was per- in individuals with PAM.246 The group used linkage
formed comparing the frequency of common SNPs in analysis of a large consanguineous family in which six
patients with fibrotic idiopathic interstitial pneumonia members were affected by PAM, and ultimately identi-
(n = 1616) with controls (n = 4683). A replication anal- fied six homozygous mutations in SCL34A2 that predict
ysis included 876 cases and 1890 controls. A MUC5B loss of protein function. A separate group in Japan used
promoter variant (rs868903) was again found to be sig- genomewide SNP mapping to identify candidate genes
nificantly associated with fibrotic lung disease, with a in six patients with PAM, and they independently identi-
metaanalysis P value of 9.2 × 10−26.232 A broad region fied SCL34A2 as a gene of interest.247 Two patients had
on 11p15, including the MUC5B, MUC2, and TOLLIP homozygous frameshift mutations and four had splice-
genes, was found to demonstrate genomewide signifi- site mutations of the gene. None of the mutations was
cance. After adjusting for the MUC5B promoter SNP identified in normal controls. Since these initial discov-
found in the earlier study (rs35705950),231 most of the eries, additional missense and frameshift mutations, as
variants in this broad region were no longer significantly well as intragenetic deletions, have been identified.248–250
associated, suggesting that the associations seen for the The gene is expressed in high levels in the lung, pre-
other SNPs may be due to linkage disequilibrium. The dominantly in type II epithelial cells.251–253 Epithelial
MUC5B promoter (rs35705950) has been found in the deletion of the mouse Npt2b gene in the lung leads to
Framingham Heart Study233 to be associated with inter- a progressive pulmonary process characterized by diffuse
stitial lung abnormalities, thus linking it to an early alveolar microliths, restrictive physiology, and alveolar
manifestation of IPF. Mucins undergo a complex matu- phospholipidosis.254 Microliths are readily dissolved by
ration process in airway cells, with glycosylation and whole-lung EDTA lavage. A low-phosphate diet prevents
disulfide multimerization, before secretion. As genetic microlith formation in young mice and decreases the
perturbations leading to increased mucin production burden of pulmonary calcium deposits in older mice.
cause elevated ER stress signaling,234–236 it is interest- The effectiveness of such a diet has not yet been studied
ing to speculate that the MUC5B promoter variant may in human patients.
CHAPTER 1 Genetic Interstitial Lung Disease 15

TABLE 1.3
Genetic Interstitial Lung Disease: Pathogenesis
Pathway Surfactant Metabolism ER Stress Telomere Shortening
Rare variants NKX2.1 SFTPC TERT, TERC
ABCA3 SFTPA1, SFTPA2 RTEL1
SFTPB HPS1, HPS2, HPS4 PARN
SFTPC COPA DKC1, TINF2
SFTPA1, SFTPA2 Other DC genes
CSF2RA, SFT2RB
Common variants MUC5B? TERT, TERC
OBFC1

DC, dyskeratosis congenita; ER, endoplasmic reticulum.

CONCLUSION identification of genes relevant to the molecular patho-


Discoveries of the genetic underpinnings of ILD have genesis of more common manifestations of ILD. How
demonstrated a few consistent themes. Many of the this knowledge leads to effective treatments across the
genes involve common pathways that lead to altered spectrum of genetic ILD will be the next grand opportu-
surfactant metabolism, increased ER stress signaling, nity to improve patient care.
and telomere shortening (Table 1.3). Mutations, or
pathologic ultrarare variants, have been found in genes
involved with the metabolism of pulmonary surfac- REFERENCES
tant. Given the central role surfactant has for reduc- 1. Avila NA, Dwyer AJ, Rabel A, Moss J. Sporadic lym-
ing surface tension of the lung, opsonizing respiratory phangioleiomyomatosis and tuberous sclerosis complex
pathogens, and regulating inflammatory responses with lymphangioleiomyomatosis: comparison of CT fea-
in the alveolar space, it is not surprising that patients tures. Radiology. 2007;242(1):277–285.
2. Lenoir S, Grenier P, Brauner MW, et al. Pulmonary lym-
with mutations in these genes have a wide range of ILD
phangiomyomatosis and tuberous sclerosis: comparison
presentations. of radiographic and thin-section CT findings. Radiology.
Another common pathway, increased ER stress sig- 1990;175(2):329–334.
naling, has been elucidated by genetic variants linked 3. Curatolo P, Bombardieri R, Jozwiak S. Tuberous sclero-
to various ILD subtypes.255,256 Production of misfolded sis. Lancet. 2008;372(9639):657–668.
(missense) surfactant proteins in type 2 alveolar epithe- 4. Henske EP, Wessner LL, Golden J, et al. Loss of tuber-
lial cells leads to increased ER stress. Mutations in the in in both subependymal giant cell astrocytomas and
HPS and COPA genes alter intracellular protein trans- angiomyolipomas supports a two-hit model for the
port and also upregulate ER stress pathways and lead pathogenesis of tuberous sclerosis tumors. Am J Pathol.
to lung fibrosis. As the common variant located in the 1997;151(6):1639–1647.
5. Carsillo T, Astrinidis A, Henske EP. Mutations in the tu-
MUC5B promoter increases gene expression, this SNP
berous sclerosis complex gene TSC2 are a cause of spo-
may lead to increased susceptibility to lung fibrosis radic pulmonary lymphangioleiomyomatosis. Proc Natl
through its effects on increased mucin expression and Acad Sci USA. 2000;97(11):6085–6090.
increased ER stress. Finally, many different rare and 6. Sato T, Seyama K, Fujii H, et al. Mutation analysis of
common variants linked to genetic ILD share a common the TSC1 and TSC2 genes in Japanese patients with
pathway of telomere shortening. Pulmonary fibrosis has pulmonary lymphangioleiomyomatosis. J Hum Genet.
been estimated to be the most common manifestation 2002;47(1):20–28.
of the monogenic short telomere syndromes, and SNPs 7. Smolarek TA, Wessner LL, McCormack FX, Mylet JC,
in three telomere-related genes reach genomewide sig- Menon AG, Henske EP. Evidence that lymphangiomy-
nificance in a large GWAS study. The link between telo- omatosis is caused by TSC2 mutations: chromosome
16p13 loss of heterozygosity in angiomyolipomas and
mere attrition and lung fibrosis is further strengthened
lymph nodes from women with lymphangiomyomato-
by the finding that telomere length predicts transplant- sis. Am J Hum Genet. 1998;62(4):810–815.
free survival in patients with sporadic (noninherited) 8. Henske EP, McCormack FX. Lymphangioleiomy-
IPF across independent cohorts.257,258 Thus, the study omatosis – a wolf in sheep’s clothing. J Clin Invest.
of rare human patients and families has led to the 2012;122(11):3807–3816.
16 Interstitial Lung Disease

9. Bissler JJ, McCormack FX, Young LR, et al. Siroli- 25. de la Fuente J, Dokal I. Dyskeratosis congenita: advances
mus for angiomyolipoma in tuberous sclerosis com- in the understanding of the telomerase defect and the
plex or lymphangioleiomyomatosis. N Engl J Med. role of stem cell transplantation. Pediatr Transplant.
2008;358(2):140–151. 2007;11(6):584–594.
10. Morton JM, McLean C, Booth SS, Snell GI, Whitford HM. 26. Giri N, Lee R, Faro A, et al. Lung transplantation for
Regression of pulmonary lymphangioleiomyomatosis pulmonary fibrosis in dyskeratosis congenita: Case Re-
(PLAM)-associated retroperitoneal angiomyolipoma port and systematic literature review. BMC Blood Disord.
post-lung transplantation with rapamycin treatment. J 2011;11:3.
Heart Lung Transplant. 2008;27(4):462–465. 27. Imokawa S, Sato A, Toyoshima M, et al. Dyskeratosis
11. Krueger DA, Care MM, Holland K, et al. Everolimus for congenita showing usual interstitial pneumonia. Intern
subependymal giant-cell astrocytomas in tuberous scle- Med. 1994;33(4):226–230.
rosis. N Engl J Med. 2010;363(19):1801–1811. 28. Verra F, Kouzan S, Saiag P, Bignon J, de Cremoux H.
12. McCormack FX, Inoue Y, Moss J, et al. Efficacy and safety Bronchoalveolar disease in dyskeratosis congenita. Eur
of sirolimus in lymphangioleiomyomatosis. N Engl J Respir J. 1992;5(4):497–499.
Med. 2011;364(17):1595–1606. 29. Parry EM, Alder JK, Qi X, Chen JJ, Armanios M. Syn-
13. Hornstein OP, Knickenberg M. Perifollicular fi- drome complex of bone marrow failure and pulmonary
bromatosis cutis with polyps of the colon–a cutaneo- fibrosis predicts germline defects in telomerase. Blood.
intestinal syndrome sui generis. Arch Dermatol Res. 2011;117(21):5607–5611.
1975;253(2):161–175. 30. Yabe M, Yabe H, Hattori K, et al. Fatal interstitial pul-
14. Toro JR, Pautler SE, Stewart L, et al. Lung cysts, spon- monary disease in a patient with dyskeratosis congenita
taneous pneumothorax, and genetic associations in 89 after allogeneic bone marrow transplantation. Bone Mar-
families with Birt-Hogg-Dube syndrome. Am J Respir Crit row Transplant. 1997;19(4):389–392.
Care Med. 2007;175(10):1044–1053. 31. Rocha V, Devergie A, Socie G, et al. Unusual complica-
15. Toro JR, Wei MH, Glenn GM, et al. BHD mutations, clin- tions after bone marrow transplantation for dyskeratosis
ical and molecular genetic investigations of Birt-Hogg- congenita. Br J Haematol. 1998;103(1):243–248.
Dube syndrome: a new series of 50 families and a review 32. Alter BP, Baerlocher GM, Savage SA, et al. Very short tel-
of published reports. J Med Genet. 2008;45(6):321–331. omere length by flow FISH identifies patients with Dys-
16. Nickerson ML, Warren MB, Toro JR, et al. Mutations in a keratosis Congenita. Blood. 2007;110(5):1439–1447.
novel gene lead to kidney tumors, lung wall defects, and 33. Heiss NS, Knight SW, Vulliamy TJ, et al. X-linked dys-
benign tumors of the hair follicle in patients with the keratosis congenita is caused by mutations in a highly
Birt-Hogg-Dube syndrome. Cancer Cell. 2002;2(2):157– conserved gene with putative nucleolar functions. Nat
164. Genet. 1998;19(1):32–38.
17. Painter JN, Tapanainen H, Somer M, Tukiainen P, Ait- 34. Cohen SB, Graham ME, Lovrecz GO, Bache N, Robin-
tomaki KA. 4-bp deletion in the Birt-Hogg-Dube gene son PJ, Reddel RR. Protein composition of catalytically
(FLCN) causes dominantly inherited spontaneous pneu- active human telomerase from immortal cells. Science.
mothorax. Am J Hum Genet. 2005;76(3):522–527. 2007;315(5820):1850–1853.
18. Graham RB, Nolasco M, Peterlin B, Garcia CK. Nonsense 35. Vulliamy T, Marrone A, Goldman F, et al. The RNA com-
mutations in folliculin presenting as isolated familial ponent of telomerase is mutated in autosomal domi-
spontaneous pneumothorax in adults. Am J Respir Crit nant dyskeratosis congenita. Nature. 2001;413(6854):
Care Med. 2005;172(1):39–44. 432–435.
19. Agarwal PP, Gross BH, Holloway BJ, Seely J, Stark P, 36. Armanios M, Chen JL, Chang YP, et al. Haploinsuf-
Kazerooni EA. Thoracic CT findings in Birt-Hogg-Dube ficiency of telomerase reverse transcriptase leads to
syndrome. AJR Am J Roentgenol. 2011;196(2):349–352. anticipation in autosomal dominant dyskeratosis con-
20. Tobino K, Gunji Y, Kurihara M, et al. Characteristics of genita. Proc Natl Acad Sci USA. 2005;102(44):15960–
pulmonary cysts in Birt-Hogg-Dube syndrome: thin- 15964.
section CT findings of the chest in 12 patients. Eur J 37. Du HY, Pumbo E, Manley P, et al. Complex inheritance
Radiol. 2011;77(3):403–409. pattern of dyskeratosis congenita in two families with
21. Devine MS, Garcia CK. Genetic interstitial lung disease. 2 different mutations in the telomerase reverse tran-
Clin Chest Med. 2012;33(1):95–110. scriptase gene. Blood. 2008;111(3):1128–1130.
22. Drachtman RA, Alter BP. Dyskeratosis congenita. Derma- 38. Aspesi A, Vallero S, Rocci A, et al. Compound heterozy-
tol Clin. 1995;13(1):33–39. gosity for two new TERT mutations in a patient with
23. Dokal I. Dyskeratosis congenita in all its forms. Br J Hae- aplastic anemia. Pediatr Blood Cancer. 2010;55(3):550–
matol. 2000;110(4):768–779. 553.
24. Zinsser F. Atrophia Cutis Reticularis cum Pigmentations, 39. Savage SA, Giri N, Baerlocher GM, Orr N, Lansdorp PM,
Dystrophia Unguium et Leukoplakis oris (Poikiooder- Alter BP. TINF2, a component of the shelterin telomere
mia atrophicans vascularis Jacobi). Ikonogr Dermatol. protection complex, is mutated in dyskeratosis congen-
1910;5:219–223. ita. Am J Hum Genet. 2008;82(2):501–509.
CHAPTER 1 Genetic Interstitial Lung Disease 17

40. Walne AJ, Vulliamy T, Beswick R, Kirwan M, Dokal I. 55. Vulliamy T, Marrone A, Szydlo R, Walne A, Mason PJ,
TINF2 mutations result in very short telomeres: analysis Dokal I. Disease anticipation is associated with pro-
of a large cohort of patients with dyskeratosis congen- gressive telomere shortening in families with dyskera-
ita and related bone marrow failure syndromes. Blood. tosis congenita due to mutations in TERC. Nat Genet.
2008;112(9):3594–3600. 2004;36(5):447–449.
41. Guo Y, Kartawinata M, Li J, et al. Inherited bone mar- 56. Goldman F, Bouarich R, Kulkarni S, et al. The effect of TERC
row failure associated with germline mutation of haploinsufficiency on the inheritance of telomere length.
ACD, the gene encoding telomere protein TPP1. Blood. Proc Natl Acad Sci USA. 2005;102(47):17119–17124.
2014;124(18):2767–2774. 57. Devgan SS, Sanal O, Doil C, et al. Homozygous deficien-
42. Kocak H, Ballew BJ, Bisht K, et al. Hoyeraal-Hreidarsson syn- cy of ubiquitin-ligase ring-finger protein RNF168 mim-
drome caused by a germline mutation in the TEL patch of ics the radiosensitivity syndrome of ataxia-telangiectasia.
the telomere protein TPP1. Genes Dev. 2014;28(19):2090– Cell Death Differ. 2011;18(9):1500–1506.
2102. 58. Stewart GS, Stankovic T, Byrd PJ, et al. RIDDLE immu-
43. Keller RB, Gagne KE, Usmani GN, et al. CTC1 Mutations nodeficiency syndrome is linked to defects in 53BP1-
in a patient with dyskeratosis congenita. Pediatr Blood mediated DNA damage signaling. Proc Natl Acad Sci
Cancer. 2012;59(2):311–314. USA. 2007;104(43):16910–16915.
44. Walne AJ, Bhagat T, Kirwan M, et al. Mutations in the 59. Doil C, Mailand N, Bekker-Jensen S, et al. RNF168 binds
telomere capping complex in bone marrow failure and and amplifies ubiquitin conjugates on damaged chro-
related syndromes. Haematologica. 2013;98(3):334–338. mosomes to allow accumulation of repair proteins. Cell.
45. Walne AJ, Vulliamy T, Marrone A, et al. Genetic heterogene- 2009;136(3):435–446.
ity in autosomal recessive dyskeratosis congenita with one 60. Stewart GS, Panier S, Townsend K, et al. The RIDDLE syn-
subtype due to mutations in the telomerase-associated pro- drome protein mediates a ubiquitin-dependent signaling
tein NOP10. Hum Mol Genet. 2007;16(13):1619–1629. cascade at sites of DNA damage. Cell. 2009;136(3):420–
46. Trahan C, Martel C, Dragon F. Effects of dyskeratosis 434.
congenita mutations in dyskerin, NHP2 and NOP10 61. Okamoto K, Bartocci C, Ouzounov I, Diedrich JK,
on assembly of H/ACA pre-RNPs. Hum Mol Genet. Yates 3rd JR, Denchi EL. A two-step mechanism for
2010;19(5):825–836. TRF2-mediated chromosome-end protection. Nature.
47. Vulliamy T, Beswick R, Kirwan M, et al. Mutations in the 2013;494(7438):502–505.
telomerase component NHP2 cause the premature age- 62. Hermansky F, Pudlak P. Albinism associated with hem-
ing syndrome dyskeratosis congenita. Proc Natl Acad Sci orrhagic diathesis and unusual pigmented reticular cells
USA. 2008;105(23):8073–8078. in the bone marrow: report of two cases with histochem-
48. Zhong F, Savage SA, Shkreli M, et al. Disruption of tel- ical studies. Blood. 1959;14(2):162–169.
omerase trafficking by TCAB1 mutation causes dyskera- 63. Gahl WA, Brantly M, Kaiser-Kupfer MI, et al. Genetic de-
tosis congenita. Genes Dev. 2011;25(1):11–16. fects and clinical characteristics of patients with a form
49. Walne AJ, Vulliamy T, Kirwan M, Plagnol V, Dokal I. of oculocutaneous albinism (Hermansky-Pudlak syn-
Constitutional mutations in RTEL1 cause severe dyskera- drome). N Engl J Med. 1998;338(18):1258–1264.
tosis congenita. Am J Hum Genet. 2013;92(3):448–453. 64. Witkop CJ, Krumwiede M, Sedano H, White JG. Reliabil-
50. Le Guen T, Jullien L, Touzot F, et al. Human RTEL1 de- ity of absent platelet dense bodies as a diagnostic cri-
ficiency causes Hoyeraal-Hreidarsson syndrome with terion for Hermansky-Pudlak syndrome. Am J Hematol.
short telomeres and genome instability. Hum Mol Genet. 1987;26(4):305–311.
2013;22(16):3239–3249. 65. Dell’Angelica EC, Shotelersuk V, Aguilar RC, Gahl WA,
51. Ballew BJ, Yeager M, Jacobs K, et al. Germline mutations Bonifacino JS. Altered trafficking of lysosomal proteins
of regulator of telomere elongation helicase 1, RTEL1, in in Hermansky-Pudlak syndrome due to mutations
Dyskeratosis congenita. Hum Genet. 2013;132(4):473– in the beta 3A subunit of the AP-3 adaptor. Mol Cell.
480. 1999;3(1):11–21.
52. Tummala H, Walne A, Collopy L, et al. Poly(A)-specific 66. Morgan NV, Pasha S, Johnson CA, et al. A germline mu-
ribonuclease deficiency impacts telomere biology and tation in BLOC1S3/reduced pigmentation causes a novel
causes dyskeratosis congenita. J Clin Invest. 2015;125(5): variant of Hermansky-Pudlak syndrome (HPS8). Am J
2151–2160. Hum Genet. 2006;78(1):160–166.
53. Dhanraj S, Gunja SM, Deveau AP, et al. Bone marrow 67. Martina JA, Moriyama K, Bonifacino JS. BLOC-3, a
failure and developmental delay caused by mutations protein complex containing the Hermansky-Pudlak
in poly(A)-specific ribonuclease (PARN). J Med Genet. syndrome gene products HPS1 and HPS4. J Biol Chem.
2015;52(11):738–748. 2003;278(31):29376–29384.
54. Moon DH, Segal M, Boyraz B, et al. Poly(A)-specific 68. Suzuki T, Li W, Zhang Q, et al. The gene mutated in
ribonuclease (PARN) mediates 3’-end maturation of the cocoa mice, carrying a defect of organelle biogenesis,
telomerase RNA component. Nat Genet. 2015;47(12): is a homologue of the human Hermansky-Pudlak syn-
1482–1488. drome-3 gene. Genomics. 2001;78(1–2):30–37.
18 Interstitial Lung Disease

