Вы находитесь на странице: 1из 8

Drugs & Aging

https://doi.org/10.1007/s40266-019-00647-y

LEADING ARTICLE

Idiopathic Pulmonary Fibrosis: New and Emerging Treatment Options


Richard J. Hewitt1,2 · Toby M. Maher1,2 

© Springer Nature Switzerland AG 2019

Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and debilitating, scarring lung disease with a worse prognosis than some
cancers. The incidence of IPF is increasing and while current antifibrotic therapies slow disease progression, they do not offer
a cure. The pathobiology of IPF is complex and is driven by aging-associated cellular dysfunction, epithelial injury, and an
aberrant wound-healing response characterised by fibroblast activation and extracellular matrix accumulation (ECM) in the
interstitium. As understanding of the underlying mechanisms has evolved, new targets for pharmacotherapy have emerged.
Novel drugs currently in development for pulmonary fibrosis have diverse molecular properties and mechanisms of action,
as well as different routes of administration. A shared primary goal of these agents is reduction of the profibrotic activity of
fibroblasts and limitation of ECM deposition, which hinders gas exchange and ultimately leads to respiratory failure. This
article provides an overview of some promising new therapeutic options for IPF and considers the challenges for future
drug development.

Key Points  1 Introduction

Idiopathic pulmonary fibrosis (IPF) is a devastating lung Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal
disease associated with aging that causes progressive lung disease of unknown cause characterised by relentless
interstitial scarring, respiratory failure and death. interstitial scarring [1]. The incidence of IPF is estimated
at three to nine cases per 100,000 population per year, and
There are currently only two antifibrotic drugs licensed
increases with age [2, 3]. There is interindividual variability
for the treatment of IPF in the UK. These drugs slow
in the disease trajectory, but ultimately prognosis is poorer
disease progression but do not provide a cure.
than several other types of cancer, with a median survival of
There is a growing portfolio of novel therapies that target 3 years [4, 5]. As normal lung parenchyma is replaced with
diverse pathways in the intricate pathobiology of IPF scar tissue, there is an irreversible deterioration in lung func-
which have been shown to be safe in early-phase clinical tion, which has a devastating impact on the quality of life of
trials. Many of these drugs aim to reduce the profibrotic sufferers who experience debilitating dyspnoea and cough.
activity of fibroblasts and limit extracellular matrix Currently, no pharmacological therapy is available that
deposition. can reverse lung fibrosis or cure IPF. There was a paradigm
shift in treatment strategy following the PANTHER-IPF
clinical trial published in 2012 [6], which demonstrated
that patients treated with a combination of prednisolone,
azathioprine and N-acetylcysteine (NAC) had an increased
risk of hospitalisation and death compared with placebo [6].
This led to a reconsideration of the fundamental pathobiol-
* Toby M. Maher ogy of IPF, a move away from conventional immunosup-
t.maher@imperial.ac.uk pression, and the development of novel antifibrotic agents.
Two orally administered antifibrotic drugs—pirfenidone
1
National Heart and Lung Institute, Imperial College London, and nintedanib—were recommended for the treatment of
Sir Alexander Fleming Building, London SW7 2AZ, UK
IPF in the 2015 ATS/ERS/JRS/ALAT Clinical Practice
2
Interstitial Lung Disease Unit, Royal Brompton Hospital, Guideline on the basis of clinical trials that demonstrate
London SW3 6NP, UK