69. Zhang Q, Zhao B, Li W, et al. Ru2 and Ru encode 83. Guttentag SH, Akhtar A, Tao JQ, et al. Defective surfactant
mouse orthologs of the genes mutated in human secretion in a mouse model of Hermansky-Pudlak syn-
Hermansky-Pudlak syndrome types 5 and 6. Nat Genet. drome. Am J Respir Cell Mol Biol. 2005;33(1):14–21.
2003;33(2):145–153. 84. Gras D, Jonard L, Roze E, et al. Benign hereditary chorea:
70. Li W, Zhang Q, Oiso N, et al. Hermansky-Pudlak syn- phenotype, prognosis, therapeutic outcome and long
drome type 7 (HPS-7) results from mutant dysbindin, term follow-up in a large series with new mutations in
a member of the biogenesis of lysosome-related orga- the TITF1/NKX2-1 gene. J Neurol Neurosurg Psychiatry.
nelles complex 1 (BLOC-1). Nat Genet. 2003;35(1): 2012;83(10):956–962.
84–89. 85. Young LR, Deutsch GH, Bokulic RE, Brody AS, Nogee LM.
71. Wildenberg SC, Oetting WS, Almodovar C, Krumwiede M, A mutation in TTF1/NKX2.1 is associated with famil-
White JG, King RA. A gene causing Hermansky-Pudlak ial neuroendocrine cell hyperplasia of infancy. Chest.
syndrome in a Puerto Rican population maps to chro- 2013;144(4):1199–1206.
mosome 10q2. Am J Hum Genet. 1995;57(4):755–765. 86. Hamvas A, Deterding RR, Wert SE, et al. Heterogene-
72. Brantly M, Avila NA, Shotelersuk V, Lucero C, Huizing ous pulmonary phenotypes associated with mutations
M, Gahl WA. Pulmonary function and high-resolution in the thyroid transcription factor gene NKX2-1. Chest.
CT findings in patients with an inherited form of pul- 2013;144(3):794–804.
monary fibrosis, Hermansky-Pudlak syndrome, due to 87. Carre A, Szinnai G, Castanet M, et al. Five new TTF1/
mutations in HPS-1. Chest. 2000;117(1):129–136. NKX2.1 mutations in brain-lung-thyroid syndrome:
73. Avila NA, Brantly M, Premkumar A, Huizing M, rescue by PAX8 synergism in one case. Hum Mol Genet.
Dwyer A, Gahl WA. Hermansky-Pudlak syndrome: radi- 2009;18(12):2266–2276.
ography and CT of the chest compared with pulmonary 88. Peca D, Petrini S, Tzialla C, et al. Altered surfactant ho-
function tests and genetic studies. AJR Am J Roentgenol. meostasis and recurrent respiratory failure secondary to
2002;179(4):887–892. TTF-1 nuclear targeting defect. Respir Res. 2011;12:115.
74. Anderson PD, Huizing M, Claassen DA, White J, Gahl 89. Burkhalter JL, Morano JU, McCay MB. Diffuse inter-
WA. Hermansky-Pudlak syndrome type 4 (HPS-4): stitial lung disease in neurofibromatosis. South Med J.
clinical and molecular characteristics. Hum Genet. 1986;79(8):944–946.
2003;113(1):10–17. 90. Massaro D, Katz S. Fibrosing alveolitis: its occurrence,
75. Bachli EB, Brack T, Eppler E, et al. Hermansky-Pudlak roentgenographic, and pathologic features in von Reck-
syndrome type 4 in a patient from Sri Lanka with pul- linghausen’s neurofibromatosis. Am Rev Respir Dis.
monary fibrosis. Am J Med Genet A. 2004;127A(2):201– 1966;93(6):934–942.
207. 91. Webb WR, Goodman PC. Fibrosing alveolitis in patients
76. Gahl WA, Brantly M, Troendle J, et al. Effect of pirfeni- with neurofibromatosis. Radiology. 1977;122(2):289–293.
done on the pulmonary fibrosis of Hermansky-Pudlak 92. Porterfield JK, Pyeritz RE, Traill TA. Pulmonary hyperten-
syndrome. Mol Genet Metab. 2002;76(3):234–242. sion and interstitial fibrosis in von Recklinghausen neu-
77. Gochuico BR, Huizing M, Golas GA, et al. Interstitial rofibromatosis. Am J Med Genet. 1986;25(3):531–535.
lung disease and pulmonary fibrosis in Hermansky- 93. Khumalo NP, Pillay K, Beighton P, et al. Poikilo-
Pudlak syndrome type 2, an adaptor protein-3 complex derma, tendon contracture and pulmonary fibrosis: a
disease. Mol Med. 2012;18:56–64. new autosomal dominant syndrome? Br J Dermatol.
78. Nakatani Y, Nakamura N, Sano J, et al. Interstitial pneu- 2006;155(5):1057–1061.
monia in Hermansky-Pudlak syndrome: significance of 94. Mercier S, Kury S, Shaboodien G, et al. Mutations in
florid foamy swelling/degeneration (giant lamellar body FAM111B cause hereditary fibrosing poikiloderma with
degeneration) of type-2 pneumocytes. Virchows Arch. tendon contracture, myopathy, and pulmonary fibrosis.
2000;437(3):304–313. Am J Hum Genet. 2013;93(6):1100–1107.
79. Pierson DM, Ionescu D, Qing G, et al. Pulmonary fibro- 95. Has C, Sparta G, Kiritsi D, et al. Integrin alpha3 muta-
sis in hermansky-pudlak syndrome. a case report and tions with kidney, lung, and skin disease. N Engl J Med.
review. Respiration. 2006;73(3):382–395. 2012;366(16):1508–1514.
80. O’Brien K, Troendle J, Gochuico BR, et al. Pirfenidone 96. Yalcin EG, He Y, Orhan D, Pazzagli C, Emiralioglu N,
for the treatment of Hermansky-Pudlak syndrome pul- Has C. Crucial role of posttranslational modifications of
monary fibrosis. Mol Genet Metab. 2011;103(2):128–134. integrin alpha3 in interstitial lung disease and nephrotic
81. Lyerla TA, Rusiniak ME, Borchers M, et al. Aberrant lung syndrome. Hum Mol Genet. 2015;24(13):3679–3688.
structure, composition, and function in a murine model 97. Liu Y, Jesus AA, Marrero B, et al. Activated STING in
of Hermansky-Pudlak syndrome. Am J Physiol Lung Cell a vascular and pulmonary syndrome. N Engl J Med.
Mol Physiol. 2003;285(3):L643–L653. 2014;371(6):507–518.
82. Mahavadi P, Korfei M, Henneke I, et al. Epithelial stress 98. Jeremiah N, Neven B, Gentili M, et al. Inherited STING-
and apoptosis underlie Hermansky-Pudlak syndrome- activating mutation underlies a familial inflammatory
associated interstitial pneumonia. Am J Respir Crit Care syndrome with lupus-like manifestations. J Clin Invest.
Med. 2010;182(2):207–219. 2014;124(12):5516–5520.
CHAPTER 1 Genetic Interstitial Lung Disease 19

99. Lohr NJ, Molleston JP, Strauss KA, et al. Human 114. Salzer U, Chapel HM, Webster AD, et al. Mutations in
ITCH E3 ubiquitin ligase deficiency causes syndromic TNFRSF13B encoding TACI are associated with com-
multisystem autoimmune disease. Am J Hum Genet. mon variable immunodeficiency in humans. Nat Genet.
2010;86(3):447–453. 2005;37(8):820–828.
100. Hustad CM, Perry WL, Siracusa LD, et al. Molecular ge- 115. Castigli E, Wilson SA, Garibyan L, et al. TACI is mutant
netic characterization of six recessive viable alleles of the in common variable immunodeficiency and IgA defi-
mouse agouti locus. Genetics. 1995;140(1):255–265. ciency. Nat Genet. 2005;37(8):829–834.
101. Perry WL, Hustad CM, Swing DA, O’Sullivan TN, Jenkins 116. Warnatz K, Salzer U, Rizzi M, et al. B-cell activating fac-
NA, Copeland NG. The itchy locus encodes a novel ubiq- tor receptor deficiency is associated with an adult-onset
uitin protein ligase that is disrupted in a18H mice. Nat antibody deficiency syndrome in humans. Proc Natl Acad
Genet. 1998;18(2):143–146. Sci USA. 2009;106(33):13945–13950.
102. Watkin LB, Jessen B, Wiszniewski W, et al. COPA mu- 117. van Zelm MC, Reisli I, van der Burg M, et al. An antibody-
tations impair ER-Golgi transport and cause hereditary deficiency syndrome due to mutations in the CD19 gene.
autoimmune-mediated lung disease and arthritis. Nat N Engl J Med. 2006;354(18):1901–1912.
Genet. 2015;47(6):654–660. 118. Kuijpers TW, Bende RJ, Baars PA, et al. CD20 deficiency
103. Ostergaard P, Simpson MA, Connell FC, et al. Mutations in humans results in impaired T cell-independent anti-
in GATA2 cause primary lymphedema associated with body responses. J Clin Invest. 2010;120(1):214–222.
a predisposition to acute myeloid leukemia (Emberger 119. van Zelm MC, Smet J, Adams B, et al. CD81 gene defect
syndrome). Nat Genet. 2011;43(10):929–931. in humans disrupts CD19 complex formation and leads
104. Hahn CN, Chong CE, Carmichael CL, et al. Heritable to antibody deficiency. J Clin Invest. 2010;120(4):1265–
GATA2 mutations associated with familial myelodysplastic 1274.
syndrome and acute myeloid leukemia. Nat Genet. 120. Thiel J, Kimmig L, Salzer U, et al. Genetic CD21 deficien-
2011;43(10):1012–1017. cy is associated with hypogammaglobulinemia. J Allergy
105. Dickinson RE, Griffin H, Bigley V, et al. Exome sequenc- Clin Immunol. 2012;129(3):801–810.e806.
ing identifies GATA-2 mutation as the cause of dendritic 121. Lopez-Herrera G, Tampella G, Pan-Hammarstrom Q,
cell, monocyte, B and NK lymphoid deficiency. Blood. et al. Deleterious mutations in LRBA are associated with
2011;118(10):2656–2658. a syndrome of immune deficiency and autoimmunity.
106. Hsu AP, Sampaio EP, Khan J, et al. Mutations in GATA2 Am J Hum Genet. 2012;90(6):986–1001.
are associated with the autosomal dominant and sporad- 122. Alangari A, Alsultan A, Adly N, et al. LPS-responsive
ic monocytopenia and mycobacterial infection (Mono- beige-like anchor (LRBA) gene mutation in a family with
MAC) syndrome. Blood. 2011;118(10):2653–2655. inflammatory bowel disease and combined immunode-
107. Spinner MA, Sanchez LA, Hsu AP, et al. GATA2 deficien- ficiency. J Allergy Clin Immunol. 2012;130(2):481–488.
cy: a protean disorder of hematopoiesis, lymphatics, and e482.
immunity. Blood. 2014;123(6):809–821. 123. Salzer E, Kansu A, Sic H, et al. Early-onset inflammatory
108. Hsu AP, Johnson KD, Falcone EL, et al. GATA2 haplo- bowel disease and common variable immunodeficiency-
insufficiency caused by mutations in a conserved in- like disease caused by IL-21 deficiency. J Allergy Clin Im-
tronic element leads to MonoMAC syndrome. Blood. munol. 2014;133(6):1651–1659.e1612.
2013;121(19):3830–3837. S3831–S3837. 124. Fliegauf M, Bryant VL, Frede N, et al. Haploinsufficiency
109. Aghamohammadi A, Allahverdi A, Abolhassani H, et al. of the NF-kappaB1 Subunit p50 in common variable im-
Comparison of pulmonary diseases in common variable munodeficiency. Am J Hum Genet. 2015;97(3):389–403.
immunodeficiency and X-linked agammaglobulinae- 125. Chen K, Coonrod EM, Kumanovics A, et al. Germline
mia. Respirology. 2010;15(2):289–295. mutations in NFKB2 implicate the noncanonical NF-
110. Verma N, Grimbacher B, Hurst JR. Lung disease in primary kappaB pathway in the pathogenesis of common variable
antibody deficiency. Lancet Respir Med. 2015;3(8):651–660. immunodeficiency. Am J Hum Genet. 2013;93(5):812–
111. Maarschalk-Ellerbroek LJ, de Jong PA, van Montfrans JM, 824.
et al. CT screening for pulmonary pathology in common 126. Goldman FD, Gurel Z, Al-Zubeidi D, et al. Congenital
variable immunodeficiency disorders and the correla- pancytopenia and absence of B lymphocytes in a neo-
tion with clinical and immunological parameters. J Clin nate with a mutation in the Ikaros gene. Pediatr Blood
Immunol. 2014;34(6):642–654. Cancer. 2012;58(4):591–597.
112. Gregersen S, Aalokken TM, Mynarek G, et al. High reso- 127. Kuehn HS, Boisson B, Cunningham-Rundles C,
lution computed tomography and pulmonary function et al. Loss of B cells in patients with heterozygous
in common variable immunodeficiency. Respir Med. mutations in IKAROS. N Engl J Med. 2016;374(11):
2009;103(6):873–880. 1032–1043.
113. Grimbacher B, Hutloff A, Schlesier M, et al. Homozygous 128. Chen Y, Stirling RG, Paul E, Hore-Lacy F, Thompson BR,
loss of ICOS is associated with adult-onset common Douglass JA. Longitudinal decline in lung function in
variable immunodeficiency. Nat Immunol. 2003;4(3): patients with primary immunoglobulin deficiencies. J
261–268. Allergy Clin Immunol. 2011;127(6):1414–1417.
20 Interstitial Lung Disease

129. Vetrie D, Vorechovsky I, Sideras P, et al. The gene in- 144. Partington MW, Prentice RS. X-linked cutaneous amyloi-
volved in X-linked agammaglobulinaemia is a member dosis: further clinical and pathological observations. Am
of the src family of protein-tyrosine kinases. Nature. J Med Genet. 1989;32(1):115–119.
1993;361(6409):226–233. 145. Ades LC, Rogers M, Sillence DO. An X-linked reticulate
130. Yel L, Minegishi Y, Coustan-Smith E, et al. Mutations in pigmentary disorder with systemic manifestations: report
the mu heavy-chain gene in patients with agammaglob- of a second family. Pediatr Dermatol. 1993;10(4):344–351.
ulinemia. N Engl J Med. 1996;335(20):1486–1493. 146. Anderson RC, Zinn AR, Kim J, Carder KR. X-linked retic-
131. Minegishi Y, Coustan-Smith E, Wang YH, Cooper MD, ulate pigmentary disorder with systemic manifestations:
Campana D, Conley ME. Mutations in the human lamb- report of a third family and literature review. Pediatr Der-
da5/14.1 gene result in B cell deficiency and agamma- matol. 2005;22(2):122–126.
globulinemia. J Exp Med. 1998;187(1):71–77. 147. Starokadomskyy P, Gemelli T, Rios JJ, et al. DNA
132. Minegishi Y, Coustan-Smith E, Rapalus L, Ersoy F, polymerase-alpha regulates the activation of type I
Campana D, Conley ME. Mutations in Igalpha (CD79a) interferons through cytosolic RNA: DNA synthesis. Nat
result in a complete block in B-cell development. J Clin Immunol. 2016;17(5):495–504.
Invest. 1999;104(8):1115–1121. 148. Kerem E, Elstein D, Abrahamov A, et al. Pulmonary func-
133. Minegishi Y, Rohrer J, Coustan-Smith E, et al. An essen- tion abnormalities in type I Gaucher disease. Eur Respir J.
tial role for BLNK in human B cell development. Science. 1996;9(2):340–345.
1999;286(5446):1954–1957. 149. Miller A, Brown LK, Pastores GM, Desnick RJ. Pulmonary
134. Dobbs AK, Yang T, Farmer D, Kager L, Parolini O, involvement in type 1 Gaucher disease: functional and
Conley ME. Cutting edge: a hypomorphic mutation exercise findings in patients with and without clinical in-
in Igbeta (CD79b) in a patient with immunodeficiency terstitial lung disease. Clin Genet. 2003;63(5):368–376.
and a leaky defect in B cell development. J Immunol. 150. Goitein O, Elstein D, Abrahamov A, et al. Lung involve-
2007;179(4):2055–2059. ment and enzyme replacement therapy in Gaucher’s dis-
135. Boisson B, Wang YD, Bosompem A, et al. A recur- ease. QJM. 2001;94(8):407–415.
rent dominant negative E47 mutation causes agam- 151. Jmoudiak M, Futerman AH. Gaucher disease: pathologi-
maglobulinemia and BCR(-) B cells. J Clin Invest. cal mechanisms and modern management. Br J Haema-
2013;123(11):4781–4785. tol. 2005;129(2):178–188.
136. Freeman AF, Holland SM. Clinical manifestations, etiol- 152. McGovern MM, Wasserstein MP, Giugliani R, et al. A
ogy, and pathogenesis of the hyper-IgE syndromes. Pedi- prospective, cross-sectional survey study of the natu-
atr Res. 2009;65(5 Pt 2):32R–37R. ral history of Niemann-Pick disease type B. Pediatrics.
137. Freeman AF, Kleiner DE, Nadiminti H, et al. Causes of 2008;122(2):e341–e349.
death in hyper-IgE syndrome. J Allergy Clin Immunol. 153. Mendelson DS, Wasserstein MP, Desnick RJ, et al. Type
2007;119(5):1234–1240. B Niemann-Pick disease: findings at chest radiography,
138. Minegishi Y, Saito M, Tsuchiya S, et al. Dominant- thin-section CT, and pulmonary function testing. Radiol-
negative mutations in the DNA-binding domain of STAT3 ogy. 2006;238(1):339–345.
cause hyper-IgE syndrome. Nature. 2007;448(7157): 154. Parto K, Svedstrom E, Majurin ML, Harkonen R, Simell O.
1058–1062. Pulmonary manifestations in lysinuric protein intolerance.
139. Milner JD, Brenchley JM, Laurence A, et al. Impaired Chest. 1993;104(4):1176–1182.
T(H)17 cell differentiation in subjects with autosomal 155. Nogee LM. Genetic basis of children’s interstitial lung
dominant hyper-IgE syndrome. Nature. 2008;452(7188): disease. Pediatr Allergy Immunol Pulmonol. 2010;23(1).
773–776. 156. Nogee LM, de Mello DE, Dehner LP, Colten HR. Brief
140. Angulo I, Vadas O, Garcon F, et al. Phosphoinositide report: deficiency of pulmonary surfactant protein
3-kinase delta gene mutation predisposes to respiratory B in congenital alveolar proteinosis. N Engl J Med.
infection and airway damage. Science. 2013;342(6160): 1993;328(6):406–410.
866–871. 157. Nogee LM, Garnier G, Dietz HC, et al. A mutation in the
141. Lucas CL, Kuehn HS, Zhao F, et al. Dominant-activating surfactant protein B gene responsible for fatal neonatal
germline mutations in the gene encoding the PI(3)K respiratory disease in multiple kindreds. J Clin Invest.
catalytic subunit p110delta result in T cell senescence and 1994;93(4):1860–1863.
human immunodeficiency. Nat Immunol. 2014;15(1): 158. Andersen C, Ramsay JA, Nogee LM, et al. Recurrent fa-
88–97. milial neonatal deaths: hereditary surfactant protein B
142. Kuehn HS, Ouyang W, Lo B, et al. Immune dysregu- deficiency. Am J Perinatol. 2000;17(4):219–224.
lation in human subjects with heterozygous ger- 159. Cole FS, Hamvas A, Rubinstein P, et al. Population-
mline mutations in CTLA4. Science. 2014;345(6204): based estimates of surfactant protein B deficiency. Pedi-
1623–1627. atrics. 2000;105(3 Pt 1):538–541.
143. Schubert D, Bode C, Kenefeck R, et al. Autosomal domi- 160. Somaschini M, Wert S, Mangili G, Colombo A, Nogee L.
nant immune dysregulation syndrome in humans with Hereditary surfactant protein B deficiency resulting from
CTLA4 mutations. Nature Med. 2014;20(12):1410–1416. a novel mutation. Intensive Care Med. 2000;26(1):97–100.
CHAPTER 1 Genetic Interstitial Lung Disease 21