Vol.:(0123456789)
R. J. Hewitt, T. M. Maher

they slow disease progression and improve survival [7–10]. epithelial vulnerability to injury [13]. It is probable that the
Treatment-associated adverse effects (including nausea, interplay of multiple pathways with inbuilt redundancy is the
dyspepsia and a photosensitive rash with pirfenidone, and reason that many single pathway-targeted drug candidates
nausea and diarrhoea with nintedanib), limit tolerability and have failed in IPF clinical trials.
lead to drug discontinuation in up to 26% of patients [11]. The histological lesion associated with IPF, usual inter-
Although pirfenidone and nintedanib represent a step-change stitial pneumonia (UIP), is characterised by spatial and
in the management of IPF, there remains a significant unmet temporal heterogeneity with normal lung interposed with
need for new therapies for this inevitably progressive and active fibrotic foci and honeycombing in a subpleural distri-
fatal disease. This review aims to highlight promising novel bution [1]. It is important to recognise that the development
agents for the treatment of IPF. of fibrosis in IPF is not only associated with the accumula-
tion of collagen and extracellular matrix but is also defined
by architectural destruction of the lung, with abnormal
2 Idiopathic Pulmonary Fibrosis (IPF) remodelling of distal airspaces, resulting in loss of the gas
Pathobiology exchange surface area. The nature of this fibrotic destruction
raises the challenge of how best to deliver drugs in IPF, and
Understanding of the mechanisms underlying IPF has shapes treatment aspirations. The loss of lung architecture in
improved significantly over the last 20 years, catalysed in IPF makes it unlikely that even highly effective antifibrotic
part by the advent of transcriptomics, which has allowed drugs will restore normal lung structure and function. As
unbiased analysis of human samples to identify novel path- such, the primary treatment intention in IPF is to stabilise
ways [12]. Current evidence supports a pathogenic model lung function and prevent further loss rather than, as would
in which alveolar epithelial injury provokes a dysregulated be desirable, to reverse disease.
wound healing response with fibroblast proliferation and
activation, and excess extracellular matrix deposition in the
interstitium (Fig. 1) [13]. This impedes gas exchange, lead- 3 Emerging Targets and Pharmacotherapy
ing to respiratory failure and ultimately death. for IPF
IPF is a complex disease, with a number of potential envi-
ronmental triggers and abnormalities in multiple pathways 3.1 Autotaxin: GLPG1690
driving the disease [14]. Aging-associated cellular distur-
bances, including mitochondrial dysfunction and senes- Lysophosphatidic acid (LPA), a phospholipid that signals
cence, with genetic variants, also play a significant role in via G protein-coupled receptors to illicit a range of cellular

Injury Galectin- 3 inhibitor - TD139

Cell- based therapies


Alveolar space

• Anti- αVβ6 - BG00011 & GSK3008348


• Anti-CTGF - FG-3019 (Pamrevlumab)
Epithelium • Autotaxin inhibitor - GLPG1690
• LPA1 receptor antagonist - BMS-
986020
• Pentraxin- 2 - PRM151
• PI3K/mTOR inhibition; GSK2126458
Mesenchyme (Omipalisib)
• GPR40 agonist/ GPR84 antagonist –
PB4050
• Anti-IL-13 - Lebrikizumab
Endothelium

Vascular space

Fig. 1  Schematic overview of the complex pathobiology of IPF and the fibrotic cascade, with the common aim of limiting matrix depo-
novel drug candidates. Injury to an aging alveolar epithelium pro- sition. Adapted from Maher [15]. IPF idiopathic pulmonary fibrosis,
motes activation of fibroblasts and differentiation to myofibroblasts, ECM extracellular matrix, CTGF connective tissue growth factor,
which deposit excess ECM in the interstitium, leading to impaired gas LPA lysophosphatidic acid, PI3K/mTOR phosphatidylinositol-3-ki-
exchange. Many of the agents listed target more than one aspect of nase (PI3K)/mammalian target of rapamycin, IL interleukin
IPF: New and Emerging Treatment Options

responses, including proliferation and migration, is increased a significant role in this process [21]. Pentraxin-2 (PTX-2),
in the bronchoalveolar lavage (BAL) fluid of IPF patients also known as serum amyloid P (SAP), is a blood plasma
[16, 17]. In a bleomycin model of fibrosis, LPA, via the pattern recognition receptor synthesised by the liver during
receptor ­LPA1, mediates fibroblast recruitment and increases steady state. The pentraxin family also includes pentraxin-1,
vascular permeability following lung injury [17]. Concentra- more commonly known as C-reactive protein (CRP), and
tions of autotaxin (ATX; ENPP2), the enzyme responsible pentraxin-3. Administration of human SAP inhibits fibro-
for converting lysophospholipids to LPA, were found to be sis in a mouse kidney by opsonising apoptotic cell debris
increased in IPF lung tissue, prompting further investigation and acting as a ligand for the Fcγ receptor, downregulating
as a potential therapeutic target [18]. monocyte and macrophage activity [22]. Administration
A multicentre, phase IIa randomised placebo-controlled of recombinant human SAP reduces lung collagen content
trial (RCT) of GLPG1690 (Galapagos), an orally adminis- in bleomycin challenged and transforming growth factor
tered, selective autotaxin inhibitor was published in 2018 (TGF)-β1 overexpressing mice, and is associated with a
[19]. Twenty-three patients with comparable demographics reduction in alternatively activated (M2) macrophages [23].
were enrolled in the trial to take either 600 mg GLPG1690 Blood plasma levels of SAP are reduced in IPF patients com-
or placebo once daily for 12 weeks. Treatment-emergent pared with controls, and correlate with FVC [24].
adverse events of mild to moderate severity were registered Intravenous recombinant human pentraxin-2, PRM-151,
in the placebo and GLP1690 groups in similar proportions. was assessed in a small, multiple ascending dose, phase I
Two (12%) patients in the GLP1690 group encountered RCT in treatment-naive subjects with IPF [25]. It was well
events directly related to the treatment, but, importantly, no tolerated, with no severe infusion reactions or adverse events
patients suffered acute exacerbations of IPF or died during at any dose (1, 5, or 10 mg/kg); however, there was one
the trial. Measured concentrations of LPA species C18:2 episode of treatment-related hypotension. Pharmacokinetic
were reduced with a once-daily 600 mg dose of GLP1690 studies showed, as expected, that pentraxin-2 led to a dose-
for 12 weeks. The study was not statistically powered to dependent increase in PTX-2 levels.
evaluate efficacy, but spirometry results were encouraging; A phase II, double-blind, multicentre RCT of pentraxin-2
mean forced vital capacity (FVC) remained stable in the administered intravenously (10 mg/kg) every 4 weeks for
GLPG1690 group compared with the placebo group, where 24 weeks has now been conducted [26]. Randomisation
it declined (mean − 70 mls). Parallel phase III trials assess- was stratified according to background therapy, with sub-
ing the safety and efficacy of GLPG1690 with standard med- jects being permitted to take concomitant pirfenidone or
ical therapy are now recruiting (ISABELA1, ClinicalTrials. nintedanib provided a stable dose was established for at
gov identifier: NCT03711162; and ISABELA2, ClinicalTri- least 3 months prior to study entry. A total of 111 subjects
als.gov identifier: NCT03733444). completed the trial, in which a significant difference in least
squares mean change in FVC percentage predicted from
3.2 Lysophosphatidic Acid Receptor: BMS‑986020 baseline to 28 weeks was seen in the pentraxin-2 group
(− 2.5) versus the placebo group (− 4.8); a difference of 2.3
An alternative strategy for targeting the LPA pathway has (90% confidence interval [CI] 1.1–3.5; p = 0.001). The most
been undertaken using a small molecule inhibitor of recep- common adverse event in the pentraxin-2 group was cough,
tor ­LPA1, BMS-986020 (Bristol-Myers Squibb), and a and although there were four infusion-related reactions, none
multicentre, phase II, double-blind RCT of BMS-986020 were considered by the authors as serious. The authors rec-
has been completed [20]. A total of 108 IPF patients com- ognized the inclusion of subjects with possible UIP and lack
pleted the 26-week study, in which they were randomized to of central review of radiology as limitations of the study.
receive placebo or BMS-986020 600 mg once or twice daily.
A slower rate of FVC decline was seen in patients treated 3.4 Connective Tissue Growth Factor: FG‑3019
with BMS-986020 twice daily compared with placebo. (Pamrevlumab)
Furthermore, a dose-related increase in liver enzymes and
three cases of cholecystitis were reported in the treatment Key pathways of fibrogenesis, including fibroblast differen-
arm and were postulated to be the result of off-target BMS- tiation to and activation of myofibroblasts, ECM deposition
986020-induced inhibition of hepatobiliary transporters. and TGF-β expression, are modulated by the matricellular
protein, connective tissue growth factor (CTGF/CCN2) [27].
3.3 Pentraxin‑2: PRM‑151 CTGF does not appear to act via a unique receptor, thus
complicating the therapeutic targeting of this pathway. How-
At the centre of the underlying pathobiology of IPF is an ever, an anti-CTGF human monoclonal antibody that binds
aberrant wound-healing response to alveolar injury. Mono- CTGF directly with high affinity, FG-3019, also known as
cyte-derived alveolar macrophages have been shown to play pamrevlumab (FibroGen), was identified as a candidate in
R. J. Hewitt, T. M. Maher