161. Wegner DJ, Hertzberg T, Heins HB, et al. A major 176. Stevens PA, Pettenazzo A, Brasch F, et al. Nonspecific
deletion in the surfactant protein-B gene causing le- interstitial pneumonia, alveolar proteinosis, and abnor-
thal respiratory distress. Acta Paediatr. 2007;96(4): mal proprotein trafficking resulting from a spontaneous
516–520. mutation in the surfactant protein C gene. Pediatr Res.
162. Dunbar 3rd AE, Wert SE, Ikegami M, et al. Prolonged 2005;57(1):89–98.
survival in hereditary surfactant protein B (SP-B) defi- 177. Woischnik M, Sparr C, Kern S, et al. A non-BRICHOS
ciency associated with a novel splicing mutation. Pediatr surfactant protein c mutation disrupts epithelial cell
Res. 2000;48(3):275–282. function and intercellular signaling. BMC Cell Biol.
163. Ballard PL, Nogee LM, Beers MF, et al. Partial deficiency 2010;11:88.
of surfactant protein B in an infant with chronic lung 178. Bridges JP, Xu Y, Na CL, Wong HR, Weaver TE. Adapta-
disease. Pediatrics. 1995;96(6):1046–1052. tion and increased susceptibility to infection associated
164. Nogee LM, Dunbar 3rd AE, Wert SE, Askin F, Hamvas A, with constitutive expression of misfolded SP-C. J Cell
Whitsett JA. A mutation in the surfactant protein C gene Biol. 2006;172(3):395–407.
associated with familial interstitial lung disease. N Engl J 179. Garmany TH, Wambach JA, Heins HB, et al. Population
Med. 2001;344(8):573–579. and disease-based prevalence of the common muta-
165. Thomas AQ, Lane K, Phillips 3rd J, et al. Heterozy- tions associated with surfactant deficiency. Pediatr Res.
gosity for a surfactant protein C gene mutation as- 2008;63(6):645–649.
sociated with usual interstitial pneumonitis and 180. Somaschini M, Nogee LM, Sassi I, et al. Unexplained
cellular nonspecific interstitial pneumonitis in one neonatal respiratory distress due to congenital sur-
kindred. Am J Respir Crit Care Med. 2002;165(9): factant deficiency. J Pediatr. 2007;150(6):649–653.
1322–1328. 653.e641.
166. Abou Taam R, Jaubert F, Emond S, et al. Familial inter- 181. Weaver TE. Synthesis, processing and secretion of
stitial disease with I73T mutation: a mid- and long-term surfactant proteins B and C. Biochim Biophys Acta.
study. Pediatr Pulmonol. 2009;44(2):167–175. 1998;1408(2–3):173–179.
167. Brasch F, Griese M, Tredano M, et al. Interstitial lung dis- 182. Serrano AG, Perez-Gil J. Protein-lipid interactions and
ease in a baby with a de novo mutation in the SFTPC surface activity in the pulmonary surfactant system.
gene. Eur Respir J. 2004;24(1):30–39. Chem Phys Lipids. 2006;141(1–2):105–118.
168. Bullard JE, Nogee LM. Heterozygosity for ABCA3 mu- 183. Matsumura Y, Ban N, Inagaki N. Aberrant catalytic cycle
tations modifies the severity of lung disease associated and impaired lipid transport into intracellular vesicles
with a surfactant protein C gene (SFTPC) mutation. Pedi- in ABCA3 mutants associated with nonfatal pediatric in-
atr Res. 2007;62(2):176–179. terstitial lung disease. Am J Physiol Lung Cell Mol Physiol.
169. Cameron HS, Somaschini M, Carrera P, et al. A common 2008;295(4):L698–L707.
mutation in the surfactant protein C gene associated 184. Matsumura Y, Ban N, Ueda K, Inagaki N. Characteriza-
with lung disease. J Pediatr. 2005;146(3):370–375. tion and classification of ATP-binding cassette transport-
170. Soraisham AS, Tierney AJ, Amin HJ. Neonatal respira- er ABCA3 mutants in fatal surfactant deficiency. J Biol
tory failure associated with mutation in the surfactant Chem. 2006;281(45):34503–34514.
protein C gene. J Perinatol. 2006;26(1):67–70. 185. Cheong N, Madesh M, Gonzales LW, et al. Functional
171. van Moorsel CH, van Oosterhout MF, Barlo NP, and trafficking defects in ATP binding cassette A3 mu-
et al. Surfactant protein C mutations are the basis tants associated with respiratory distress syndrome. J Biol
of a significant portion of adult familial pulmonary Chem. 2006;281(14):9791–9800.
fibrosis in a dutch cohort. Am J Respir Crit Care Med. 186. Trapnell BC, Whitsett JA, Nakata K. Pulmonary alveolar
2010;182(11):1419–1425. proteinosis. N Engl J Med. 2003;349(26):2527–2539.
172. Lawson WE, Grant SW, Ambrosini V, et al. Genetic muta- 187. Prakash UB, Barham SS, Carpenter HA, Dines DE,
tions in surfactant protein C are a rare cause of sporadic Marsh HM. Pulmonary alveolar phospholipoproteino-
cases of IPF. Thorax. 2004;59(11):977–980. sis: experience with 34 cases and a review. Mayo Clin
173. Markart P, Ruppert C, Wygrecka M, et al. Surfactant Proc. 1987;62(6):499–518.
protein C mutations in sporadic forms of idiopathic 188. Goldstein LS, Kavuru MS, Curtis-McCarthy P, Christie HA,
interstitial pneumonias. Eur Respir J. 2007;29(1): Farver C, Stoller JK. Pulmonary alveolar proteinosis: clini-
134–137. cal features and outcomes. Chest. 1998;114(5):1357–1362.
174. Wert SE, Whitsett JA, Nogee LM. Genetic disorders of 189. Seymour JF, Presneill JJ. Pulmonary alveolar proteinosis:
surfactant dysfunction. Pediatr Dev Pathol. 2009;12(4): progress in the first 44 years. Am J Respir Crit Care Med.
253–274. 2002;166(2):215–235.
175. Mulugeta S, Maguire JA, Newitt JL, Russo SJ, Kotorash- 190. Kitamura T, Tanaka N, Watanabe J, et al. Idiopathic
vili A, Beers MF. Misfolded BRICHOS SP-C mutant pro- pulmonary alveolar proteinosis as an autoimmune
teins induce apoptosis via caspase-4- and cytochrome disease with neutralizing antibody against granulo-
c-related mechanisms. Am J Physiol Lung Cell Mol Physiol. cyte/macrophage colony-stimulating factor. J Exp Med.
2007;293(3):L720–L729. 1999;190(6):875–880.
22 Interstitial Lung Disease

191. Bonfield TL, Russell D, Burgess S, Malur A, Kavuru MS, 205. Hadchouel A, Wieland T, Griese M, et al. Biallelic muta-
Thomassen MJ. Autoantibodies against granulocyte tions of methionyl-tRNA synthetase cause a specific type
macrophage colony-stimulating factor are diagnostic for of pulmonary alveolar proteinosis prevalent on reunion
pulmonary alveolar proteinosis. Am J Respir Cell Mol Biol. island. Am J Hum Genet. 2015;96(5):826–831.
2002;27(4):481–486. 206. van Meel E, Wegner DJ, Cliften P, et al. Rare recessive
192. Lin FC, Chang GD, Chern MS, Chen YC, Chang SC. loss-of-function methionyl-tRNA synthetase mutations
Clinical significance of anti-GM-CSF antibodies in presenting as a multi-organ phenotype. BMC Med Genet.
idiopathic pulmonary alveolar proteinosis. Thorax. 2013;14:106.
2006;61(6):528–534. 207. Armanios MY, Chen JJ, Cogan JD, et al. Telomerase mu-
193. Inoue Y, Trapnell BC, Tazawa R, et al. Characteristics of tations in families with idiopathic pulmonary fibrosis. N
a large cohort of patients with autoimmune pulmonary Engl J Med. 2007;356(13):1317–1326.
alveolar proteinosis in Japan. Am J Respir Crit Care Med. 208. Tsakiri KD, Cronkhite JT, Kuan PJ, et al. Adult-onset pul-
2008;177(7):752–762. monary fibrosis caused by mutations in telomerase. Proc
194. Carey B, Trapnell BC. The molecular basis of pulmo- Natl Acad Sci USA. 2007;104(18):7552–7557.
nary alveolar proteinosis. Clin Immunol. 2010;135(2): 209. Cronkhite JT, Xing C, Raghu G, et al. Telomere shorten-
223–235. ing in familial and sporadic pulmonary fibrosis. Am J
195. Uchida K, Beck DC, Yamamoto T, et al. GM-CSF autoan- Respir Crit Care Med. 2008;178(7):729–737.
tibodies and neutrophil dysfunction in pulmonary al- 210. Diaz de Leon A, Cronkhite JT, Katzenstein AL, et al.
veolar proteinosis. N Engl J Med. 2007;356(6):567–579. Telomere lengths, pulmonary fibrosis and telomerase
196. Uchida K, Nakata K, Trapnell BC, et al. High-affinity (TERT) mutations. PLoS One. 2010;5(5):e10680.
autoantibodies specifically eliminate granulocyte- 211. Armanios M. Telomerase and idiopathic pulmonary fi-
macrophage colony-stimulating factor activity in the brosis. Mutat Res. 2012;730(1–2):52–58.
lungs of patients with idiopathic pulmonary alveolar 212. Yamaguchi H, Calado RT, Ly H, et al. Mutations in TERT,
proteinosis. Blood. 2004;103(3):1089–1098. the gene for telomerase reverse transcriptase, in aplastic
197. Hansen G, Hercus TR, McClure BJ, et al. The structure of anemia. N Engl J Med. 2005;352(14):1413–1424.
the GM-CSF receptor complex reveals a distinct mode 213. Calado RT, Brudno J, Mehta P, et al. Constitutional tel-
of cytokine receptor activation. Cell. 2008;134(3): omerase mutations are genetic risk factors for cirrhosis.
496–507. Hepatology. 2011;53(5):1600–1607.
198. Suzuki T, Sakagami T, Rubin BK, et al. Familial pulmo- 214. Hartmann D, Srivastava U, Thaler M, et al. Telomerase
nary alveolar proteinosis caused by mutations in CS- gene mutations are associated with cirrhosis formation.
F2RA. J Exp Med. 2008;205(12):2703–2710. Hepatology. 2011;53(5):1608–1617.
199. Suzuki T, Sakagami T, Young LR, et al. Hereditary pul- 215. Diaz de Leon A, Cronkhite JT, Yilmaz C, et al. Subclini-
monary alveolar proteinosis: pathogenesis, presenta- cal lung disease, macrocytosis, and premature graying
tion, diagnosis, and therapy. Am J Respir Crit Care Med. in kindreds with telomerase (TERT) mutations. Chest.
2010;182(10):1292–1304. 2011;140(3):753–763.
200. Tanaka T, Motoi N, Tsuchihashi Y, et al. Adult-onset 216. Calado RT, Regal JA, Hills M, et al. Constitutional hy-
hereditary pulmonary alveolar proteinosis caused pomorphic telomerase mutations in patients with
by a single-base deletion in CSF2RB. J Med Genet. acute myeloid leukemia. Proc Natl Acad Sci USA.
2011;48(3):205–209. 2009;106(4):1187–1192.
201. Dranoff G, Crawford AD, Sadelain M, et al. Involvement of 217. Kirwan M, Vulliamy T, Marrone A, et al. Defining the
granulocyte-macrophage colony-stimulating factor in pul- pathogenic role of telomerase mutations in myelodys-
monary homeostasis. Science. 1994;264(5159):713–716. plastic syndrome and acute myeloid leukemia. Hum Mu-
202. Stanley E, Lieschke GJ, Grail D, et al. Granulocyte/mac- tat. 2009;30(11):1567–1573.
rophage colony-stimulating factor-deficient mice show 218. Newton C, Batra K, Torrealba J, et al. Telomere-related
no major perturbation of hematopoiesis but develop a lung fibrosis is diagnostically heterogeneous but uni-
characteristic pulmonary pathology. Proc Natl Acad Sci formly progressive. Eur Respir J. 2016;48(6):1710–1720.
USA. 1994;91(12):5592–5596. 219. Silhan LL, Shah PD, Chambers DC, et al. Lung
203. Nishinakamura R, Nakayama N, Hirabayashi Y, et al. transplantation in telomerase mutation carriers
Mice deficient for the IL-3/GM-CSF/IL-5 beta c receptor with pulmonary fibrosis. Eur Respir J. 2014;44(1):
exhibit lung pathology and impaired immune response, 178–187.
while beta IL3 receptor-deficient mice are normal. Im- 220. Borie R, Kannengiesser C, Hirschi S, et al. Severe hema-
munity. 1995;2(3):211–222. tologic complications after lung transplantation in pa-
204. Robb L, Drinkwater CC, Metcalf D, et al. Hematopoi- tients with telomerase complex mutations. J Heart Lung
etic and lung abnormalities in mice with a null mu- Transplant. 2014.
tation of the common beta subunit of the receptors 221. Tokman S, Singer JP, Devine MS, et al. Clinical out-
for granulocyte-macrophage colony-stimulating fac- comes of lung transplantation in patients with telom-
tor and interleukins 3 and 5. Proc Natl Acad Sci USA. erase mutations. J Heart Lung Transplant. 2015;34(10):
1995;92(21):9565–9569. 1318–1324.
CHAPTER 1 Genetic Interstitial Lung Disease 23

222. Cogan JD, Kropski JA, Zhao M, et al. Rare variants 237. Mushiroda T, Wattanapokayakit S, Takahashi A, et al. A ge-
in RTEL1 are associated with familial interstitial nome-wide association study identifies an association of a
pneumonia. Am J Respir Crit Care Med. 2015;191(6): common variant in TERT with susceptibility to idiopathic
646–655. pulmonary fibrosis. J Med Genet. 2008;45(10):654–656.
223. Stuart BD, Choi J, Zaidi S, et al. Exome sequencing 238. Calado RT, Young NS. Telomere diseases. N Engl J Med.
links mutations in PARN and RTEL1 with familial pul- 2009;361(24):2353–2365.
monary fibrosis and telomere shortening. Nat Genet. 239. Levy D, Neuhausen SL, Hunt SC, et al. Genome-wide
2015;47(5):512–517. association identifies OBFC1 as a locus involved in hu-
224. Kannengiesser C, Borie R, Menard C, et al. Heterozygous man leukocyte telomere biology. Proc Natl Acad Sci USA.
RTEL1 mutations are associated with familial pulmo- 2010;107(20):9293–9298.
nary fibrosis. Eur Respir J. 2015. 240. Codd V, Nelson CP, Albrecht E, et al. Identification of
225. Kropski JA, Mitchell DB, Markin C, et al. A novel dysk- seven loci affecting mean telomere length and their as-
erin (DKC1) mutation is associated with familial inter- sociation with disease. Nat Genet. 2013;45(4):422–427.
stitial pneumonia. Chest. 2014;146(1):e1–e7. 427e421–e422.
226. Alder JK, Stanley SE, Wagner CL, Hamilton M, Hanu- 241. Castellana G, Lamorgese V. Pulmonary alveolar micro-
manthu VS, Armanios M. Exome sequencing identi- lithiasis. World cases and review of the literature. Respira-
fies mutant TINF2 in a family with pulmonary fibrosis. tion. 2003;70(5):549–555.
Chest. 2015;147(5):1361–1368. 242. Sosman MC, Dodd GD, Jones WD, Pillmore GU. The fa-
226a. Stanley SE, Gable DL, Wagner CL, et al. Loss-of-function milial occurrence of pulmonary alveolar microlithiasis. Am
mutations in the RNA biogenesis factor NAF1 predis- J Roentgenol Radium Ther Nucl Med. 1957;77(6):947–1012.
pose to pulmonary fibrosis–emphysema. Sci Transl Med. 243. Ucan ES, Keyf AI, Aydilek R, et al. Pulmonary alveo-
2016;8(351):351ra107. lar microlithiasis: review of Turkish reports. Thorax.
227. Wang Y, Kuan PJ, Xing C, et al. Genetic defects in 1993;48(2):171–173.
surfactant protein A2 are associated with pulmo- 244. Melamed JW, Sostman HD, Ravin CE. Interstitial thick-
nary fibrosis and lung cancer. Am J Hum Genet. ening in pulmonary alveolar microlithiasis: an underap-
2009;84(1):52–59. preciated finding. J Thorac Imaging. 1994;9(2):126–128.
228. van Moorsel CH, Ten Klooster L, van Oosterhout MF, 245. Helbich TH, Wojnarovsky C, Wunderbaldinger P,
et al. SFTPA2 mutations in familial and sporadic idio- Heinz-Peer G, Eichler I, Herold CJ. Pulmonary al-
pathic interstitial pneumonia. Am J Respir Crit Care Med. veolar microlithiasis in children: radiographic and
2015;192(10):1249–1252. high-resolution CT findings. AJR Am J Roentgenol.
229. Nathan N, Giraud V, Picard C, et al. Germline SFTPA1 1997;168(1):63–65.
mutation in familial idiopathic interstitial pneumonia 246. Corut A, Senyigit A, Ugur SA, et al. Mutations in
and lung cancer. Hum Mol Genet. 2016. SLC34A2 cause pulmonary alveolar microlithiasis and
230. Maitra M, Wang Y, Gerard RD, Mendelson CR, Garcia CK. are possibly associated with testicular microlithiasis. Am
Surfactant protein A2 mutations associated with pulmo- J Hum Genet. 2006;79(4):650–656.
nary fibrosis lead to protein instability and endoplasmic 247. Huqun, Izumi S, Miyazawa H, et al. Mutations in
reticulum stress. J Biol Chem. 2010;285(29):22103–22113. the SLC34A2 gene are associated with pulmonary
231. Seibold MA, Wise AL, Speer MC, et al. A common alveolar microlithiasis. Am J Respir Crit Care Med.
MUC5B promoter polymorphism and pulmonary fibro- 2007;175(3):263–268.
sis. N Engl J Med. 2011;364(16):1503–1512. 248. Wang H, Yin X, Wu D. Novel human pathological muta-
232. Fingerlin TE, Murphy E, Zhang W, et al. Genome-wide tions. SLC34A2. Pulmonary alveolar microlithiasis. Hum
association study identifies multiple susceptibility loci Genet. 2010;127(4):471.
for pulmonary fibrosis. Nat Genet. 2013;45(6):613–620. 249. Dogan OT, Ozsahin SL, Gul E, et al. A frame-shift muta-
233. Hunninghake GM, Hatabu H, Okajima Y, et al. MUC5B tion in the SLC34A2 gene in three patients with pulmo-
promoter polymorphism and interstitial lung abnormal- nary alveolar microlithiasis in an inbred family. Intern
ities. N Engl J Med. 2013;368(23):2192–2200. Med. 2010;49(1):45–49.
234. Schroeder BW, Verhaeghe C, Park SW, et al. AGR2 is 250. Ishihara Y, Hagiwara K, Zen K, Huqun, Hosokawa Y,
induced in asthma and promotes allergen-induced Natsuhara A. A case of pulmonary alveolar microlithiasis
mucin overproduction. Am J Respir Cell Mol Biol. with an intragenetic deletion in SLC34A2 detected by a
2012;47(2):178–185. genome-wide SNP study. Thorax. 2009;64(4):365–367.
235. Zhao F, Edwards R, Dizon D, et al. Disruption of 251. Hashimoto M, Wang DY, Kamo T, et al. Isolation and
Paneth and goblet cell homeostasis and increased en- localization of type IIb Na/Pi cotransporter in the devel-
doplasmic reticulum stress in Agr2-/- mice. Dev Biol. oping rat lung. Am J Pathol. 2000;157(1):21–27.
2010;338(2):270–279. 252. Feild JA, Zhang L, Brun KA, Brooks DP, Edwards RM.
236. Tsuru A, Fujimoto N, Takahashi S, et al. Negative Cloning and functional characterization of a sodium-
feedback by IRE1beta optimizes mucin production dependent phosphate transporter expressed in human
in goblet cells. Proc Natl Acad Sci USA. 2013;110(8): lung and small intestine. Biochem Biophys Res Commun.
2864–2869. 1999;258(3):578–582.
24 Interstitial Lung Disease

253. Traebert M, Hattenhauer O, Murer H, Kaissling B, Biber J. 256. Tanjore H, Blackwell TS, Lawson WE. Emerging evidence
Expression of type II Na-P(i) cotransporter in alveolar for endoplasmic reticulum stress in the pathogenesis of
type II cells. Am J Physiol. 1999;277(5 Pt 1):L868–L873. idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol
254. Saito A, Nikolaidis NM, Amlal H, et al. Modeling pul- Physiol. 2012;302(8):L721–L729.
monary alveolar microlithiasis by epithelial deletion of 257. Stuart BD, Lee JS, Kozlitina J, et al. Effect of telomere
the Npt2b sodium phosphate cotransporter reveals pu- length on survival in patients with idiopathic pulmonary
tative biomarkers and strategies for treatment. Sci Transl fibrosis: an observational cohort study with independ-
Med. 2015;7(313):313ra181. ent validation. Lancet Respir Med. 2014;2(7):557–565.
255. Mulugeta S, Nureki S, Beers MF. Lost after transla- 258. Dai J, Cai H, Li H, et al. Association between telomere
tion: insights from pulmonary surfactant for under- length and survival in patients with idiopathic pulmo-
standing the role of alveolar epithelial dysfunction nary fibrosis. Respirology. 2015;20(6):947–952.
and cellular quality control in fibrotic lung disease.
Am J Physiol Lung Cell Mol Physiol. 2015;309(6):
L507–L525.
CHAPTER 2

Pathobiology of Novel Approaches


to Treatment
SILVIA PUGLISI • CARLO VANCHERI

KEY POINTS

• By virtue of basic research and new clinical observations, the amount of information and data on the pathobiology of
IPF is growing day by day, offering new and more ample opportunities to either develop new therapies or reposition
known drugs for IPF.
• A number of preclinical and clinical studies are currently under way with the aim of finding new and more potent
drugs or effective cell-based treatments for IPF and possibly to personalize treatments, identifying subcategories of
patients on the basis of genetic backgrounds and response to treatment.
• This will certainly be possible in a future not so distant, provided that the tree of knowledge for the pathobiology of
IPF continues to grow florid and robust.

An understanding of the pathogenesis of any disease is result of the interaction between the individual genetic
crucial to identifying those cellular and molecular mech- background, the chronic action of different exogenous
anisms through which a disease arises and evolves. This risk factors such as smoking, and the relentless action of
may lead to the discovery and development of new com- aging. In susceptible and aged individuals, chronic dam-
pounds and drugs that, acting on specific targets, may age caused by risk factors elicits an abnormal response
be effective in slowing or stopping the disease. Unfor- of alveolar–bronchiolar cells that subsequently leads
tunately, the pathogenesis of many complex diseases to the activation of fibroblasts and ultimately to tissue
caused by the combination of genetic, environmental, fibrosis. During the last 10 years some of these patho-
and lifestyle factors is still not clear. Idiopathic pulmo- biologic mechanisms, involved in the abnormal
nary fibrosis (IPF) is no exception to this rule, and its response to damage of epithelial cells and fibroblasts,
pathogenesis represents the perfect example of a complex have been disclosed, and today the overall picture of the
disease due to the combination of genetic predisposition pathogenesis of IPF seems less obscure. Based on these
and exogenous risk factors. Nevertheless, during the last considerations this chapter will review some aspects of
few years, we have gathered useful information about IPF pathobiology that have inspired research studies
some of the cellular and molecular pathways involved in that emerged into clinical trials leading to the develop-
the origin and progression of this dreadful disease. How- ment of new drugs. The pathobiologic foundations of
ever, it must be admitted that some of the data collected some ongoing trials will be also reviewed.
derive either from diagnostic biopsies and explanted
lung tissues or from in vitro and in vivo animal studies,
and for this reason they may have some limitations. THE APPROACH TO TREATMENT IN THE
Lung tissue from biopsy, and even more explanted OLD DAYS
tissue, mirrors disease in specific moments and very For many years lung fibrosis was considered the result
often when it is in an advanced stage, whereas in vitro of a chronic inflammatory process of the alveolar tissue
and in vivo studies, although very suggestive and that could evolve into fibrosis. Based on this belief and
extremely useful, need to be confirmed in the context a long empiric experience (not supported by scientific
of the human disease. In spite of these objective limita- evidence), the treatment of IPF was built for decades
tions, there is scientific agreement in considering IPF the on high doses of steroids associated with immunosup-

25
26 Interstitial Lung Disease

pressants, such as azathioprine. This approach was also PANTHER study, a clinical trial designed to assess the
endorsed by the ATS/ERS statement published in 2000, efficacy and safety of triple therapy (prednisone, aza-
although two Cochrane reviews1 underlined the lack thioprine, and N-acetylcysteine [NAC]) in IPF patients.
of randomized controlled trials exploring the effect of Quite surprisingly, this trial was stopped before its con-
steroids and immunosuppressants in IPF. Both reviews clusion because of an excessive number of deaths, hos-
concluded that there was very little evidence support- pitalizations, and adverse events in the treated group,5
ing the role of these drugs for the treatment of IPF. and the absence of any efficacy. The PANTHER study
This is not surprising considering that the presence put an end to the “traditional therapy” with steroids
of inflammatory and immune cells is not a dominant and immunosuppressants for the treatment of IPF.
feature of IPF lung tissue.2 In addition, even if it is In the meantime, additional experimental data had
difficult to conceive, fibrosis may take place without confirmed the marginal role of inflammation in IPF
being preceded by chronic inflammation. It has been showing instead the key role of AECs and fibroblasts.
demonstrated that it is possible to induce a fibrotic According to more recent studies, the inception of tis-
reaction in a lung culture system free of blood cells sue fibrosis is due to the exposure of AECs to chronic
and soluble mediators. This model of “washed lung” microinjuries, such as those caused by smoking or
suggested that the disruption of the normal epithe- environmental and/or professional exposures that may
lial–fibroblast interaction, induced by hyperoxia as the damage the epithelium, causing, in susceptible indi-
fibrogenic stimulus, may be sufficient, in the absence viduals, the activation of incorrect repair mechanisms
of the classical inflammatory cells, to induce epithelial and stress response pathways that eventually promote
damage and to promote fibroblast growth and collagen fibroblast recruitment and proliferation.6 During nor-
deposition, in other words, to create tissue fibrosis.3 mal tissue repair processes, when wound healing is
This is possible because epithelial cells and fibroblasts, over, the activity of fibroblasts ends, and they undergo
commonly considered structural cells, are not merely apoptosis. However, in some circumstances, and IPF is
bystander cells forming the scaffold of the lung, but are one of those, fibroblast functions are altered resulting
instead able to produce and release mediators, to pro- in their permanence into the tissue, ongoing deposi-
liferate and differentiate into more active and aggres- tion of extracellular matrix (ECM) proteins, parenchy-
sive phenotypes, and eventually to affect the behavior mal distortion, and ultimately fibrosis. Starting from
of the surrounding microenvironment. The turning this pathogenic evidence, new preclinical studies and
point in defining the real nature of the pathogenesis of clinical trials have been designed, shifting their focus
IPF was made by Selman in 2002; he described IPF as a from antiinflammatory molecules to targeting compo-
noninflammatory fibroproliferative disorder triggered nents of the wound healing cascade and fibrogenesis.
by an unknown stimulus responsible for alveolar epi-
thelial cell (AEC) injury. According to this hypothesis,
epithelial damage and the subsequent altered crosstalk THE ADVENT OF PIRFENIDONE
between fibroblasts and epithelium caused the release Pirfenidone (5-methyl-1-phenyl-2-[1H]-pyridone) is
of growth factors and cytokines involved in the recruit- an orally available synthetic pyridone compound that
ment and differentiation of mesenchymal cells into has been shown to inhibit the progression of fibrosis
myofibroblasts.4 in animal models and in in vitro systems. Pirfenidone
In spite of the clear evidence showing a limited role has been used for the treatment of different models
for inflammation in the pathogenesis of IPF, the treat- of fibrotic diseases, including liver, renal, and cardiac
ment of this disease remained based on steroids and fibrosis, all situations characterized by the abnormal
immunosuppressants. This was likely due to at least deposition of collagen. The mechanism of action is still
three reasons: (1) In the recent past the diagnosis of not fully understood, but preclinical studies suggest
IPF was not as accurate as today, and in some patients, that pirfenidone combines antiinflammatory, antioxi-
with other forms of lung fibrosis such as nonspecific dant, and above all antifibrotic properties. Pirfenidone
interstitial pneumonia (NSIP), the “traditional treat- exerts its antiinflammatory action mainly through the
ment” with steroids and immunosuppressants could regulation of tumor necrosis factor alpha and beta
be effective. (2) There is a complete lack of any other (TNF-α and TNF-β) pathways,7,8 whereas its antioxi-
alternative treatment with the exception of lung trans- dant activity is exerted through the reduction of lipid
plantation. (3) Controlled clinical trials demonstrating peroxidation and partly because of its scavenger activ-
whether or not this treatment was effective are lacking. ity for toxic hydroxyl radicals.9 However, pirfenidone is
This final gap was eventually filled in 2012 with the best known for its antifibrotic properties.
CHAPTER 2 Pathobiology of Novel Approaches to Treatment 27

Fibroblasts are normally quiescent cells unless tis- pirfenidone group.16 These encouraging results led to
sue injury occurs. In this situation they undergo a another study in Japan, whose primary endpoint was
phenotypic transition into myofibroblasts acquiring the change in vital capacity from baseline at 52 weeks.
contractile and secretory properties and synthesiz- This time the study met its primary endpoint showing
ing collagen and ECM proteins. This is an important that the loss in vital capacity was significantly higher in
step in the pathogenesis of IPF, and many studies the placebo group. It was also proved that pirfenidone
have proposed TGF-β as the key fibrogenic cytokine increased the progression-free survival (PFS) defined as
responsible for fibroblast differentiation.10 The major the time until death and 10% or more decline in vital
signaling pathway of TGF-β is through its transmem- capacity from baseline.17
brane receptor serine/threonine kinases that in turn In the meantime two multinational randomized
activate the cytoplasmic Smad proteins, and more double-blind placebo-controlled Phase III trials (004
specifically Smad3, inducing alpha smooth muscle and 006) named CAPACITY started in Europe, Austra-
actin (SMA) gene expression and subsequently fibro- lia, and North America to test the efficacy of pirfenidone
blast differentiation.11 It has been also demonstrated in patients affected by mild to moderate IPF. The pri-
that Smad-independent molecular pathways such as mary endpoint, common to both studies, was defined
phosphatidylinositol-3-kinase/protein kinase B (PI3K/ as the change in forced vital capacity (FVC) percentage
Akt) might be involved in myofibroblast differentiation from baseline to 72 weeks. In study 004, pirfenidone
induced by TGF-β.12 significantly reduced FVC decline at week 72 but did
A number of in vitro and in vivo animal studies not reduce decline in 6-min walk test (6MWT) distance,
have demonstrated that pirfenidone inhibits collagen while study 006 failed to meet its primary endpoint
synthesis and ECM deposition and reduces fibroblast but reduced the decline in 6MWT distance.18 Based on
proliferation and, most important, their differentia- the positive results of the one CAPACITY and Japanese
tion into myofibroblasts.13 Other studies have shown studies, pirfenidone was approved in 2011 by the Euro-
that pirfenidone also decreases TGF-β–induced α-SMA pean Medicines Agency (EMA) for the treatment of mild
and procollagen (Col) I at mRNA and protein levels. to moderate IPF. The Food and Drug Administration
These “antifibrotic” effects are likely related to the inhi- (FDA) required an additional trial to approve the use
bition exerted by pirfenidone on the phosphorylation of pirfenidone in the United States; this was called the
induced by TGF-β of Smad3, p38, and Akt, key factors ASCEND trial. In ASCEND, pirfenidone significantly
for the regulation of this pathway.14 Other studies per- reduced the proportion of patients who experienced a
formed on the bleomycin model of lung fibrosis have decline of 10% or more in FVC and the relative risk of
described a diminished presence of profibrotic fac- death or disease progression.19 All patients randomized
tors such as TGF-β in the bronchoalveolar lavage fluid to pirfenidone 2403 mg/day or placebo in the CAPACITY
(BALF) of pirfenidone-treated animals and a concur- and ASCEND trials were included in a pooled analy-
rent reduction of the accumulation of hydroxyproline, sis aimed to assess safety and efficacy of pirfenidone at
Col I, and Col III in BAL and lung tissue.15 1 year of treatment. This study suggested that pirfeni-
Even if the exact mechanisms through which pir- done reduces the risk of death by 48% and improves
fenidone acts were only partially known, the identifica- PFS by 38% in IPF patients compared to placebo.20
tion of its antioxidant and antiinflammatory properties As already underlined in this chapter, the under-
along with a surprising antifibrotic activity redefined standing of the pathogenic mechanisms of a disease
the clinical interest for this pleiotropic drug. In spite of can be important to developing new drugs. Similarly,
that, it took a few years to have the first double-blind the comprehension of the mechanisms of action of a
randomized clinical trial testing the efficacy of pirfeni- drug is useful to better understand its potential thera-
done in IPF patients. This was a Japanese Phase II study peutic effect as well as to predict and possibly avoid side
that was stopped after only 9 months because it was effects. For this reason, preclinical and clinical studies
considered unethical to continue the study consider- thoroughly evaluated the safety profile of pirfenidone.
ing the increased number of acute exacerbations in the To further evaluate the long-term safety and toler-
placebo group compared with the group treated with ability of the drug, patients who participated in five
pirfenidone. The primary endpoint of the study was not different clinical trials were analyzed: three Phase III
achieved—there was no difference in the saturation of multinational trials (CAPACITY 004, 006, and ASCEND)
peripheral oxygen under exertion between the placebo and the two ongoing open label studies RECAP (002,
and the pirfenidone arm, but a significant reduction 012). This study collected data from a large and well-
in the decline of vital capacity was described in the defined cohort of IPF patients who were treated with
28 Interstitial Lung Disease

pirfenidone and followed for up to 9.9 years. The major side effect does not resolve spontaneously. Pharmaco-
adverse events were gastrointestinal (GI) events [nau- logic treatment with oral and/or topical steroids and
sea (37.6%), diarrhea (28.1%), dyspepsia (18.4%), antihistamines is suggested in cases of persistence of
and vomiting (15.9%)] and rash (25.0%). Elevations the skin reaction.
in blood levels of aminotransferase greater than three On one hand, it is helpful to reduce the dose of pir-
times the upper limit of normal occurred in 3.1% of fenidone to reduce the risk of adverse events, but on the
patients. All of these events were mild to moderate in other it is necessary to maintain the correct serum con-
severity, transient with dose modification, and without centration of the drug to maximize its effectiveness. It
clinical long-term consequences.21 is important to adjust the dose of pirfenidone carefully
Many studies have been conducted to explain the in each patient to balance tolerability with expected
molecular mechanism responsible for GI events. Ani- efficacy.
mal studies showed that pirfenidone reduces the rate of It has been estimated that 70–80% of pirfenidone is
gastric emptying and small intestinal transit, and this metabolized by cytochrome CYP1A2 and by CYP2C9,
effect can be relieved by the administration of proki- 2C19, 2D6, and 2E1. Thus the concomitant use of
netic drugs. It was demonstrated that the concomitant inhibitors of CYP1A2, such as grapefruit and fluvox-
administration of mosapride and splitting pirfenidone amine, should be avoided during the treatment with
doses during meals improve the decrease in gastric pirfenidone because they could increase the exposure
emptying rate caused by pirfenidone.22 to the drug and the risk of side effects. Conversely,
Rubino et al. studied the pharmacokinetic features strong inducers of CYP1A2, such as cigarette smoke,
of pirfenidone in a group of healthy volunteers and should be avoided because they may accelerate drug
described the effects of the simultaneous assumption clearance reducing its effectiveness.
with food. The combination of pirfenidone with food The oral administration of pirfenidone is followed
caused a reduction and delay in the absorption of the by a rapid degradation of the molecule in two metabo-
drug and a reduced plasma concentration of both drug lites, 5-hydroxypirfenidone and 5-carboxypirfenidone,
and metabolites. These results have an important clini- and their elimination in the urine.26 These two metabo-
cal repercussion because a correlation between plasma lites still have antifibrotic properties, as demonstrated
concentration and the risk of adverse events has been by Togami et al., and their distribution in lung tissue is
demonstrated, suggesting that food may reduce the low if compared with the liver and kidneys because of a
incidence of GI events.23 low affinity between metabolites and lung tissue.27 The
On the basis of these previous studies, it is suggested direct consequence of this observation is the intriguing
that patients take the drug during or at the end of the idea that pirfenidone and its metabolites could reach
meal, start with a longer initial dosing titration, and higher concentrations in the lung when administered
take a prokinetic drug if necessary to reduce the inci- by inhalation instead of the traditional oral route.
dence of GI events. If GI events occur, it is suggested
to reduce the dose and, if the adverse event persists,
temporarily discontinue treatment until the symptoms THE STORY OF NINTEDANIB
become tolerable.24 Based on its prognosis, IPF has been generically com-
Photosensitivity is the second most common pared to malignant disease. Similar to cancer, IPF is
adverse event that may be caused by pirfenidone. A associated with risk factors such as smoking and/or
study performed on guinea pigs demonstrated that environmental or professional exposure, and the pres-
pirfenidone is photoreactive, which means that it can ence of a specific genetic background is considered fun-
absorb ultraviolet B and A (UVB, UVA) radiation, caus- damental for the occurrence of the disease. In addition,
ing the generation of reactive oxygen species (ROS) and IPF and cancer share a number of pathogenic pathways
lipid peroxidations that may be responsible for sun- such as genetic and epigenetic alterations, abnormal
burn. After oral administration, pirfenidone distribu- expression of miRNAs, cellular and molecular aber-
tion to the UV-exposed tissue (skin) is even higher than rance including an altered response to regulatory
in lung tissue.25 Based on these findings, it is suggested signals, delayed apoptosis, reduced cell-to-cell commu-
that patients avoid sun exposure, protect skin with sun- nication, and above all the activation of specific signal
screen active against both UVA and UVB radiation, and transduction pathways.28 These signaling pathways are
to wear protective clothes. Photosensitivity reaction is fundamental to translate any extracellular signal in an
also related to pirfenidone dose, so that dosage adjust- equivalent cell response and action. In this process, an
ment or temporary discontinuation is indicated if the extracellular signaling molecule stimulates a specific
CHAPTER 2 Pathobiology of Novel Approaches to Treatment 29

receptor inside the cell or on its surface, producing a fibroblast. The expression of PI3K p110, alpha, beta, and
series of intracellular events that lead, depending on delta isoforms does not differ between normal and IPF
the cellular type, to gene activation, metabolism altera- tissue–derived fibroblasts, whereas immune reactivity
tions, or in general, changes in cell behavior. for p110 gamma was much stronger in both IPF lung
A large variety of signal transduction pathways homogenates and ex vivo fibroblast cell lines. Further-
are activated both in cancer and in IPF, and many more, both p110 gamma pharmacologic inhibition and
studies have demonstrated their involvement in the gene silencing significantly inhibited the proliferation
pathogenesis of these two diseases. It has been largely rate as well as α-SMA expression in IPF fibroblasts.
demonstrated that alterations of the Wnt signaling These data strongly suggest that this isoform of PI3K
transduction pathway have clinical relevance in the may have a role in the pathogenesis of IPF, representing
pathogenesis of cancer.29 Indeed, the Wnt/β-catenin a novel specific pharmacologic target.33
signaling pathway regulates the expression of mol- In this regard, a recent study has demonstrated that
ecules involved in tissue invasion such as matrilysin, oral administration of a p110 gamma inhibitor pre-
laminin, and cyclin-D1, and it is also involved in a bio- vents bleomycin-induced pulmonary fibrosis in rats.
logically relevant crosstalk with TGF-β. It is interesting More recently researchers have focused their atten-
to note that the Wnt/β-catenin pathway is also strongly tion on another signal transduction pathway activated
activated in IPF lung tissue as evidenced by extensive in IPF and frequently altered in many cancers: the
nuclear accumulation of β-catenin at different involved JAK–STAT signaling pathway. This system is a major
sites such as bronchiolar proliferative lesions, damaged signaling alternative to the second messenger system
alveolar structures, and fibroblast foci.30 The functional such as cyclic AMP, cyclic GMP, inositol triphosphate,
significance of β-catenin has been well demonstrated diacylglycerol, and calcium. It transmits chemical sig-
by the description of an intense immunoreactivity for nals from outside the cell, through the cell membrane
β-catenin and by the subsequent expression of high lev- toward the cytoplasm, and then into the nucleus. Here,
els of cyclin-D1 and matrilysin. Caraci et al. have dem- it affects the activity of gene promoter regions, causing
onstrated that the Wnt/β-catenin pathway may also be DNA transcription and cell activation. One of the regu-
activated by TGF-β through the induction of α-SMA latory mechanisms of the JAK-STAT signaling pathway
expression via extracellular-regulated kinases (ERK)1/2 is represented by the SOCS family (suppressor of cyto-
activation, glycogen synthase kinase-3beta (GSK-3beta) kine signaling proteins). Bao et al. have demonstrated
inhibition, and nuclear β-catenin translocation that that in IPF patients there is a lower expression of SOCS1
leads to fibroblast activation and collagen production and this finding has been related to more severe mani-
in human lung fibroblasts.31 festations of the disease and to a worse prognosis.34
Several studies are currently exploring the anticancer One of the most studied signaling pathways,
effect of specific inhibitors of the Wnt pathway such as strongly activated not only in many cancers, but also
ICG-001. It is interesting that the same compound has in IPF, is the tyrosine kinase pathway. Tyrosine kinase
already been used with success in an animal model of is an enzyme that acts as a sort of “on” or “off” switch
lung fibrosis. Another key process not only described that regulates many cellular functions. Mutations can
in carcinogenesis but also likely involved in causing turn tyrosine kinases in a nonstop functional state that,
IPF is the altered regulation of apoptosis. In this case under specific circumstances, may lead to the initia-
the PI3K/Akt signaling pathway plays an important tion or progression of cancer. More recently this path-
role. This pathway is involved in the regulation of cell way has also been studied in the context of the wound
growth, proliferation, and survival. Phosphoinositol- healing process and fibrosis.35 Indeed, tyrosine kinases
3-kinase (PI3K) stimulates the synthesis of phospha- catalyze the phosphorylation of tyrosine residues in
tidylinositol-3,4,5-triphosphate, which causes the proteins such as platelet-derived growth factor (PDGF),
activation of Akt-modulated cellular processes such as fibroblast growth factor (FGF), or vascular endothelial
protection from apoptosis.32 Based on its primary struc- growth factor (VEGF), regulating a wide array of cel-
ture and in vitro lipid substrate specificity, it is possible lular functions, including cell growth, differentiation,
to recognize three different classes of PI3K: classes I, II, adhesion, motility, and regulation of cell death.
and III. Recently, Conte et al. assessed the expression PDGF, a ligand of tyrosine kinase receptor, is a
of class I PI3K p110 isoform in IPF lung tissue as well heterodimeric molecule expressed in different cells,
as in tissue-derived fibroblast cell lines and evaluated including fibroblasts. Binding of PDGF to its receptor
the effect of the selective inhibition of p110 isoforms (PDGFr) leads to autophosphorylation of the receptor
on the proliferation and fibrogenic activity of the IPF and activates intracellular pathways, such as Ras, Raf,
30 Interstitial Lung Disease