preclinical models of disease [27]. A tenfold increase in and type 2 alveolar epithelial cells [35, 36]. Mice with a
CTGF messenger RNA (mRNA) transcripts was detected in genetic deletion of galectin-3 (galectin-3−/−) exhibit less
the BAL fluid of IPF patients compared with controls [28]. fibrosis in TGF-β1 and bleomycin murine models [34]. In
In a radiation-induced pulmonary fibrosis mouse model, response to TFG-β1, alveolar epithelial cells from galec-
FG-3019 attenuates or reverses lung fibrosis, with improve- tin-3−/− mice show less epithelial–mesenchymal transition
ments in lung function and survival, depending on the time than wild-type, while galectin-3−/− fibroblasts produce less
of administration in relation to the initial irradiation injury collagen. Targeting galectin-3 with a small molecule inhibi-
[29]. tor, TD-139, instilled directly into the lungs of mice follow-
In a phase IIa, open-label, single-arm, multicentre trial, ing bleomycin, reduced fibrosis [34].
89 subjects received intravenous pamrevlumab at either Three different doses (0.3, 3 and 10 mg) of a dry pow-
15 or 30 mg/kg every 3 weeks for 45 weeks [30]. Subjects der inhaler formulation of the galectin-3 inhibitor TD-139
were allowed to continue prednisolone (≤ 10 mg) and/or (Galecto Biotech) have been tested in a proof-of-concept,
NAC (≤ 1800 mg) provided these had been taken for at multicenter, double-blind, phase IIa RCT in 24 subjects
least 4 months prior to study enrolment. Inclusion criteria [38]. Galectin-3 was assessed in BAL and plasma predos-
included those with a history of disease progression (defined ing and 14 days postdosing. No significant drug-related
radiologically or by an FVC decline of ≥ 10%) in the pre- adverse effects were reported in the abstract presented at
ceding 12 or 18 months, and ≥ 10% to < 50% whole-lung the ATS 2017 International Conference. There was a high
fibrosis quantified by high-resolution computed tomography dose-dependent concentration of TD-139 in BAL pellets,
(HRCT) to allow evaluation of any reversal of lung fibro- suggesting inhaled delivery of the drug to the distal air-
sis, as was suggested in the preclinical studies. The safety space was achieved. TD-139 effectively lowered expression
profile was acceptable; treatment-emergent serious adverse of galectin-3 in flow-sorted BAL macrophages at the 3 and
events occurred in 29% of subjects and none of the deaths 10 mg doses. A phase IIb trial is anticipated to start recruit-
that occurred were attributed to the treatment. An average ing within the next year.
decline in FVC of 140 mls was seen over the 48-week study
period, with 30% of subjects showing a 0.2–14.1% increase 3.6 αvβ6 Integrin: BG00011 (Formerly STX‑100)
in FVC. Interestingly, a subset also showed reduced reticular
fibrosis by quantitative HRCT. Integrins are transmembrane receptors that modulate cell
Following on from this, a phase II, double-blind, placebo- adhesion to the extracellular matrix and adjacent cells,
controlled trial, ‘PRAISE’, randomised 103 IPF patients to which also interact with the cell cytoskeleton to activate
receive 16 doses of pamrevlumab (30 mg/kg) or placebo intracellular signalling [39]. Integrin αvβ6 is strongly
for 48 weeks (ClinicalTrials.gov identifier: NCT03733444). expressed in type I and II alveolar epithelial cells in fibrotic
A late-breaking abstract at the American Thoracic Society areas assessed by immunostaining of lung tissue from indi-
(ATS) 2018 International Conference reported pamrevlumab viduals with IPF, with higher levels of αvβ6 immunostaining
slowed progression of fibrosis, as assessed by quantitative being associated with an increased risk of death [40, 41].
HRCT at weeks 24 and 48 [31]. Although the results of TGF-β, a key mediator of fibrosis, is secreted in a latent
this study have not yet been published, pamrevlumab has form coupled with latency-associated protein (LAP). Inte-
received fast-track status from the US FDA and is expected grin αvβ6 binds to an arginine-glycine-aspartic (RGD)
to enter into phase III trials in the near future. sequence on LAP to activate TGF-β1 [42]. Low-dose treat-
ment with a monoclonal antibody to αvβ6 attenuates fibrosis
3.5 Galectin‑3: TD139 in a murine bleomycin model [40].
Details of a phase IIa, escalating-dose RCT of BG00011
A pleiotropic, β-galactoside-binding lectin known as galec- (formerly STX-100, Biogen), a humanized anti-αvβ6 immu-
tin-3 (Gal-3) has been implicated in the development of noglobulin (Ig) G1 monoclonal antibody, was presented at
fibrosis in multiple organs, including the liver and kidney the ATS 2018 International Conference [43]. Forty-one
[32, 33]. Elevated levels of galectin-3 in bronchoalveolar IPF patients received either the drug or placebo by weekly
lavage fluid from IPF subjects have been reported, albeit subcutaneous injection for 8 weeks, in five dose cohorts
inconsistently across studies [34–36]. Interestingly, in the (range 0.015–3 mg/kg). Adverse events were reported in
Framingham Heart Study cohort, higher serum galectin-3 four patients treated with BG00011 but were judged not
concentrations were associated with lower FVC, reduced to be related to the drug. pSMAD2, a signal transducer
diffusing capacity of the lung for carbon monoxide ­(DLCO) of TGF-β, was reduced in the BAL fluid by BG00011 in
and interstitial lung abnormalities on CT [37]. a dose-dependent manner. A phase IIb safety and efficacy
Galectin-3 is expressed in numerous cell types, but in the trial over 52 weeks (SPIRIT; ClinicalTrials.gov identifier:
context of IPF is mainly localized to alveolar macrophages NCT03573505) is now recruiting.
IPF: New and Emerging Treatment Options