and MEK, and extracellular ones such as ERK and PI3K. carcinoma and other cancers, have been tested for the
PDGF is a potent growth factor for fibroblasts in vitro, treatment of IPF. Imatinib mesylate, a specific inhibi-
and some mediators such as TGF-β and basic FGF have tor of PDGFr, was evaluated for its potential antifibrotic
PDGF-dependent profibrotic activities. PDGF is more effects in preclinical and clinical studies. It was able to
highly expressed in epithelial cells and alveolar mac- inhibit fibroblast proliferation and collagen deposition
rophages in the lungs of patients with IPF.36 Moreover, in vitro and in vivo, but when it was evaluated in a clini-
high levels of PDGF have been shown in irradiated cal trial, no benefit in slowing disease progression was
mice, and the use of a PDGFr inhibitor has attenuated found.48 To achieve a more potent antifibrotic effect,
the development of pulmonary fibrosis in animal mod- other compounds able to exert an inhibition of multiple
els induced by radiation.37 receptors were tested. BIBF 1000, an inhibitor of PDGFr,
In addition to PDGF, FGF is involved not only in VEGFr, and FGFr, was first evaluated in a mice model of
carcinogenesis but also in fibrogenesis. FGF receptors bleomycin-induced pulmonary fibrosis and in an ex vivo
(FGFrs) are present on epithelial cells and fibroblasts and fibroblast differentiation assay. It was found to attenuate
mediate epithelial–mesenchymal transition and fibro- fibrosis by reducing the expression of profibrotic factors
blast transition into myofibroblasts.38 FGF and more and by decreasing collagen deposition.49
specifically two members of this family of growth factors Along the same lines, BIBF1120 (nintedanib), a
such as FGF1 and FGF2 are potent activators of fibroblast triple kinase inhibitor, with potent suppressing effects
proliferation and collagen production. The binding of on VEGFr, PDGFr, and FGFr, was also evaluated. Nint-
FGF to FGFr causes autophosphorylation and activation edanib exerts its inhibitory effect on PDGF, FGF, and
of downstream signaling via PI3K/Akt, ERK1/2, and Ras/ VEGF through the occupation of the intracellular ATP-
Raf/MAPK pathways.39 Interestingly, TGF-β regulates the binding pocket of the tyrosine kinases.50 This drug
FGFr cascade, inducing the upregulation of FGFr-1 and interferes with fibroblast proliferation as well as with
the release of FGF2 in human lung fibroblasts.40,41 In the secretion and deposition of ECM. It has been shown
animal models of pulmonary fibrosis FGFr-1 is strongly in a mice model that nintedanib reduces TIMP1 and
upregulated, whereas it is not expressed in control ani- TIMP2, which in turn inhibit matrix metalloprotein-
mals, suggesting its involvement in the fibrotic process.42 ases responsible for the degradation of collagen type I
This is substantiated by the observation that inhibition and II. It also reduces collagen deposition and inhibits
of FGF signaling ameliorates bleomycin-induced pulmo- the transformation of fibroblasts into myofibroblasts
nary fibrosis.43 In addition, increased levels of FGF have in human lung fibroblasts of IPF patients51 by TGF-β.52
been detected in lung cells of patients affected by IPF.44 More recently, nintedanib was studied in clinical
If the role of VEGF in cancer is well recognized, its trials as a potential antifibrotic therapy in IPF, demon-
involvement in IPF is less clear. VEGF is produced by strating that treatment with this drug may reduce the
alveolar and bronchial epithelial cells, airway smooth decline in lung function of IPF patients by about 50%.
muscle cells, fibroblasts, and endothelial cells. VEGFr Based on these results, nintedanib has been approved
is characterized by the typical intracellular domain that as a new therapy in patients with IPF. This is relevant,
undergoes autophosphorylation as a consequence of because for the first time, based on the observation of
the binding VEGF-VEGFr, activating different signaling the pathogenic similarities between IPF and cancer, a
pathways (p38, PI3K, Ras) involved in cell prolifera- drug “borrowed” from another field, in this case oncol-
tion and migration. In IPF patients, VEGF expression is ogy, has been “repositioned” for IPF. “Drug reposi-
low in fibroblasts and leukocytes in fibrotic lesions, but tioning” is an interesting and convenient strategy that
increased in endothelial cells and AECs.45 Parkas et al. may save time and reduce costs in the development of
reported direct effects of VEGF-A in lung fibroblasts, a specific drug. The safety profile of the explored drug
enhancing collagen I expression induced by β.46 More is already known so that the risk of adverse events is
recently, high VEGF serum concentration in IPF patients reduced. In addition, the mechanism of action of a
was related to a worse 5-year survival rate.47 The precise repositioned drug is known and can be used in those
role of VEGF-VEGFr signaling in IPF is controversial, diseases where a similar mechanism is involved. Today,
and further studies are required to clarify its exact role. computational strategies are available to explore the
Nevertheless, VEGF and even more PDGF and FGF have mechanisms of action of approved drugs and to match
a potent effect on fibroblast activation and proliferation, them with cellular and/or molecular targets involved in
and their inhibition is expected to reduce fibrosis in IPF. the pathogenesis of virtually any disease.
Based on the previous discussion, tyrosine kinases The first randomized, double-blind, placebo-con-
receptor inhibitors, widely used in non–small cell lung trolled, Phase II trial conducted to evaluate the efficacy
CHAPTER 2 Pathobiology of Novel Approaches to Treatment 31

of nintedanib in IPF patients (TOMORROW study) diarrhea develops. It has been supposed that patients
demonstrated, in the group receiving BIBF1120, a reduc- treated with tyrosine kinase inhibitors that are directed
tion of 68.4% in the annual rate of decline in FVC and against EGFR experience diarrhea because of the exces-
a lower incidence of acute exacerbations compared with sive chloride secretion and deficient sodium absorption.
placebo.53 Following the results of this clinical trial, In the normal colon, sodium absorption and chlo-
two other randomized double-blind Phase III trials ride secretion are stimulated directly by intracellular
(INPULSIS-1, INPULSIS-2) were performed. A total of messengers such as c-AMP and intracellular calcium.
1066 IPF patients were randomized in a 3:2 ratio to receive EGFR is a negative regulator of chloride secretion and
placebo or BIBF1120. This treatment reduced the rate of is expressed in GI normal mucosa. EGFR inhibitors
decline in FVC over the 52-week study period in both could increase chloride secretion, inducing a secretory
INPULSIS trials. The secondary endpoints were repre- diarrhea.57 Diarrhea following imatinib has also been
sented by the time to the first exacerbation and the change related to c-Kit inhibition, highly expressed in the inter-
in total St. George’s Respiratory Questionnaire (SGRQ) stitial cells of Cajal, which have a pacemaker function in
score, but no consistent differences were found between the intestine, so that c-Kit inhibition can cause an altera-
the placebo and the nintedanib groups.54 Recently, pooled tion in the intestinal motility resulting in diarrhea.58
analyses of data from the three trials (­ TOMORROW, Nintedanib dose reduction rapidly lowers the
INPULSIS 1 and 2) were performed to obtain an overall incidence and severity of diarrhea. Sometimes the
estimate of the clinical efficacy of nintedanib in 1231 IPF addition of loperamide is indicated as a useful treat-
patients. Nintedanib consistently slowed disease progres- ment to decrease intestinal motility.
sion, reducing the annual rate decline in FVC compared
with the placebo group and causing a modest change in
SGRQ total score from the baseline. This analysis also NAC (N-ACETYLCYSTEINE) OR NOT NAC,
demonstrated a positive trend toward a reduction in risk THAT IS THE QUESTION
for all-cause and respiratory mortality.55 The use of antioxidants in the treatment of IPF is based
Nintedanib, after its oral administration, is rapidly on the hypothesis that the disease can develop from an
metabolized by hepatocytes into metabolite BIBF1202. oxidant-antioxidant imbalance responsible for repeated
This metabolite is eliminated through the liver and AEC injuries followed by fibrotic repair. ROS play an
feces, while the excretion in the urine is minimal.56 important role in the fibrotic process as demonstrated
The hepatic metabolism of nintedanib is CYP450 by the reduced levels of glutathione (GSH) in BAL of
independent; therefore, the drug interactions are very IPF patients.59 NAC is a precursor of GSH and a power-
limited if compared with pirfenidone. Particular care ful antioxidant scavenger of ROS. The first clinical trial
should be used in patients affected by coagulopathy at to test oral NAC combined with prednisone and aza-
risk of bleeding because nintedanib, as an inhibitor of thioprine was the IFIGENIA study, which showed that
VEGF, can theoretically cause bleeding. The most fre- NAC combination therapy given for 1 year significantly
quent adverse events recorded during the studies were reduced the functional decline of IPF in terms of vital
GI events, especially diarrhea, which occurred in 61.5% and diffusing capacity compared to prednisone and
of patients in INPULSIS-1 and 63.2% in INPULSIS-2, azathioprine alone. In spite of these encouraging results
and increased liver enzymes levels, which occurred in it took several years to have additional information on
4.9% of patients in INPULSIS-1 and 5.2% of patients the therapeutic role of NAC in IPF. In 2011 a specific
in INPULSIS-2, respectively. In the pooled data analy- study, named “PANTHER,” was initiated to assess the
sis, diarrhea was the most frequent adverse event in the efficacy of the triple therapy (prednisone, azathioprine,
nintedanib group (61.5% of patients treated with nint- and NAC) compared to NAC alone and placebo in a
edanib vs. 17.9% of patients treated with placebo). In 1:1:1 ratio. It is well known that the trial failed, showing
general, adverse events are reversible with reducing the an increase in the number of deaths and hospitaliza-
dose of nintedanib or stopping the treatment, without tions in the group treated with the triple therapy. The
clinically significant consequences.55 study was continued to explore the difference between
Diarrhea was described as the most frequent adverse NAC monotherapy and placebo. This part of the
event following nintedanib assumption. The pathophys- PANTHER study showed quite clearly that NAC alone
iologic mechanism of drug-induced diarrhea is still not was not superior to placebo in reducing FVC decline.60
completely understood, but some studies conducted for A noteworthy recent observation suggests that a single
oncologic tyrosine kinase inhibitors could be helpful to nucleotide polymorphism in the TOLLIP gene, known
understand the molecular mechanisms through which to be involved in lung host defense, may influence the
32 Interstitial Lung Disease

effect of antioxidant therapy in IPF patients. These data score and longer survival in those patients receiving ant-
suggest that IPF patients with the rs3750920 TOLLIP acid treatment.67 It was also reported that medication
TT genotype may respond to NAC treatment.61 Inhaled for GERD and Nissen fundoplication were indepen-
NAC in monotherapy has also been tested in a multi- dent predictors of longer survival for IPF. A more recent
center, perspective, randomized, controlled clinical trial study has demonstrated a slower decline of functional
in Japanese patients with early-stage IPF. This study capacity in IPF patients affected by GERD and treated
demonstrated a possible beneficial effect of the drug with antacid therapy suggesting, once again, a possible
in patients with early-stage IPF.62 Recently, a retrospec- benefit for antacid treatment.68 More recently, Lee et al.
tive observational study involving 22 Japanese patients have reported in a cohort of 786 IPF patients a protec-
affected by early untreated IPF was conducted to assess tive effect of PPI used for at least 4 months.69 Based on
the efficacy on FVC and GSH levels. This study suggested these results the current guidelines for IPF treatment
that patients with higher oxidative stress could develop provide a conditional recommendation for the use
a better response to NAC treatment, likely because NAC of PPIs in IPF patients.70 Recently, a post hoc analysis
replenishes GSH levels, correcting the oxidant–antioxi- from the placebo groups of three trials of pirfenidone
dant imbalance.63 Thus oxidative stress can be a possible (CAPACITY and ASCEND) showed that antacid therapy
marker of response to NAC treatment. These results sup- is not related to a clinical significant improvement in
port new perspective clinical trials to test the efficacy of outcomes after 52 weeks of treatment in either PFS or
either oral NAC based on genotypic stratification of IPF mortality.71 According to this study IPF patients with
patients or inhaled NAC based on oxidative stress levels advanced disease (defined as FVC <70%) receiving ant-
of patients with an early-stage disease. These approaches acid treatment had a higher infection rate than patients
will allow for identifying subgroups of patients respond- who did not receive antacid treatment. New studies are
ing to NAC, hopefully opening a fascinating view on the required to assess the real efficacy of antacid treatment,
first attempt of personalized therapy for IPF. As things especially in advanced IPF stages.
stand, the real effectiveness of NAC is still debated and
more studies are needed.
CURRENT CLINICAL TRIALS: THE FUTURE
IS COMING
ANTACID TREATMENT: MORE DOUBTS The pathobiology of IPF is complex; several cellular
THAN CERTAINTIES and molecular mechanisms are involved in its occur-
Gastroesophageal reflux is highly prevalent in patients rence and progression. Some of these mechanisms are
with IPF compared with patients affected by other inter- obscure, some others are partially clear, and some of
stitial lung diseases and healthy people.64 It has been them are well understood and are being used to develop
proposed that in some predisposed patients, gastro- new therapeutic strategies. By virtue of this, a great deal
esophageal reflux disease (GERD) and repetitive micro- of preclinical and clinical work has been performed in
aspiration of acid in the airways can cause continuous the last few years, and much more is currently under way
AEC injury, triggering those pathogenic mechanisms to explore novel treatments and therapeutic strategies
underlying IPF.65 Microaspiration also may be involved for IPF. All of them are obviously inspired by the new
in acute exacerbations of IPF as suggested by the dis- pathogenic knowledge that comes every day from the
covery of high levels of pepsin in BALF of IPF patients research field. Recently, there has been interest in novel
with acute exacerbations. The logical consequence of agents that target type 2 T-helper cell (Th2) inflamma-
these observations was to investigate the role of proton tory pathways such as interleukin 13 (IL-13). Phase II
pump inhibitors (PPI) in IPF. Indeed in the literature randomized, double-blind, placebo-controlled trials of
there is some evidence showing a positive role for ant- two monoclonal antibodies against IL-13, lebrikizumab
acid treatment in IPF. and tralokinumab, are currently under way. The ratio-
It has been also demonstrated that PPI could also nale for the use of antimonoclonal antibodies against
have antifibrotic and antiinflammatory properties IL-13 is based on some studies that nicely showed
through the direct suppression of proinflammatory that IL-13, a cytokine secreted by Th2 cells, is a strong
cytokines (such as TNF-α, IL1-β, IL-6) and via the stimulator of fibroblast proliferation, acting through
downregulation of TGF-β receptors involved in fibro- multiple mechanisms, including interaction with TG-
blast proliferation.66 beta and CCL2, thus inducing fibrosis.72 However, the
In a noncontrolled retrospective study of 204 IPF exact mechanism whereby IL-13 promotes pulmonary
patients, Lee et al. observed a lower radiologic fibrosis fibrosis has not been completely defined, with studies
CHAPTER 2 Pathobiology of Novel Approaches to Treatment 33

showing either a TGF-β–dependent73 or a TGF-β–inde- Lung fibroblasts are dependent on CTGF that regulates
pendent pathway.74 IL-13 signals through its respective their attachment to the matrix and subsequently their
receptor subunits via JAK2–STAT6–dependent and AP1- migration, differentiation, and apoptosis.82 CTGF is
dependent pathways,75 but some studies have suggested upregulated in tissues subjected to mechanical stress
that IL-13 Ra2 also has a role in the signaling mecha- as may happen in lung fibrosis,83 and it is believed
nisms.76 Thus, it has been suggested that IL-13 and its that activation of fibroblasts by TGF-β also induces
receptor subunits are involved in the pathogenesis of CTGF expression. Increased levels of CTGF have been
pulmonary fibrosis, and even if the correct mechanism recorded in BALF and lung tissue of IPF patients.84 It
is still not completely understood, they are considered has also been proven in animal models that targeting
targetable. In this regard Murray et al. studied biopsy CTGF diminishes fibrosis.85 Raghu et al. for the first
samples from IPF patients who experimented a rapidly time studied the efficacy of anti-CTGF monoclonal
progressing form of IPF, demonstrating that the IL-13 antibodies for the treatment of IPF patients in a Phase
pathway was enhanced compared to IPF patients with II clinical trial,86 and following these results a new
a slower decline in FVC. It has also been suggested that phase II clinical trial was started to assess efficacy and
the inhibition of IL-13 promotes lung repair and helps safety of this new molecule.87
to restore epithelial integrity; therefore, targeting IL-13 In parallel to the numerous pharmacological inves-
inhibits fibrotic processes and promotes the repairing tigations, cell-based strategies have been explored as
process in the lung.77 a possible alternative to pharmacologic treatments.
According to the recent evidence, fibroblasts in IPF Cell-based therapies represent an innovative possibil-
lungs could partially derive from circulating monocytes ity to treat patients with lung fibrosis because trans-
that leave the circulation, reach the lung, and then dif- planted cells could be potentially able to proliferate
ferentiate into fibrocytes through a process mediated and differentiate into alveolar cells replacing the
by a family of proteins named pentraxins.78 There are damaged epithelial alveolar cells and some benefits
three main systemic pentraxins in humans: SAP (serum could be obtained from the paracrine properties of
amyloid P component), CRP (C-reactive protein), and the newly administered cells. Different kinds of cells
PTX3. It has been demonstrated that PTX3 promotes have been proposed for a possible new IPF treatment,
fibrocyte differentiation, whereas injections of SAP in including not only AECs, but also lung mixed epithe-
a mouse model of pulmonary fibrosis inhibit fibro- lial cells, mesenchymal cells, stem cells, and circulat-
sis.79 A previous study has also showed the potential ing endothelial progenitor cells.
for pentraxin SAP to inhibit bleomycin-induced lung Serrano-Mollar et al. have reported that trans-
fibrosis through the inhibition of monocyte differentia- planting AECsII, obtained by in vitro differentiation
tion in fibrocytes in vitro and in vivo.80 On the basis of healthy organs, in bleomycin-induced pulmonary
of these evidences, a new drug PRM-151 (recombinant fibrosis lung could reverse the fibrotic process.88 Stem
human pentraxin 2, known as serum amyloid P) was cells also represent a promising option for therapeu-
tested in IPF patients in a randomized, double-blind, tic application in IPF and because of their capacity to
placebo-controlled trial that assessed safety, tolerability, differentiate into different cell lines and to exert both
and pharmacokinetics of single ascending intravenous antiproliferative and antiinflammatory effects. Stem
doses of this drug administered to healthy subjects and cell properties have been investigated in murine mod-
IPF patients. The drug was well tolerated, and just tran- els of lung fibrosis in different studies, showing that
sient skin reactions were observed as adverse events. A murine bone marrow–derived mesenchymal stem cells
30–40% decrease in the number of fibrocytes 24 h post are able to target damaged pulmonary areas and to
dose was also registered, suggesting that drug admin- reduce inflammation and collagen deposition in the
istration may be associated with a reduction of these lung.89 Similar results were achieved using transplanted
cells in IPF. Interestingly, FVC showed a trend toward placenta-derived stem cells, demonstrating neutrophil
improvement in the treated group.81 However, the num- infiltration decrease and a significative reduction in the
ber of patients of this preliminary, although very inter- severity of fibrosis.90
esting, study was very low. Further and larger studies are The most important issues, concerning the type
needed to test the real efficacy of this molecule. A clini- of cells used, regard cell recruitment to the lung, the
cal trial exploring the efficacy of pentraxin 2 on a larger induction of differentiation in epithelial lung cells,
population of patients with IPF is currently under way. and the risk of cancer, especially teratoma. Differ-
Connective tissue growth factor (CTGF) is a matri- ent routes of cell administration were tested, intra-
cellular protein that connects cell surfaces and ECM. venously or through intraperitoneal or intratracheal
34 Interstitial Lung Disease

instillation, showing different capacity to reach the For these reasons, during the last few years the
lung, but it is still debatable which is the more effec- scientific attention has focused on some steps of the
tive. Other issues are unsolved such as the effective cell fibrogenic process, characterized by the activation of
dose equivalent for cell therapy, defined as the mini- multiple and different pathways that may represent
mum cell number required to have a significant result. specific therapeutic targets. Indeed, pirfenidone was
Few clinical trials have been conducted so far on cell- tested for its pleiotropic properties, as a drug able to
based therapy in IPF, and only some of these have act in different pathways of the fibrogenic process,
been published, with limited results mainly focused and its efficacy has been proved in several preclinical
on safety. Safety and tolerability are the most impor- and clinical studies that have made pirfenidone the
tant issues because of the risk of teratoma and other first drug approved for the treatment of IPF. The story
risks associated with isolation, culture, and storage of of nintedanib, a drug born to be used in oncology for
cells. The most commonly used cells are mesenchymal non–small cell lung cancer, is also interesting. By vir-
cells, which are characterized by low immunogenic- tue of the observation of some pathobiologic similari-
ity and low risk of teratoma. The future development ties between cancer and IPF, such as the activation of
of cell therapy is fascinating and possibly realistic the same signaling transduction pathways, nintedanib
using bioengineering approaches that according to has been repositioned and eventually approved for the
recent studies will make it possible to use decellular- treatment of IPF. The approval of pirfenidone first and
ized whole lungs as a scaffold for subsequent cellular then of nintedanib represents a fundamental if not
implantation.91 historic moment for patients affected by IPF and their
families. The opportunity to have a treatment slowing
down the progression of the disease or reducing the
CONCLUSION: IS PATHOBIOLOGY risk of death has helped to attenuate the fatalism that
IMPORTANT FOR NOVEL APPROACHES for a long time has been associated with the diagno-
TO TREATMENT? sis of IPF. The current approach to treatment is based
Is the understanding of the pathobiology of IPF really on sequential therapy, so that one of the two approved
important for exploring novel approaches to treat- drugs is started and when it is believed to be ineffective
ment? The answer to this question is simple and obvi- or side effects due to the drug are severe, it is possible to
ous: yes. In spite of that, and against any clinical and switch to the alternative drug. Similar to cancer, for the
pathobiologic evidence, IPF has been treated with first time exists the possibility to have a first-line and,
steroids and immunosuppressants for many years. As as alternative, a second-line of treatment. Two differ-
already discussed in this chapter, this was mainly due ent clinical trials are currently under way exploring the
to the absence of other effective medical treatments, to safety of the association of pirfenidone and nintedanib,
some diagnostic inaccuracy that considered as IPF/UIP and hopefully, as in other diseases, there will be the
some forms of NSIP responding to steroids, and to the possibility to use in the same patients a combination
incomprehensible lack of proper studies showing the of drugs. The mechanisms of action of the two drugs
exact position of steroids and immunosuppressants in seem different, so it is reasonable to think of a possible
the therapeutic “scenario” of IPF. From this point of increase in their efficacy without necessarily having a
view the PANTHER study did justice to the “noninflam- sum of side effects.
matory hypothesis” confirming that antiinflammatory By virtue of basic research and new clinical obser-
treatment with steroids is not effective in IPF and even vations, the amount of information and data on the
harmful. Fortunately, in other cases the potential drugs pathobiology of IPF is growing day by day, offering new
studied were more in tune with the pathobiology of and more ample opportunities to either develop new
IPF. Today, IPF is considered an epithelial-driven pro- therapies or reposition known drugs for IPF. A number
cess triggered by an abnormal response of these cells of preclinical and clinical studies are currently under
to chronic injuries likely caused by specific risk factors way with the aim of finding new and more potent drugs
such as smoking or environmental exposures. Genetic or effective cell-based treatments for IPF and possibly
predisposition and aging play a fundamental role to personalize treatments, identifying subcategories
in altering the response of epithelial cells to chronic of patients on the basis of genetic backgrounds and
damage. The final result of all this is the activation of response to treatment. This will certainly be possible in
fibroblasts, and their recruitment, proliferation, and a future not so distant, provided that the tree of knowl-
differentiation into myofibroblasts, resulting in scar edge for the pathobiology of IPF continues to grow
formation and loss of lung function. florid and robust.
CHAPTER 2 Pathobiology of Novel Approaches to Treatment 35