A small molecule inhibitor of the αvβ6 integrin, analogue that has been developed as an orally administered
GSK3008348 (GlaxoSmithKline), has been formulated for small molecule for the treatment of IPF [52]. It targets the
administration as a nebulised solution. In a phase I trial of G-protein-coupled receptors GPR40 and GPR84, where it
29 healthy participants, it was well tolerated at clinically acts as an agonist and antagonist, respectively [52]. PBI-
relevant doses and demonstrated promising pharmacokinet- 4050 reduced activation and profibrotic gene expression
ics, with no serious adverse events [44]. in human dermal fibroblasts, and histological evidence of
lung fibrosis by 47% in a bleomycin mouse model [52]. In
3.7 Interleukin‑13: Lebrikizumab a phase II, open-label, single-arm study (ClinicalTrials.
gov identifier: NCT02538536), PBI-4050 taken at a dose
Interleukin (IL)-13 is a T-helper (Th) 2 cytokine that medi- of 800 mg daily for 12 weeks was well tolerated and there
ates tissue fibrosis and remodelling, in part through TGF-β1 was no significant decline in FVC when used alone and in
activation [45]. It is increased in lung tissue of bleomycin- combination with nintedanib [53]. A drug–drug interaction
treated mice, and antibody blocking of IL-13 reduces fibro- with pirfenidone resulted in a significant decline in FVC
sis in this model [46]. Lebrikizumab (Roche) is a human- over 12 weeks. This was an exploratory study of 41 patients,
ised monoclonal antibody to IL-13. A phase II, randomised, therefore an appropriately powered RCT is required to ascer-
double-blind, placebo-controlled trial, ‘RIFF’, was under- tain the effectiveness of this agent.
taken to assess lebrikizumab (250 mg subcutaneously every
4 weeks) as monotherapy (Cohort A, n = 154) or in combina- 3.10 Cell‑Based Therapies
tion with background pirfenidone (Cohort B, n = 351) over
52 weeks (ClinicalTrials.gov identifier: NCT01872689). At the core of IPF pathogenesis is failed epithelial repair.
Data from this study were presented at the ATS 2018 Inter- Bone marrow-derived mesenchymal stem cells (MSCs)
national Conference [47, 48]. Despite an acceptable safety transplanted into the lungs of mice contributed to the
profile, lebrikizumab was not associated with a benefit in repair and resolution of injury induced by bleomycin [54].
either the rate of FVC decline or mortality over 52 weeks ‘ASTIS’, a phase III trial of autologous haematopoietic stem
in either cohort. To make an informed judgement on the cell transplantation in patients with diffuse cutaneous sys-
future potential of lebrikizumab in IPF, we await the fully temic sclerosis (83% of whom had interstitial lung disease
published results of the RIFF trial. on HRCT), improved lung function [55]. Utilisation of the
regenerative properties of MSCs for the treatment of IPF
3.8 Phosphatidylinositol‑3‑Kinase/AKT is now under investigation in human trials. Eight patients
and Mammalian Target of Rapamycin Pathway: with moderate–severe IPF received an intravenous infusion
GSK2126458 (Omipalisib) of placenta-derived MSCs from an unrelated donor at a
dose of either 1 × 106 or 2 × 106 MSCs/kg in a single-centre,
For over a decade, phosphatidylinositol-3-kinase (PI3K) and phase Ib trial. No significant short-term safety issues were
its serine/threonine kinases Akt and mTOR, have garnered identified in this study [56]. In accordance with these find-
significant traction in the field of oncology as they play a ings, a phase I trial, ‘AETHER’, administered human bone
critical role in cell survival, growth and proliferation [49]. marrow-derived MSCs via intravenous infusion to nine IPF
PI3K signalling activity is evident in macrophages, bron- patients with no serious adverse events [57]. In an alternative
chial epithelium and fibrotic foci in IPF lung tissue [50]. A approach, three endobronchial infusions of autologous adi-
potent small molecule inhibitor of the PI3K/mammalian tar- pose-derived stem cells–stromal vascular fraction (ADSCs-
get of rapamycin (mTOR) pathway, GSK2126458 (Omipal- SVF) at a dose of 0.5 × 106 cells/kg, in a phase Ib study of
isib), reduces fibroblast proliferation and collagen I synthesis 14 patients with mild–moderate IPF, was associated with no
induced by TGF-β in vitro [50]. In a double-blind, placebo- adverse events [58]. Up to 5 years of longitudinal data from
controlled, dose-escalation, phase Ib study of omipalisib in this study confirm an acceptable safety profile, but has failed
17 IPF patients, the drug had an acceptable safety profile and to demonstrate any alteration in disease course as assessed
[18F]-fluorodeoxyglucose-positron emission tomography by FVC, diffusion capacity of the lungs for carbon monoxide
([18F]-FDG-PET)/CT scans confirmed target engagement (DLco), and 6-min walk test (6MWT) [59].
in the lungs [51]. Diarrhoea was the most common adverse
event, reported by four participants.
4 Priorities for Future Research
3.9 GPR40 and GPR84: PBI‑4050
The early-phase trials discussed in this article underscore
PBI-4050, 3-pentylbenzeneacetic acid sodium salt (Prometic the diversity of therapeutic targets currently under investiga-
BioSciences Inc.), is a synthetic medium-chain fatty acid tion in IPF, a complex disease driven by multiple pathways.
R. J. Hewitt, T. M. Maher