REFERENCES 18. Noble PW, Albera C, Bradford WZ, et al. Pirfenidone in


1. Richeldi L, Davies HR, Ferrara G, et al. Corticosteroids for patients with idiopathic pulmonary fibrosis (CAPACITY):
idiopathic pulmonary fibrosis. Cochrane Database Syst Rev. two randomised trials. Lancet. 2011;377(9779):1760–
2003;3:CD002880. 1769.
2. Funke M, Geiser T. Idiopathic pulmonary fibrosis: the 19. King Jr TE, Bradford WZ, Castro-Bernardini S, et al. A
turning point is now! Swiss Med Wkly. 2015;145:w14139. phase 3 trial of pirfenidone in patients with idiopathic
3. Adamson IY, Young L, Bowden DH. Relationship of alveo- pulmonary fibrosis. N Engl J Med. 2014;370(22):2083–
lar epithelial injury and repair to the induction of pulmo- 2092.
nary fibrosis. Am J Pathol. 1988;130(2):377–383. 20. Noble PW, Albera C, Williamson Z, et al. Pirfenidone
4. Pardo A, Selman M. Idiopathic pulmonary fibrosis: for idiopathic pulmonary fibrosis: analysis of pooled
new insights in its pathogenesis. Int J Biochem Cell Biol. data from three multinational phase 3 trials. Eur Respir J.
2002;34(12):1534–1538. 2016;47:243–253.
5. Idiopathic Pulmonary Fibrosis Clinicl Research Network, 21. Lancaster L, Albera C, Williamson Z, et al. Safety of pir-
Raghu G, Anstrom KJ, et al. Prednisone, azathioprine, and fenidone in patients with idiopathic pulmonary fibrosis:
N-acetylcysteine for pulmonary fibrosis. N Engl J Med. integrated analysis of cumulative data from 5 clinical tri-
2012;366(21):1968–1977. als. BMJ Open Resp Res. 2016;3:e000105.
6. Ward HE, Nicholas TE. Alveolar type I and type II cells. 22. Itoh T, Koyabu K, Morimoto A, et al. Ameliorative effects
Aust N Z J Med. 1984;14(5, suppl 3):731–734. of mosapride or Rikkunshi-to on the suppression of gas-
7. Oku H, Nakazato H, Horikawa, et al. Pirfenidone sup- trointestinal motility by pirfenidone in rats. Jpn Pharmacol
presses tumor necrosis factor-alpha, enhances interleu- Ther. 2012;40:405–411.
kin-10 and protects mice from endotoxic shock. Eur J 23. Rubino CM, Bhavnani SM, Ambrose PG, et al. Effect
Pharmacol. 2002;446:167–176. of food and antiacids on the pharmacokinetics of pi-
8. Iyer SN, Gurujeyalakshmi G, Giri SN. Effects of pirfeni- rfenidone in older healthy adults. Pulm Pharmacol Ther.
done on transforming growth factor-beta gene expression 2009;22:279–285.
at the transcriptional level in bleomycin hamster model 24. Costabel U, Bendstrup E, Cottin V, et al. Pirfenidone in
of lung fibrosis. J Pharmacol Exp Ther. 1999;291:367–373. idiopathic pulmonary fibrosis: expert panel discussion on
9. Misra HP, Rabideau C. Pirfenidone inhibits NADPH- the management of drug-related adverse events. Adv Ther.
dependent microsomal lipid peroxidation and scavenges 2014;31:375–391.
hydroxyl radicals. Mol Cell Biochem. 2000;204:119–126. 25. Seto Y, Inoue R, Kato M, et al. Photosafety assessments
10. Raghu G, Masta S, Meyers D, Narayanan AS. Collagen on pirfenidone: photochemical, photobiological, and
synthesis by normal and fibrotic human lung fibroblasts pharmacokinetic characterization. J Photochem Photobiol
and the effect of transforming growth factor-beta. Am Rev B. 2013;120:44–51.
Respir Dis. 1989;140(1):95–100. 26. Huang NY, Ding L, Wang J, et al. Pharmacokinetics,
11. Shi J, Massague J. Mechanisms of TGFbeta signaling from safety and tolerability of pirfenidone and its major me-
cell membrane to the nucleus. Cell. 2003;113:685–700. tabolite after single and multiple oral doses in healthy
12. Gu L, Zhu YJ, Yang X, et al. Effect of TGF-beta/Smad sign- Chinese subjects under fed conditions. Drug Res (Stuttg).
aling pathway on lung myofibroblast differentiation. Acta 2013;63:388–395.
Pharmacol Sin. 2007;28(3):382–391. 27. Togami K, Kanehira Y, Tada H. Pharmacokinetic evalua-
13. Conte E, Gili E, Fagone E, et al. Effect of pirfenidone on tion of tissue distribution of pirfenidone and its metabo-
proliferation, TGf-beta-induced myofibroblast differentia- lites for idiopathic pulmonary fibrosis therapy. Biopharm
tion and fibrogenic activity of primary human lung fibro- Drug Dispos. 2015;36:205–215.
blasts. Eur J Pharm Sci. 2014;58:13–19. 28. Vancheri C. Idiopathic pulmonary fibrosis and cancer: do
14. Conte E, Fruciano M, Fagone E, et al. Inhibition of PI3K they really look similar? BMC Med. 2015;13:220.
prevents the proliferation and differentiation of human 29. Konigshoff M, Balsara N, Pfaff EM, et al. Functional Wnt
lung fibroblasts into myofibroblasts: the role of class I signalling is increased in idiopathic pulmonary fibrosis.
P110 isoforms. PLoS One. 2011;6(10):e24663. PloS One. 2008;3(5):e2142.
15. Nakayama S, Mukae H, Sakamoto N, et al. Pirfenidone 30. Chilosi M, Poletti V, Zamò A, et al. Aberrant Wnt/beta-
inhibits the expression of HSP47 in TGF-beta1-stimulated catenin pathway activation in idiopathic pulmonary
human lung fibroblasts. Life Sci. 2008;82(3,4):210–217. fibrosis. Am J Pathol. 2003;162(5):1495–1502.
16. Azuma A, Nukiwa T, Tsuboi E, et al. Double-blind, pla- 31. Caraci F, Gili E, Calafiore M, et al. TGF-beta1 targets the
cebo-controlled trial of pirfenidone in patients with idi- GSK-3beta/beta-catenin pathway via ERK activation in the
opathic pulmonary fibrosis. Am J Respir Crit Care Med. transition of human lung fibroblasts into myofibroblasts.
2005;171(9):1040–1047. Pharmacol Res. 2008;57(4):274–282.
17. Taniguchi H, Ebina M, Kondoh Y, et al. Pirfenidone in idi- 32. Bowley E, O’Gorman DB, Gan BS. Beta-catenin signalling
opathic pulmonary fibrosis. Eur Respir J. 2010;35(4):821– in fibro-proliferative disease. J Surg Res. 2014;138(1):141–
829. 150.
36 Interstitial Lung Disease

33. Conte E, Gili E, Fruciano M, et al. PI3Kp110gamma over- 48. Daniels CE, Lasky JA, Limper AH, et al. Imatinib treat-
expression in idiopathic pulmonary fibrosis lung tissue ment for idiopathic pulmonary fibrosis: randomized
and fibroblast cells: in vitro effects of its inhibition. Lab placebo-controlled trial results. Am J Respir Crit Care Med.
Invest. 2013;93(5):566–576. 2010;181(6):604–610.
34. Bao Z, Zhang Q, Wan H, et al. Expression of suppres- 49. Chaudhary NI, Roth GJ, Hilberg F, et al. Inhibition of
sor of cytokine signalling 1 in the peripheral blood of PDGF, VEGF and FGF signalling attenuates fibrosis. Eur
patient with idiopathic pulmonary fibrosis. Chin Med J. Respir J. 2007;29:976–985.
2014;127(11):2117–2120. 50. Hostettler KE, Zhong J, Papakonstantinou E, et al. Anti-
35. Grimminger F, Günther A, Vancheri C. The role of tyros- fibrotic effects of nintedanib in lung fibroblasts derived
ine kinases in the pathogenesis of idiopathic pulmonary from patients with idiopathic pulmonary fibrosis. Respir
fibrosis. Eur Respir J. 2015;45(5):1426–1433. Res. 2014;15:157.
36. Antoniades HN, Bravo MA, Avila RE, et al. Platelet-derived 51. Pardo A, Cabrera S, Maldonado M, et al. Role of matrix
growth factor in idiopathic pulmonary fibrosis. J Clin In- metallopreoteinases in idiopathic pulmonary fibrosis.
vest. 1990;86:1055–1064. Respir Res. 2016;17:23.
37. Abdollahi A, Li M, Ping G, et al. Inhibition of platelet- 52. Wollin L, Maillet I, Quesniaux V, et al. Anti fibrotic and
derived growth factor signalling attenuates pulmonary antiinflammatory activity of the tyrosine kinase inhibi-
fibrosis. J Exp Med. 2005;201(6):925–935. tor nintedanib in experimental models of lung fibrosis. J
38. Zou H, Nie XH, Zhang T, et al. Effect of basic fibroblast Pharmacol Exp Ther. 2014;349:209–220.
growth factor on the proliferation, migration and phe- 53. Richeldi L, Costabel U, Selman M, et al. Efficacy of a
notypic modulation of airway smooth muscle cells. Chin tyrosine kinase inhibitor in idiopathic pulmonary fibro-
Med J. 2008;121:424–429. sis. N Engl J Med. 2011;365(12):1079–1087.
39. Wollin L, Wex E, Pautsch A, et al. Mode of action of nint- 54. Richeldi L, du Bois RM, Raghu G, et al. Efficacy and safety
edanib in the treatment of idiopathic pulmonary fibrosis. of nintedanib in idiopathic pulmonary fibrosis. N Engl J
Eur Respir J. 2015;45:1434–1445. Med. 2014;370(22):2071–2082.
40. Thannickal VJ, Aldweib KD, Rajan T, et al. Upregulated 55. Richeldi L, Cottin V, du Bois RM, et al. Nintedanib in
expression of fibroblast growth factor (FGF) receptors patients with idiopathic pulmonary fibrosis: combined
by transforming growth factor beta 1(TGF beta1) medi- evidence from the TOMORROW and INPULSIS trials.
ates enhanced mitogenic responses to FGFs in cultured Respir Med. 2016;113:74–79.
human lung fibroblasts. Biochem Biophys Res Commun. 56. Woodcock HV, Molyneaux PL, Maher TM, et al. Reducing
1998;251:437–441. lung function decline in patients with idiopathic pulmo-
41. Khalil N, Xu YD, O’Connor R, et al. Proliferation of pul- nary fibrosis: potential of nintedanib. Drug Des Devel Ther.
monary interstitial fibroblasts is mediated by transform- 2013;7:503–510.
ing growth factor-beta1-induced release of extracellular 57. Uribe JM, Keely SJ, Traynor-Kaplan AE, et al. Phosphati-
fibroblast growth factor 2 and phosphorylation of p38 dylinositol 3-kinase mediates the inhibitory effect of
MAPK and JNK. J Biol Chem. 2005;280:43000–43009. epidermal growth factor on calcium-dependent chloride
42. Barrios R, Pardo A, Ramos C, et al. Upregulation of acid secretion. J Biol Chem. 1996;271:26588–26595.
fibroblast growth factor during development of experi- 58. Deininger MWN, O’Brien SG, For JM, et al. Practical
mental lung fibrosis. Am J Physiol. 1997;273:L451–L458. management of patients with chronic myeloid leukemia
43. Inoue Y, King Jr TE, Barker E, et al. Basic fibroblast growth receiving imatinib. J Clin Oncol. 2003;21:1637–1647.
factor and its receptors in idiopathic pulmonary fibrosis 59. Behr J, Degenkolb B, Krombach F, et al. Intracellular glu-
and lymphangioleiomyomatosis. Am J Respir Crit Care tathione and bronchoalveolar cells in fibrosing alveolitis:
Med. 2002;166:765–773. effects of N-acetylcysteine. Eur Respir J. 2002;19:906–911.
44. Yu ZH, Wang DD, Zhou ZY, et al. Mutant soluble ectodo- 60. Martinez FJ, de Andrade JA, Anstrom KJ, et al. Rand-
main of fibroblast growth factor receptor-2 III c attenu- omized trial of acetylcysteine in idiopathic pulmonary
ates bleomycin induced pulmonary fibrosis in mice. Biol fibrosis. N Engl J Med. 2014;370(22):2093–2101.
Pharm Bull. 2012;35:731–736. 61. Oldham JM, Ma SF, Martinez FJ, et al. TOLLIP, MUC5B
45. Ebina M, Shimizukawa M, Shibata N, et al. Heteroge- and the response to N-acetylcysteine among individuals
neous increase in CD 43 positive alveolar capillaries in with idiopathic pulmonary fibrosis. Am J Respir Crit Care
idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. Med. 2015;192(12):1475–1482.
2004;169:1203–1208. 62. Homma S, Azuma A, Taniguchi H, et al. Efficacy of in-
46. Farkas L, Farkas D, Ask K, et al. VEGF ameliorates pulmo- haled N-acetylcysteine monotherapy in patients with
nary hypertension through inhibition of endothelial ap- early stage idiopathic pulmonary fibrosis. Respirology.
optosis in experimental lung fibrosis in rats. J Clin Invest. 2012;17:467–477.
2009;119:1298–1311. 63. Muramatsu Y, Sugino K, Ishida F, et al. Effect of inhaled
47. Ando M, Miyazaki E, Ito T, et al. Significance of serum N-acetylcysteine monotherapy on lung function and
vascular endothelial growth factor level in patients with redox balance in idiopathic pulmonary fibrosis. Respir
idiopathic pulmonary fibrosis. Lung. 2010;188:247–252. Invest. 2016;54:170–178.
CHAPTER 2 Pathobiology of Novel Approaches to Treatment 37

64. Morehead RS. Gastro-oesophageal reflux disease and non- 78. Pilling D, Cox N, Vakil V, et al. The Long pentraxin
asthma lung disease. Eur Respir Rev. 2009;18(114):233– PTX3 promotes fibrocyte differentiation. PLoS One.
243. 2015;10(3):e0119709.
65. Schachter LM, Dixon J, Pierce RJ, et al. Severe gastroesoph- 79. Pilling D, Buckley CD, Salmon M, et al. Inhibition of
ageal reflux is associated with reduced carbon monoxide fibrocyte differentiation by serum amyloid P. J Immunol.
diffusing capacity. Chest. 2003;123(6):1932–1938. 2003;171:5537–5546.
66. Ghebremariam YT, Cooke JP, Gerhart W, et al. Pleiotropic 80. Pilling D, Gomer RH. Persistent lung inflammation and
effect of the proton pump inhibitor esomeprazole leading fibrosis in serum amyloid P component (APCs-/-) knock-
to suppression of lung inflammation and fibrosis. J Transl out mice. PLoS One. 2014;9(4):e93730.
Med. 2015;13:249. 81. Van den Blink B, Dillingh MR, Ginns LC, et al. Recom-
67. Lee JS, Ryu JH, Elicker BM, et al. Gastroesophageal reflux binant human pentraxin-2 therapy in patients with idi-
therapy is associated with longer survival in patients with opathic pulmonary fibrosis: safety, pharmacokinetics and
idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. exploratory efficacy. Eur Respir J. 2016;47(3):889–897.
2011;184:1390–1394. 82. Leask A, Abraham DJ. All in the CCN family: essential ma-
68. Lee JS, Collard HR, Anstrom KJ, et al. Anti-acid therapy tricellular signaling modulators emerge from the bunker. J
and disease progression in idiopathic pulmonary fibrosis: Cell Sci. 2006;119:4803–4810.
an analysis of data from three randomized controlled tri- 83. Liu F, Mih JD, Shea BS, et al. Feedback amplification of
als. Lancet Respir Med. 2013;1(5):369–376. fibrosis through matrix stiffening and COX-2 suppres-
69. Lee CM, Lee DH, Ahn BK, et al. Protective effect of proton sion. J Cell Biol. 2010;190:693–706.
pump inhibitor for survival in patients with gastroesoph- 84. Pan LH, Yamauchi K, Uzuki M, et al. Type II alveolar epi-
ageal reflux disease and idiopathic pulmonary fibrosis. J thelial cells and interstitial fibroblasts express connective
Neurogastroenterol Motil. 2016:15–192. tissue growth factor in IPF. Eur Respir J. 2001;17:1220–
70. Raghu G, Rochwerg B, Zhang Y, et al. An official ATS/ERS/ 1227.
JRS/ALAT clinical practice guideline:treatment of idio- 85. Lipson KE, Wong C, Teng Y, et al. CTGF is a central media-
pathic pulmonary fibrosis: executive summary. An update tor of tissue remodelling and fibrosis and its inhibition
of the 2011 clinical guideline. Am J Respir Crit Care Med. can reverse the process of fibrosis. Fibrog Tissue Repair.
2015;192(2):e3–e19. 2012;5(suppl 1):S24.
71. Kreuter M, Wuyts W, Renzoni E, et al. Antiacid therapy 86. Raghu G, Scholand MB, De Andrade J, et al. Safety and
and disease outcomes in idiopathic pulmonary fibrosis: a efficacy of anti-CTGF monoclonal antibody FG-3019 for
pooled analysis. Lancet Respir Med. 2016;4(5):381–389. the treatment of idiopathic pulmonary fibrosis (IPF): re-
72. Murray LA, Argentieri RI, Farrel FX, et al. Hyper- sults of phase II clinical trial two years after initiation. Am
responsiveness of IPF/UIP fibroblasts: interplay between J Respir Crit Care Med. 2014;189:A1426.
TGF beta1, IL13 and CCl2. Int J Biochem Cell Biol. 2008; 87. Raghu G, Scholand MB, De Andrade J, et al. FG-3019 anti-
10:2174–2182. connective tissue growth factor monoclonal antibody:
73. Lee CG, Homer RJ, Zhu Z, et al. Interleukin-13 induces tis- results of an open-label clinical trial in IPF. Eur Respir J.
sue fibrosis by selectively stimulating and activating trans- 2016;47(5):1481–1491.
forming growth factor beta(1). J Exp Med. 2001;194:809– 88. Serrano-Mollar A, Nacher M, Gay-Jordi G, et al. Intratra-
821. cheal transplantation of alveolar type II cells reverses
74. Kaviratne M, Hesse M, Leusink M, et al. IL-13 activates a bleomycin-induced lung fibrosis. Am J Resp Crit Care.
mechanism of tissue fibrosis that is completely TGF-beta 2007;176:1261–1268.
independent. J Immunol. 2004;173:4020–4029. 89. Ortiz LA, Gambelli F, McBride C, et al. Mesenchymal stem
75. Nishimura Y, Nitto T, Inoue T, Node K. IL-13 attenuates cell engraftment in lung is enhanced in response to bleo-
vascular tube formation via JAK2-STAT6 pathway. Circ J. mycin exposure and ameliorates its fibrotic effects. Proc
2008;72:469–475. Natl Acad Sci USA. 2003;100:8407–8411.
76. Zheng T, Liu W, Oh SY, et al. IL-13 receptor alpha2 se- 90. Cargnoni A, Gibelli L, Tosini A, et al. Transplantation of
lectively inhibits IL-13-induced responses in the murine allogeneic and xenogeneic placenta-derived cells reduces
lung. J Immunol. 2008;180:522–529. bleomycin-induced lung fibrosis. Cell Transplant. 2009;18:
77. Murray LA, Zhang H, Oak SR, et al. Targeting interleu- 405–422.
kin-13 with tralokinumab attenuates lung fibrosis and 91. Mendez JJ, Ghaedi M, Steinbacher D, et al. Epithelial cell
epithelial damage in a humanized SCID idiopathic differentiation of human mesenchymal stromal cells in
pulmonary fibrosis model. Am J Respir Cell Mol Biol. decellularized lung scaffolds. Tissue Eng. 2014;20:1735–
2014;50(5):985–994. 1746.
CHAPTER 3

Smoking-Related Interstitial
Lung Diseases
ROBERT VASSALLO • JAY H. RYU

KEY POINTS

• Substantial evidence implicates cigarette smoking as the principal etiologic factor responsible for the development of
respiratory bronchiolitis–interstitial lung disease (RB-ILD), desquamative interstitial pneumonia (DIP), and pulmonary
Langerhans cell histiocytosis (PLCH).
• Cigarette smoking is an important precipitant of acute eosinophilic pneumonia (AEP) and pulmonary hemorrhage
in patients with Goodpasture syndrome, and smokers are at higher risk of developing idiopathic pulmonary fibrosis
(IPF) and rheumatoid arthritis (RA)-associated ILD.
• It is important to recognize and continue to investigate the role of cigarette smoke in the pathogenesis and clinical
course of these diverse diffuse parenchymal lung diseases.
• Although relatively uncommon, these diseases are a significant health burden and frequently affect young adults in
their most productive years.