Given the destructive nature of fibrosis in IPF, there is a References


need to target the disease at its inception so as to halt the
relentless accumulation of scar tissue. This requires further 1. Daccord C, Maher TM. Recent advances in understanding
research to establish a clearer understanding of the inciting idiopathic pulmonary fibrosis. F1000Res. 2016. https​://doi.
org/10.12688​/f1000​resea​rch.8209.1.
environmental insults leading to the development of fibrosis, 2. Raghu G, Weycker D, Edelsberg J, Bradford WZ, Oster G. Inci-
and to identify individuals at risk of disease or with subclini- dence and prevalence of idiopathic pulmonary fibrosis. Am J
cal fibrosis. To translate potential therapeutic targets from Respir Crit Care Med. 2006;174(7):810–6.
bench-to-bedside, the preclinical experimental models used 3. Hutchinson J, Fogarty A, Hubbard R, McKeever T. Global inci-
dence and mortality of idiopathic pulmonary fibrosis: a systematic
to identify candidate therapies require optimisation and bet- review. Eur Respir J. 2015;46(3):795–806.
ter validation [60]. To accelerate the development of new 4. Vancheri C, Failla M, Crimi N, Raghu G. Idiopathic pulmonary
IPF treatments, there is a pressing need for trial endpoints fibrosis: a disease with similarities and links to cancer biology.
that provide an earlier readout of antifibrotic efficacy than Eur Respir J. 2010;35(3):496–504.
5. Ley B, Collard HR, King TE Jr. Clinical course and prediction of
is permitted by FVC. This would allow ineffective treat- survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care
ments to be identified and discarded at an earlier stage in Med. 2011;183(4):431–40.
the development process and would enable prioritisation of 6. Raghu G, Anstrom KJ, King TE Jr, Lasky JA, Martinez FJ. Pred-
compounds with a greater chance of success in late-phase nisone, azathioprine, and N-acetylcysteine for pulmonary fibrosis.
N Engl J Med. 2012;366(21):1968–77.
trials. 7. Noble PW, Albera C, Bradford WZ, Costabel U, Glassberg MK,
The older age and comorbidities of IPF patients must Kardatzke D, et al. Pirfenidone in patients with idiopathic pul-
be considered when exploiting therapeutic targets that may monary fibrosis (CAPACITY): two randomised trials. Lancet.
impact biological pathways involved in cellular homeostasis 2011;377(9779):1760–9.
8. King TE Jr, Bradford WZ, Castro-Bernardini S, Fagan EA,
or pose greater risk in association with other health problems Glaspole I, Glassberg MK, et al. A phase 3 trial of pirfenidone
[61]. Targeting the mechanisms of age-associated cellular in patients with idiopathic pulmonary fibrosis. N Engl J Med.
dysfunction may prove an effective strategy and has been 2014;370(22):2083–92.
reviewed extensively elsewhere [62]. 9. Richeldi L, du Bois RM, Raghu G, Azuma A, Brown KK, Cos-
tabel U, et al. Efficacy and safety of nintedanib in idiopathic pul-
monary fibrosis. N Engl J Med. 2014;370(22):2071–82.
10. Raghu G, Rochwerg B, Zhang Y, Garcia CA, Azuma A, Behr J,
et al. An Official ATS/ERS/JRS/ALAT Clinical Practice Guide-
5 Conclusions line: Treatment of Idiopathic Pulmonary Fibrosis. An Update of
the 2011 Clinical Practice Guideline. Am J Respir Crit Care Med.
2015;192(2):e3–19.
Organ fibrosis is a consequence of aging and is a growing 11. Galli JA, Pandya A, Vega-Olivo M, Dass C, Zhao H, Criner GJ.