Cigarette smoke is a complex mixture of more than from developing some other ILDs such as hypersen-
4000 chemicals, many of which exert toxic effects on sitivity pneumonitis (HP).11 We have described a clas-
cellular function. In addition to chronic obstructive sification scheme (Box 3.1) outlining these subgroups
pulmonary disease (COPD) and cancer, cigarette smok- and their relationship to smoking.12 This classification
ers may develop diffuse parenchymal lung diseases illustrates the highly complex effects of smoking on
(Box 3.1).1–4 These diffuse parenchymal lung diseases the lung parenchyma.
are referred to as “smoking-related interstitial lung dis- The Group 1 diseases (Box 3.1) include the three
eases,” a term that recognizes the suspected causal asso- diffuse lung diseases classically regarded as smoking-
ciation with cigarette smoking. Novel insights regarding related ILDs. This designation is supported by several
the relationship between smoking and interstitial lung lines of clinical, epidemiologic, and investigative
disease (ILD) are highlighted in this updated chapter. evidence showing a direct role for cigarette smoking
as witnessed in the temporal relationship to disease
onset and progression, resolution on smoking cessa-
SMOKING AND INTERSTITIAL LUNG tion, and recurrence or progression on resumption
DISEASE of smoking.13–17 Several case series have reported a
Cigarette smoking is now widely accepted as the pri- history of smoking in the overwhelming majority of
mary cause of certain ILDs, namely respiratory bronchi- Group 1 patients, with the prevalence being highest
olitis–interstitial lung disease (RB-ILD), desquamative in RB-ILD4,18 and PLCH,2,5,19,20 and less common in
interstitial pneumonia (DIP), and pulmonary Langer- DIP (Table 3.1).4,21,22 The reported coexistence of
hans cell histiocytosis (PLCH).1–6 Cigarette smoking all three lesions in the same patient,13 the potential
is also a risk factor for the development of idiopathic for disease remission with smoking cessation,4 the
pulmonary fibrosis (IPF)7 and rheumatoid arthri- recurrence of disease in transplanted lungs,23,24 and
tis (RA)–associated ILD,8,9 and has been reported to the description of analogous lesions in mice exposed
cause some cases of acute eosinophilic pneumonia to high doses of cigarette smoke15 all provide sup-
(AEP)10 and pulmonary hemorrhage syndromes. Para- port to the designation of RB-ILD, DIP, and PLCH as
doxically, cigarette smoking may confer protection smoking-induced ILDs.

39
40 Interstitial Lung Disease

For diseases allocated to Group 2 (Box 3.1) the asso- of recent studies have implicated recent-onset expo-
ciation with cigarette smoking is less robust than for sure to cigarette smoke as a principal inducer of this
Group 1 diseases. Cigarette smoking, particularly dur- disease in some patients diagnosed to have this dis-
ing the relatively early phase after initiation of smok- order.26,27,29–32 In addition, increase in the number of
ing, seems to be an important precipitating factor in cigarettes smoked per day in chronic smokers has been
some but not in all cases of Group 2 diseases. The described to induce AEP.32,33 Of particular interest is
most relevant conditions in this category include AEP the response of certain subjects with resolved AEP to a
and certain pulmonary hemorrhage syndromes.10,25–28 rechallenge with cigarette smoke exposure that triggers
AEP deserves particular attention because a number peripheral eosinophilia and other associated patho-
physiologic abnormalities, suggesting that exposure to
BOX 3.1 cigarette smoke to induce certain responses is relevant
Proposed Classification of Smoking-Related to the development of acute diffuse lung disease in sus-
Interstitial lung Diseases (ILDs) ceptible hosts.27
Diseases included in Group 3 (Box 3.1) are chronic
Group 1—Chronic ILDs that are very likely caused by diffuse lung diseases that are statistically more likely
cigarette smoking4–6,13,21,50 to develop in cigarette smokers.34,35 For instance, ciga-
Respiratory bronchiolitis–associated ILD
rette smoking is known to increase the relative risk of
Desquamative interstitial pneumonia
RA-associated ILD, possibly by triggering RA-specific
Adult pulmonary Langerhans cell histiocytosis
Group 2—Acute ILDs that may be precipitated by ciga- immune reactions to citrullinated proteins.9,34,36 Simi-
rette smoking25,27–29,160 larly, smokers have a higher risk of developing IPF than
Acute eosinophilic pneumonia nonsmokers.35 The precise significance of these obser-
Pulmonary hemorrhage syndromes vations has been a topic of substantial debate, but there
Group 3—ILDs that are statistically more prevalent in is limited evidence that smoking itself is directly fibro-
smokers7,8,34–36 genic to the lung.37,38 It is not appropriate to consider
Idiopathic pulmonary fibrosis smoking as an inducer of these diseases, but rather a
Rheumatoid arthritis–associated ILD disease modifier or potentially a cofactor that facilitates
Group 4—ILDs that are less prevalent in
the development of profibrotic responses that lead to
smokers39–42,159
these diffuse fibrotic lung diseases.
Hypersensitivity pneumonitis
Sarcoidosis The fourth and final group consists of diseases
that are less prevalent in smokers compared with

TABLE 3.1
Key Characteristics of Group 1 Chronic Smoking-Related Diffuse Lung Diseases
RB-ILD DIP PLCH
Association with 95% 60–90% 95%
cigarette smoking
Clinical features Chronic cough and dyspnea, Chronic cough and dyspnea, Chronic cough and dyspnea
inspiratory crackles inspiratory crackles Pneumothorax in 15%
High-resolution CT Centrilobular nodules and Ground-glass and reticular Peribronchiolar nodules,
findings ground-glass opacities opacities cavitated nodules, and
cysts with relative sparing
of lung bases
Key histologic Pigment-laden macrophages in Diffuse alveolar filling with Bronchiolocentric nodules,
findings the respiratory bronchioles, and pigment-laden macrophages stellate lesions, CD1a-posi-
alveolar ducts tive Langerhans cells
Response to Modest, variable Modest, variable Modest, variable
corticosteroids

DIP, desquamative interstitial pneumonia; PLCH, pulmonary Langerhans cell histiocytosis; RB-ILD, respiratory bronchiolitis–associated
interstitial lung disease.
Another random document with
no related content on Scribd:
Uakukalailalo, who next? Uakukalailalo, owai mai?
Uahaihaikaka, who next? Uahaihaikaka, owai mai?
Uanahaekaie, who next? Uanahaekaie, owai mai?
Oiukamaewa, who next? Oiukamaewa, owai mai?
Okioikekahuna, who next? Okioikekahuna, owai mai?
Okahikuokamoku, who next? Okahikuokamoku, owai mai?
Keawenuiaumi, who next? Keawenuiaumi, owai mai?

After Kuapakaa had called out Pau ke kehea ana a Kuapakaa i


the names of the men who sat na kanaka pakahi, kahea hou
singly, he then called out those keia i na kanaka palua o ka waa,
who sat two 27 in a seat: ma na inoa:

Nanaimua, Nanaihope, who


next? NA KANAKA PALUA O KA
Neneimua, Neneihope, who WAA.
next?
Kahaneeaku, Kahaneemai, who Nanaimua, Nanaihope, owai
next? mai?
Ku,—Ka, who next? Neneimua, Neneihope, owai
Kapalikua, Kapalialo, who next? mai?
Kapohina, Kapoae, who next? Kahaneeaku, Kahaneemai, owai
Kaukaiwa, Lamakani, who next? mai?
Puupuukoa, Kainei, who next? Ku,—Ka, owai mai?
Koaloa, Koapoko, who next? Kapalikua, Kapalialo, owai mai?
Hulihana, Hulilawa, who next? Kapohina, Kapoae, owai mai?
Pulale, Makaukau, who next? Kaukaiwa, Lamakani, owai mai?
Kuia, Lou, who next? Puupuukoa, Kainei, owai mai?
Hookeleihilo, Hookeleipuna, 28 Koaloa, Koapoko, owai mai?
who next? Hulihana, Hulilawa, owai mai?
Pulale, Makaukau, owai mai?
O Kuia, o Lou, owai mai?
Hookeleihilo, Hookeleipuna,
owai mai?

While Kuapakaa was calling the Ia Kuapakaa e kahea ana i na


names of the men, the double inoa, ia manawa i nalowale ai ka
canoe of the king, waa o ke ’lii o Keawenuiaumi,
Keawenuiaumi, gradually drew mai ko laua mau maka aku. A
away from their sight. When the ike laua, ua koliuliu puaiki, na
double canoe was away off, so waa o Keawenuiaumi, i aku la o
that it appeared but a mere Pakaa i ke keiki, ia Kuapakaa:
speck, Pakaa said to the boy: “Wehe ia ka ipumakani a
“Uncover the wind calabash, Laamaomao.” A wehe ae la o
Laamaomao.” Kuapakaa then Kuapakaa i ka ipumakani, ia wa
uncovered the wind calabash huai ka ino, aole o kana mai,
and the storm in all its fury came loaa mai la na waa mua i ka lae
up. The front canoes were o Kalaau, ko na ’lii a me na
caught by the waves and wind kanaka. Ike na waa nui i ka
from the Kalaau point; being make o na waa liilii, kii aku
those that contained the chiefs hoolana, paupu i ka make.
and the men. When the large Kupikipikio ka ale, ma o a ma o,
canoes saw that the small hele mai la ka makani a loaa na
canoes were swamped, the large waa o Keawenuiaumi, make iho
ones went to their rescue, but la. Olelo aku o
they too were swamped. The [109]Keawenuiaumi i ke kahuna,
waves became larger and larger ke kilo, na hookele: “Kupanaha,
and they beat from all sides. The he mea kau a hala ae ka olelo a
wind and the storm swept along ke keiki, ka olelo ana mai nei no
until the canoe of Keawenuiaumi a ke keiki, he la ino keia, he
was met and it too was malie wale no ia oukou. A laa ka
swamped. Keawenuiaumi then make o kakou, nui kuu ninau
said to the priest, the prophet ana ia oukou, no ko oukou ike,
and the sailing masters: “How he hoole ka oukou.”
strange this is! The boy’s every
word has [108]come true. When
the boy said this was a stormy
day, you all contradicted him,
saying this was a pleasant day;
but here we are nigh unto death.
I questioned you several times
about the matter, to make sure
that you were right; but you all
denied it.”

CHAPTER III. MOKUNA III.


The Swamping of the Canoes.— Ka Make ana o na Waa.—Hoi
They Return and Land on Lakou a Pae ma Molokai.—
Molokai.—The King Is Given Dry Haawiia ke Alii ke Kapa a me ka
Kapa and Malo, as Also Awa and Malo Maloo, pela nohoi me ka
Food.—Delayed by the Storm, Awa a me ka Ai.—No ka Noho
the Party Is Provided With ana o ka Huakai i ka Ino, ua
Food.—After Four Months, Haawi ia me ka Ai.—Mahope mai o
They Prepare to Embark. na Mahina Eha, Liuliu Lakou e
Holo.

The double canoe of Poho iho la na waa o


Keawenuiaumi was swamped as Keawenuiaumi, aole kekahi waa
well as all the others, not one i koe, nui ke koekoe, nui na mea
was saved. The people suffered i lilo aku, ka ai, ka ia, ke kapa,
a terrible cold and many of their na mea a pau loa. Uwe iho la ke
things were lost; the food, the ’lii i ke anuanu a me ke koekoe,
fish and meat, their apparel and a olelo aku la: “Oia kuu mea i imi
everything else. At this, the king ai i kuu kauwa ia Pakaa, o ko
wept in his agony and suffered oukou hemahema, ike ole,
severely from cold, he then said: pololei ole ke olelo. He mau
“This is the very reason why I am papakole pulu ole keia i ke kai
in search of my servant Pakaa, ina o Pakaa ka hookele, ia
because you are not equal to the oukou iho nei pulu.”
occasion; you are without
knowledge and do not know how
to tell the future. My buttocks
were never wet when Pakaa was
my sailing master; but since I
have taken you, they have
become wet.”

When Pakaa saw that the wind Ike aku la o Pakaa i ka makani,
and the storm was in its fury, he a me ka ino launa ole, i aku la ia
said to the boy: “Cover up the i ke keiki: “Poia iho ke poi o ka
wind calabash, for your master ipu, o make auanei ko haku, eia
may perish, as he is indeed la ua anuanu.” Popoi iho la o
cold.” Kuapakaa then placed the Kuapakaa i ke poi o
cover on the calabash, Laamaomao, a hikiwawe iho la
Laamaomao, and the calm came ka malie ana, a pakele ae la na
immediately and the canoes of waa o ke ’lii.
the king were saved.

After the canoes had been Ma keia pakele ana o ke ’lii,


righted, the king gave his orders olelo aku la ia i na waa a pau: “E
to all the canoes, saying: “Let us hoi kakou, malama ke lana ala
return, perchance the boy’s no ka waa o ke keiki, i olelo mai
canoe is still floating where we ia kakou e pae, ae aku kakou e
left it. Should he invite us to land pae, alaila pae kakou.”
we must obey.”
After giving his orders the Pau ka olelo ana a ke ’lii, hoe
several canoes turned about and kela waa keia waa, aia ka pono
all returned, without maintaining o ka hiki i kahi o Kuapakaa e
their order, for each was anxious lana ana. Ma keia hoe ana, oi
to get to the place where aku la ka holo o ko
Kuapakaa was floating. In this Keawenuiaumi waa, i ko na waa
return, the canoe of e ae a hoea aku la ia i ko
Keawenuiaumi being the Kuapakaa wahi e lana ana, emi
swiftest, was the first to arrive at hope mai la na waa a pau loa i
the place where Kuapakaa was hope.
waiting, while the others were
strung out behind.

When Kuapakaa saw the king’s Ike aku la o Kuapakaa i ko ke ’lii


canoe, he said to Pakaa: “Here waa, olelo aku la ia ia Pakaa:
comes the double canoe of my “Eia na waa o kuu haku o
master, Keawenuiaumi.” Pakaa Keawenuiaumi.” I aku o Pakaa i
said to the boy: “When your ke keiki: “I hiki mai ko haku, a ae
master arrives and should show i ka pae i anei, alaila, e olelo aku
a willingness to land, say to him oe, o kaua mua a kahi a kaua e
that you wish to go in ahead a hoolana ai, alaila, kahea mai. E
little ways and wait for him, for olelo aku oe, he kekee ke awa, e
the passage way is crooked.” By pae ai.” Ma keia olelo a Pakaa,
this Pakaa was anxious to keep he olelo akamai loa, manao o
the canoe of Keawenuiaumi Pakaa, o kaa ka waa o ke ’lii
behind them, for his men being mamua, pae e i loko o ke awa,
stronger, they would be able to no ka mea, he ikaika na hoewaa
get to the landing first and in that o Keawenuiaumi. Ma ia mea
way Pakaa would be recognized, noonoo o Pakaa i mea e hiki ai
so Pakaa thought out a way to laua mamua, a ike ole ia kona
get out of the difficulty, and made ano, i nalowale, nolaila, kona
believe that the way in was kuhikuhi lalau ana, o loaa ia.
crooked. As the canoe of
Keawenuiaumi was drawing Alaila, paha hou o Kuapakaa,
near, Kuapakaa again chanted, penei:
saying:

Gently! Gently! Kiauau! Kiauau!


Comes the wind, the rain; the Makani ka ua, po ka moku,
isle is in darkness, Nihinihi ka haku, [111]
The master is on the edge of Kaa ka ua, kaa ka waa,
disaster. [110] Ehuehu kai lele ka moi,
The rain drove, the canoe rolled, Hee loko ua ai ka ale,
The sea is raging, the moi leap. Lele na ukana,
The inwards are retreating, the Hoaa i ke aloha o ke keiki,
waves are being fed, Uwe i ke aloha o ka wahine,
The burden is cast away. Noho inoino kulanalana,
They look about in doubt for love Hae ka ilio i ke kai,
of the children, Nanahu i ka nuku o ka waa,
They weep for the love of the Hookoo ka pili mua,
wife. Hele ka pili hope,
The seat is unsafe, insecure. Kai ka pili a ke kahuna,
The dog barks at the sea, Kuouou, haalulu,
It bites at the prow of the canoe. Pahili, haukeke,
The old companion is become Huhuluwi na hulu i ka maha,
strained, E na holo moana hookuli,
The new companion is become Akahi la anuanu e ke ’lii.
separated, E Keawenuiaumi, e pae.
The comradeship of the priest is
also parted,
He goes alone, he shudders,
He twists, he shivers,
The hairs on the temple are wet,
Ye stubborn sailors of the ocean,
’Tis the first cold day for the king.
Say, Keawenuiaumi, come
ashore.

Keawenuiaumi made reply: “Yes, I mai o Keawenuiaumi: “Ae, e


I will come ashore for your very pae, he mea no kau a hala ko
words have come true. I was olelo i i mai ai; ua ae no au e
willing to land, but these fellows pae, o ke akamai hoi o lakou nei,
were so learned. I thought they kai no he ike io, aole ka!” I aku o
were indeed learned, but I have Kuapakaa: “Ike la i ka make, e
found that they are not.” pae i ke awa o ke keiki.” Ae mai
Kuapakaa said: “There, you ke ’lii: “Ae, e pae.” “Auhea oe e
have faced disaster. Come ke ’lii, e hoolohe mai oe; o maua
ashore at the boy’s landing.” The ke holo e, a kahi e ani mai ai na
king then expressed his lima, alaila, oukou holo ae, no ka
willingness to land. Kuapakaa mea, he kekee ke awa e pae
then said to the king: “Say, listen aku ai o uka, ua hala no hoi ka
to me; we will go in first and wa pono e pae ai. No ka mea,
when I beckon to you, you may ina oukou i ae mua e pae, alaila,
come, because the passage way o ka wa hohonu ole ia o ke kai,
is crooked, and furthermore the aole e nalo ke akoakoa. I keia
proper time for making a landing wa, ua nalowale na pukoakoa no
is past. Had you consented to ka hohonu o ke kai, nolaila, hu
make land at my first invitation, hewa kakou ke holo pu.” Ma keia
we would have had no trouble; olelo a Kuapakaa, ae mai o
for at that time the tide was low Keawenuiaumi: “Ae, ua pono ia.”
and the coral exposed; but now
the tide is high, so that the coral
is covered deep, and we will
miss our bearings if we go in
together.” To this, Keawenuiaumi
gave his consent, saying: “That
is well.”
Pakaa and his son therefore Holo mua aku la o Pakaa ma
entered the passageway first, mamua, a kahi e lana ai, alaila
and when they stopped they kahea mai i ko ke ’lii mau waa, a
beckoned to the king’s canoe as me na waa e ae. Pela no ka holo
well as the others to come in. ana, i o ianei, e hookekee ai, a
This zigzag was continued until kokoke loa e pae i uka olelo aku
they were almost in, when Pakaa o Pakaa i ke keiki: “E, e hoe
said to the boy: “Say, let us kaua, e hoe oe a ikaika loa, i pae
paddle in; you must exert all your kaua.” Ia laua i hoe ai, pae e aku
strength, that we may land la ko laua waa i uka, lehei aku la
before the others.” With this the o Pakaa mai ka waa aku a
two worked with all their might holokiki aku la a komo i ka hale
and made land before the aipuupuu, oia ka hale a Pakaa i
others. Pakaa then jumped manao ai e nalo, no ka mea, he
ashore and ran into the house hale komo ole ia e ke ’lii. Ma
reserved for the preparation of keia lele ana o Pakaa, ua ike
food, thinking that in this house mai o Lapakahoe, o Pakaa no; o
he would be safe, for such kona kumu i manao ai oia, o ka
houses were never entered by hapeepee o ka hele, e onaha ai
kings. When Pakaa jumped from na wawae, aole nae i hooiaio loa
the canoe, Lapakahoe saw and no ka manao, aia no o Pakaa i
thought he recognized Pakaa by Kaula kahi i noho ai.
the limp he made while running,
for his legs had been injured; but
he was not certain, believing that
Pakaa was in Kaula.

Late that afternoon, all the Ahiahi iho la, pau loa mai la na
canoes made land, including the waa i ka pae, a me ko
canoe of Keawenuiaumi, who Keawenuiaumi, eia nae, o
still sat on the platform and had Keawenuiaumi, i luna no ia o ka
not come ashore, for the reason pola o na waa kahi i kau ai, aole
that he did not have any clothes, i lele i uka, no ka mea, aohe
and no loin cloth, all having been kapa, aohe malo, ua pau loa i ka
wet and the spare ones had all pulu, a ua pau loa i ka lilo i ke
been lost at sea. When kai. Ike aku la o Kuapakaa i
Kuapakaa saw his master sitting kona haku i ka noho wale mai i
there naked on the canoe, he luna o na waa, hoi aku la ia a
returned to the house and told olelo i kona makuakane ia
his father of what he had seen. Pakaa. A lohe o Pakaa, unuhi
When Pakaa heard this, he took mai la ia i ka malo a haawi aku
out a loin cloth and gave it to la ia Kuapakaa, [113]a olelo aku
Kuapakaa, saying to the boy: la: “E lawe oe i keia malo a
“You take [112]this loin cloth and haawi aku i ko haku, a o ka malo
give it to your master, and the i pulu, o ia kau e lawe mai, no ka
loin cloth that is wet, you bring it mea, o kona malo nau e hume,
here, for you are privileged to pela hoi kou malo, nana e
wear his loin cloth and he hume.”
yours.” 29

Kuapakaa therefore took up the Lawe aku la o Kuapakaa i ka


loin cloth and returned to malo a hiki i mua o
Keawenuiaumi. When he came Keawenuiaumi: “Eia kuu wahi
to the king’s presence he said: malo nou, o ko malo pulu e
“Here is my loin cloth, you can haawi mai oe ia’u.” Lalau mai la
use it and let me take your wet o Keawenuiaumi i ka malo a
one.” Keawenuiaumi reached out nana iho la, ua like loa me kona
for the loin cloth and looked at it, malo i ko laua wa e noho ana
and saw that it looked like his me Pakaa; i mai la o
own, the kind he used to wear Keawenuiaumi: “E, ua like loa no
when Pakaa had charge. At keia malo me ko’u malo.” I aku o
seeing this, Keawenuiaumi said: Kuapakaa: “No’u no keia malo; o
“Say, this loin cloth looks just like oe hoi na e ke ’lii, nolaila, haawi
my own.” Kuapakaa replied: aku la au nou ia.” Hoi mai la o
“This is my own loin cloth, but Kuapakaa me ka malo pulu a
you being the king, I give it to mua o Pakaa, i mai la o Pakaa:
you.” Kuapakaa then took the “Kau ia ae ka malo o ko haku ma
wet one and returned to Pakaa, ka puka o ka hale, i ole e komo
who said to him: “Hang up your mai na kanaka i loko nei. O oe
master’s loin cloth over the door ka mea komo i keia hale, a me
way, so that the people will not ka puka i waho, no ka mea, ua
try to enter this house. You can laa oe i ke kapa a me ka malo o
enter it and can go out, because ko haku, i hele mai na aipuupuu i
all the sacred things belonging to ai, nau e haawi aku maloko nei,
your master are free to you. ma waho mai no lakou.” He
When the king’s stewards come hana maalea keia a Pakaa.
for food you can hand it to them
from the inside of this house,
while they stand outside.” This
was cunning of Pakaa. 30

When Kuapakaa looked and saw Nana aku la o Kuapakaa, o ka


that Keawenuiaumi was sitting noho wale mai o Keawenuiaumi
without any covering, he took aohe kapa, aloha iho la ia, olelo
pity on him and so told Pakaa aku la ia Pakaa; a lohe o Pakaa,
about it. When Pakaa heard this unuhi mai la ia i ke kapa, i loko o
he took out a kapa from the wind ka ipu o Laamaomao, a haawi
calabash, Laamaomao and aku la ia Kuapakaa. Olelo aku la:
handed it to Kuapakaa, saying: “E lawe oe i ke kapa a haawi aku
“You take this and give it to your i ko haku, i olelo mai ko haku, ua
master. If he should say that it like me kona kapa, e olelo aku
looks like his, you tell him, that oe, o kou kapa no keia a kou
this is your own kapa made by makuahine i kuku ai nou.” O ka
your mother.” The name given to inoa o ke kapa, o ouholowai o
such kapas was “ouholowai of Laa; ua aala loa, no ka mea, ua
Laa.” 31 They were very sweet, hooluuia i na nahelehele aala o
having been scented with the Laa a me Puna, oia ka olapa, ke
fragrant shrubs and vines of Laa kupaoa, ka mokihana, ke apiipii,
and Puna, called the olapa, the a me na mea e ae.
kupaoa, the mokihana, the apiipii
and others.

When Kuapakaa came to the A hiki aku la o Kuapakaa i mua o


presence of the king with the Keawenuiaumi me ke kapa,
kapa and handed it to haawi aku la, lalau mai la o
Keawenuiaumi, Keawenuiaumi Keawenuiaumi i ke kapa, a
took it and spread it out. As he kuehuehu ae la, po i ke ala o ka
did this he caught the sweet olapa, honi iho la i ke ala. Alaila,
scent of the olapa. He then ninau mai la i ke keiki: “Nohea
inquired of the boy: “Where did keia kapa i loaa ai ia oe?” “No
you get this kapa?” The boy Molokai nei no,” pela aku ke
replied: “It belongs here in keiki. I mai o Keawenuiaumi:
Molokai.” Keawenuiaumi said: “Aohe kapa o na wahi e ae e like
“There are no kapas in other me ko Hawaii, aole no hoi i laha i
places like those of Hawaii; and na ’lii e ae, ia’u wale no; me he
they are not common with other mea ala o kuu kapa no keia, a
chiefs. I am the only one who eia no paha i anei o Pakaa?” “Na
possesses such things. I believe ko’u makuahine no i kuku i keia
this is my kapa. It must be that kapa no’u, no ka mea, he ’lii ko’u
Pakaa is here.” “It was my makuahine no Molokai, a he
mother that made this kapa for kapa aala no hoi ko keia aina, ua
my own use, for my mother is a hooluu ia i na mea aala he nui
chiefess of Molokai and kapas loa, a ua malama ia no’u. O ka
are scented on this island, and it inoa o ko’u kapa, o wailau, oia
has been kept for my own use. koonei kapa aala loa, e like me
The name given my kapa is ko oukou he ouholowai o Laa.
wailau. 32 That is the best and Ua like na aala.” Pau ae la ko ke
most fragrant kapa in this place, ’lii manao haohao.
like what you call the ouholowai
of Laa; they smell the same.”
This satisfied the king.
That evening the chiefs came A ahiahi iho la, akoakoa ae la na
together with their men and as ’lii me ko lakou mau kanaka, a
they were sitting quite close to kahi hookahi; olelo aku o
the king, the king said: “If Pakaa Keawenuiaumi: “Ina nei la o
was here, of an evening like this, Pakaa, penei keia ahiahi la, o ka
he would have my awa ready apu awa mai la no, o na hinalea
with two fresh hinalea. 33 I would ola elua. Inu iho la a ona, ooki
drink the awa and as its effects iho la ka ona o ka awa, uwi kela
come over me, I would feel like a me he koko aha la, a ao ka po;
newly made net, nice and snug, aloha no hoi o Pakaa.”
all night. How I do miss Pakaa.”

When Kuapakaa heard this he Lohe o Kuapakaa i keia olelo a


returned to his father, Pakaa, ke ’lii, hoi aku la ia olelo ia
and said: “My [114]master is in Pakaa: “Ua ono kuu [115]haku i
want of some awa, and he has ka awa, a olelo mai nei he aloha
expressed his affection for you ia oe no ia mau mea i kou wa e
and showed that he still noho ana me ia.” A lohe o
remembers you.” When Pakaa Pakaa, unuhi mai la ia i ka apu, i
heard this, he took down the awa ke kanoa, i ka mauu, i ka puawa,
cup, the awa dish, the grass me na mana awa elua i mama
used for straining awa, the piece mua ia: “Lawe oe i keia a ko
of awa and two portions of awa haku, hoike aku, a i olelo mai
already prepared and said to the nau e mama, ae aku no. Alaila,
boy: “You take these to your huli ae oe a ma kahi poeleele,
master and show them to him. If waiho oe i ka puawa okoa, lalau
he should ask you to prepare the iho oe i na mana i wali, a hoka
awa for him, give your consent. iho i loko o ke kanoa, alaila, e
Then you turn to one side where mahalo kela i ko hikiwawe, no ke
it is dark, leave the piece that is mea, pela wau i ko’u wa e noho
not prepared, take up the ana me ia. A pau ka awa i ka
portions that are ready, strain hoka, haawi aku oe i ko haku,
them into the cup. He will alaila, holo mama oe i na hinalea
compliment you for being very elua a kaua i hooholo ai i ka
quick, for I was ever ready with hapunapuna, lawe mai oe i pupu
these things when I was with no ka awa o ko haku, i pau ka
him. After you have strained the mulea awa o ka waha o ko
awa into the cup, hand the cup haku.”
to your master, then run as fast
as you can to the pool where we
keep the hinalea and catch two
for your master, for he would
want the fish to take away the
bitter taste of the awa from his
mouth.”

When Kuapakaa came to the A hiki o Kuapakaa i mua o


presence of Keawenuiaumi, he Keawenuiaumi, olelo aku la: “Eia
said: “Here is my awa for you.” kuu wahi awa nou.” Nana mai la
The king looked and saw that it ke ’lii a ike he puawa nui, olelo
was quite a large piece, so he mai la: “Nau no e mama.” Huli
said: “You had better prepare it ae la o Kuapakaa ma kahi
for me.” Kuapakaa then turned poeleele a hoka iho la i na mana
into a dark corner, took the i wali mua, haawi aku la i ke ’lii,
portions already prepared, a holo aku la i na hinalea elua, a
strained the same and handed hoi mai la i mua o
the cup to the king. The boy then Keawenuiaumi.
ran for the fish, the two hinalea,
and shortly after he returned with
them to the king.

Because of these things No keia mau hana a ke keiki,


performed by the boy, mahalo iho la o Keawenuiaumi i
Keawenuiaumi complimented ka eleu, me he kanaka makua
him for being quick and for ala, ua noho me na ’lii a maa ka
carrying himself like a person makaukau. Inu iho la ke ’lii a
who has always lived with kings, ona, moe iho la, hui ae la ka ona
and for conducting himself so o ka awa me ka maluhiluhi o ke
well. The king then drank up the kai, o ka moe ka hana.
awa and as the effects of it stole
over him, combined with the
weariness of a hard and eventful
day, he fell into a deep sleep.

Upon seeing this, Kuapakaa Nolaila, manao o Kuapakaa, e


decided to uncover the wind huai i ka ipu makani ana ia
calabash, Laamaomao, and to Laamaomao, i pa ka makani,
keep it uncovered, so that the mau no ka ino, noho no ke ’lii me
winds would continue to blow ia. Wehe ae la o Kuapakaa i ke
and the storm hold for days; and poi o Laamaomao, a pa iho la ka
in this way keep the king with ino i kela la i keia la, ma keia ino
him. So Kuapakaa uncovered i lohi ai ka holo o Keawenuiaumi.
Laamaomao, and the storm kept Pela ko lakou kali ana i ka malie,
up day after day; and by it the a hala hookahi malama, pau ae
expedition for the search was la ko lakou koena ai, o ka hele
postponed. Because of this ana mai Hawaii mai. Ia wa, hele
storm Keawenuiaumi and his mai na ’lii o Hawaii ia
men were forced to wait for the Keawenuiaumi, hai mai la i ko
abating of the storm until one lakou pilikia nui o ka pololi, a
month went by, when their food lohe o Keawenuiaumi, i ka lakou
which had been brought from olelo. Hoouna aku la o
Hawaii was exhausted. At this, Keawenuiaumi i ke kanaka, e
the chiefs went before ninau aku i ke keiki he ai paha
Keawenuiaumi and told him of kana, aole paha. Aka, ina he ai,
their trouble, that they had run e olelo aku oe he pilikia ko
out of food. When kakou.
Keawenuiaumi heard this he
sent a man to go and ask of the
boy, if he had any food. Said the
king: “If he has any food, tell him
that we are without any.”

When the man came before A hiki aku la ke kanaka i mua o


Kuapakaa he told him what Kuapakaa, hai aku la i na olelo a
Keawenuiaumi had said to him. Keawenuiaumi, a lohe o
When Kuapakaa heard this, he Kuapakaa, olelo mai la: “He ai
said: “There is food; but you no, eia nae, e hoi oe a hai aku i
must go back to the king and tell ke ’lii ia Keawenuiaumi, he ai no,
him the food is up in the uplands. aia i uka, e olelo aku oe, eono
Tell him there are six chiefs here alii, eono kipoipoi. Eia hoi, i kii
and I have six small patches. oukou i ka ai, mai ohi oukou i ka
Furthermore, if you should go for mea nui wale no, a haalele i ka
food, don’t take the big potatoes mea liilii, ina oukou e hana pela,
only and leave the small ones; aole oukou e kii hou i ka ai.” Hoi
for if you do so you will not get aku la ka elele a mua o
another chance to go up there Keawenuiaumi, olelo aku la i ka
for food.” With this the olelo a ke keiki, a lohe o
messenger returned to the Keawenuiaumi, kena ae la ia i
presence of Keawenuiaumi and na kanaka a me na ’lii e pii i ka
reported to him of what the boy ai.
had said. When Keawenuiaumi
heard this he ordered his men
and the chiefs to go up for food.

When they came to the uplands, A hiki lakou i uka, nana aku la
where the potatoes were lakou i na mala uala eono, nui
growing they saw that there were launa ole, a loihi no hoi ke nana
six large patches, each of very aku. Olelo ae la kekahi i kekahi:
great extent, and were so long “Kupanaha, olelo mai nei hoi ua
that the other ends could not be [117]keiki nei, he mau wahi
seen. The men then said to kipoipoi wale no, eia ka hoi, he
themselves: “How wonderful! mau mala nunui.” Koki iho la
The [116]boy said there were six lakou, a nui ka ai, hoiliili iho la, e
small patches, but here there are like me ka olelo a ke keiki mai ka
six very large patches.” The men mea nui a ka mea liilii, a hoi aku
then began to dig up the la a hiki i kai, hoa ka umu, a moa
potatoes, and after they had dug ae la, ai iho la lakou. Hele mai la
up enough, they collected the o Kuapakaa a hiki olelo mai la ia
potatoes and in obedience to the lakou: “E auhea oukou o ka uala
boy’s order, they took the large nui o ka oukou ia, O ka mea liilii
ones as well as the small ones o ka’u ia.” “Kahaha, aole peia, i
and returned with the potatoes to uala nui no kekahi au, no ka
the beach, lighted the ovens, mea, nau ka ai.” “Aole,” pela aku
and after the potatoes were o Kuapakaa, “o ka ai nui na
cooked, they sat down and ate oukou no ia, o ka ai liilii o ka’u ia.
their fill. Penei nae oukou e hana ai; e ihi
oukou a pau ka ili o waho, alaila,
kaulai i ka la a maloo.” O ke ano
o keia, he ao maloo.

After this Kuapakaa came to Ninau mai la lakou: “Heaha ke


them and said: “I want you to ano o keia hana au e ke keiki?” I
take the large potatoes for your aku o Kuapakaa: “No ko’u
own use and keep the small manao, e pau ana kela mau
ones for me.” “Why, no, not so; mala uala ia oukou, a e hiki mai
you must have some of the large ana ka manawa ino o keia aina,
ones, too, because the potatoes e kaikoo ai ka moana o kai, a e
are yours.” “No,” said Kuapakaa, loihi ana no hoi ko oukou noho
“you take the large ones and ana i anei, no ka mea, ekolu
save the small ones for me. But I malama ino i koe, o Makalii,
want you to do this: peel the skin Kaelo, Kaulua. I Olana paha
and then set out the potatoes to hookau ka malie, alaila, oukou
dry.” 34 The people then asked holo, nolaila, e pau ana ka’u ai ia
Kuapakaa: “What do you intend oukou; aka, i hoi oukou, aole au
doing with the food, boy?” e wi ana, aole no hoi e pololi, no
Kuapakaa replied: “I am doing ka mea, ua ola au i ka ai liilii a
this, because I know you will eat oukou e hoiliili nei, loaa hoi ko’u
up those potato patches and the o e mahiai aku ai i ai na’u.” O
bad weather of this land keia olelo a Kuapakaa, he olelo
generally comes about this time, maalea, he olelo huna, aole ia o
when the sea will be rough, ke ano maoli. Ua ike no o
which will keep you here for Kuapakaa, e koi ana no o
some time, for there are three Keawenuiaumi, e holo e imi ia
bad months yet to come; Makalii, Pakaa, ke malie, nolaila, ua
Kaelo and Kaulua. 35 In the malama loa na kanaka i ka ai
month of Olana, 36 it is possible liilii, i kela umu keia umu ke
that fine weather will come, then kalua ai lakou.
you people will be able to get
away. By that time my potatoes
will all be consumed by you, but
by doing this, saving and drying
out the small potatoes, I will not
be without food and will not be
hungry, for I shall then live on the
small potatoes which I ask you to
keep for me. With this food I will
be supplied during the time of
planting and care of a new crop.”
This talk by Kuapakaa, although
true, was intended to deceive
them, for Kuapakaa well knew
that Keawenuiaumi was going to
urge that he go along with him in
the search for Pakaa, when good
weather once more prevailed.
The men, in obedience to this
order, faithfully kept all the small
potatoes after every cooking day.
When Keawenuiaumi left Hawaii Eia nae, ia Keawenuiaumi ma i
on this expedition, he left word holo mai ai mai Hawaii mai, olelo
with the chiefs and the common aku ia i na ’lii o hope a me na
people that he would take up a makaainana, hookahi mahina e
month in the search for Pakaa. holo mai ai e imi ia Pakaa, aka,
He was, however, mistaken in ua hala na mahina eha ia lakou
this, for he was in Molokai for ma Molokai, o ka noho ana. Ma
four months. In this prolonged keia noho loihi ana o
absence the people of Hawaii Keawenuiaumi ma, kanikau na
began to mourn for their king, makaainana o Hawaii ia ia, e
believing that he was dead. After manao ana ua make. A hala na
staying in Molokai for four mahina eha i ka noho ana ma
months, the followers of Molokai, hu mai la ke aloha o ka
Keawenuiaumi began to think of wahine, ke keiki, a me ka
their wives, children and parents, makua, nolaila, pau ka manao
and there was a general feeling hele e imi ia Pakaa, o ka hoi
amongst them that they abandon wale no i Hawaii ka pono.
the search for Pakaa and return
to Hawaii.

At the expiration of the four A pau na malama ino eha a


months, during which bad Kuapakaa i olelo ai, popoi iho la
weather was to prevail as ia i ka ipu makani o
predicted by Kuapakaa, he Laamaomao, hookau mai la ka
closed the wind calabash of malie. Olelo aku la ia: “Olana
Laamaomao, and good weather keia o ka malama malie, o Welo,
was once more experienced. He o Ikiiki, o ke aho pulu a ka
then said to the people: “This is lawaia, he mau malama malie
Olana, the pleasant month, then wale no keia; nolaila, maloo ole
come Welo and Ikiikii (May and ai ke aho a ka lawaia, no ka holo
June), the period of time when mau i kai.” I aku la ia i na ’lii me
the fisherman’s fish line is na kanaka: “E hoa na waa a
always wet. These months are paa, no ka mea, ua kaohi aku
the pleasant months, and hence wau ia oukou no na malama ino
the fisherman’s line is never dry, eha, a ua malie, e hoi oukou.” A
because they go out fishing lohe na ’lii i keia olelo, makaukau
every day.” He then said to the iho la na waa a paa i ka hoa,
chiefs and men: “Bind the hoolana aku la i loko o ke kai,
lashings of the canoes, for I have me ka paa i ka hekau ia, no ka
kept you for four months mea, he huakai [119]pulale ka ke
because of the bad weather; ’lii. A ahiahi loa, aneane e
now that good weather has aumoe, kahea aku la o
come, you must return home.” Kuapakaa ma ka paha penei:
When the chiefs heard this, they
made ready their canoes,
renewed the lashings, and
pushed the canoes out into the
sea and moored them, for the
expedition [118]of kings is ever
alert. Very late that evening,
when it was almost midnight,
Kuapakaa called out in a chant,
as follows:

Arise! Arise! Arise! E ala! e ala! e ala!


The night is spent, the night is Ua kulu ka po! ua kulu ka po!
spent. Pau ka luhi, ka eha, ka opa,
All tiredness, soreness and Ka maka pouli o na waa la.
weariness have vanished; E ala! e ala! e ala!
Also darkness that prevents the Aia i luna o Hikiliimakaounulau,
sailing of canoes. Ka hoku i ka palena o ka aina.
Arise! Arise! Arise! E ala! e eu! E ala! e eu!
Hikiliimakaounulau 37 is up,
The star at the end of the land.
Arise, make a move! Arise, make
a move!

Upon hearing this, the chiefs Ma keia olelo a ke keiki, olelo ae


said: “How strange! it is not yet la na ’lii: “Kupanaha! Aole hoi i
anywheres near daylight, but the kokoke aku i ke ao, o ka hea
boy is calling us to sail off. This okoa mai nei no ia e holo, eia no
is only in the early evening.” ka i ke ahiahi okoa.”

CHAPTER IV. MOKUNA IV.


Departure from Molokai.—The Ka Haalele ana ia Molokai.—Na
Names of the Six Districts of Inoa o na Moku Hawaii Eono.—
Hawaii.—The King Desires Makemake ke ’Lii e Holo pu o
Kuapakaa to Accompany Him.— Kuapakaa me ia.—Ae ke keiki
The Boy Consents Under Malalo nae o na Kumuaelike.—
Conditions.—They Start Off.— Hoomaka Lakou e Holo.—
Meeting With Adverse, Cold Halawai me na Makani Anu
Winds, the Two Sailing Masters Pahili, Haule na Hookele Elua
Fall Overboard. mai ka Waa.

By this expression used by the O ko na ’lii manao ma keia olelo


chiefs, it was their intention to a ke keiki, e waiho a huli ka ia i
delay their sailing until the ke kau o ka po, alaila holo.
change of the Milky Way, after Nolaila, hoomaka hou o
midnight, when they would make Kuapakaa e kahea ma ka inoa o
the start. Hearing this na moku eono o Hawaii, e pili
expression, Kuapakaa again ana i na ’lii eono o Hawaii:
called out; this time naming the

You might also like