problem across developed countries. IPF is an exemplar Pirfenidone and nintedanib for pulmonary fibrosis in clinical
fibrotic disease characterised by progressive destruction practice: tolerability and adverse drug reactions. Respirology.
2017;22(6):1171–8.
of the lung, with consequent respiratory failure and death. 12. Vukmirovic M, Kaminski N. Impact of transcriptomics on our
The successful approval of both pirfenidone and nintedanib understanding of pulmonary fibrosis. Front Med (Lausanne).
for IPF has proven that the mechanisms driving progressive 2018;5:87.
fibrosis are therapeutically tractable. The growing portfolio 13. Selman M, Pardo A. Revealing the pathogenic and aging-
related mechanisms of the enigmatic idiopathic pulmonary
of novel treatments targeting multiple potential pathways fibrosis. An integral model. Am J Respir Crit Care Med.
involved in fibrosis speaks to the molecular complexity of 2014;189(10):1161–72.
IPF. Nonetheless, data from early-phase studies provides 14. Maher TM, Wells AU, Laurent GJ. Idiopathic pulmonary fibro-
hope that a new wave of antifibrotic treatments will reach sis: multiple causes and multiple mechanisms? Eur Respir J.
2007;30(5):835–9.
the clinic in the near future. 15. Maher TM. Idiopathic pulmonary fibrosis: pathobiology of novel
approaches to treatment. Clin Chest Med. 2012;33(1):69–83.
Acknowledgements  Toby M. Maher is supported by a National Insti- 16. van Meeteren LA, Moolenaar WH. Regulation and bio-
tute for Health Research (NIHR) Clinician Scientist Fellowship (NIHR logical activities of the autotaxin-LPA axis. Prog Lipid Res.
Ref: CS-2013-13-017) and a British Lung Foundation Chair in Respira- 2007;46(2):145–60.
tory Research (C17-3). 17. Tager AM, LaCamera P, Shea BS, Campanella GS, Selman M,
Zhao Z, et al. The lysophosphatidic acid receptor LPA1 links pul-
Compliance with Ethical Standards  monary fibrosis to lung injury by mediating fibroblast recruitment
and vascular leak. Nat Med. 2008;14(1):45–54.
18. Oikonomou N, Mouratis MA, Tzouvelekis A, Kaffe E, Valavanis
Conflict of Interest  Richard J. Hewitt has no conflicts of interest. Toby C, Vilaras G, et al. Pulmonary autotaxin expression contributes
M. Maher has, via his institution, received industry-academic fund- to the pathogenesis of pulmonary fibrosis. Am J Respir Cell Mol
ing from GlaxoSmithKline R&D and UCB, and has received consul- Biol. 2012;47(5):566–74.
tancy or speakers’ fees from Apellis, AstraZeneca, Bayer, Biogen Idec, 19. Maher TM, van der Aar EM, Van de Steen O, Allamassey L,
Boehringer Ingelheim, Galapagos, GlaxoSmithKline R&D, Indalo, Desrivot J, Dupont S, et al. Safety, tolerability, pharmacokinetics,
Pliant, ProMetic, Roche, Samumed and UCB.
IPF: New and Emerging Treatment Options

and pharmacodynamics of GLPG1690, a novel autotaxin inhibi- 34. Mackinnon AC, Gibbons MA, Farnworth SL, Leffler H, Nils-
tor, to treat idiopathic pulmonary fibrosis (FLORA): a phase son UJ, Delaine T, et al. Regulation of transforming growth fac-
2a randomised placebo-controlled trial. Lancet Respir Med. tor-beta1-driven lung fibrosis by galectin-3. Am J Respir Crit Care
2018;6(8):627–35. Med. 2012;185(5):537–46.
20. Palmer SM, Snyder L, Todd JL, Soule B, Christian R, Anstrom 35. Nishi Y, Sano H, Kawashima T, Okada T, Kuroda T, Kikkawa K,
K, et al. Randomized, double-blind, placebo-controlled, phase 2 et al. Role of galectin-3 in human pulmonary fibrosis. Allergol Int.
trial of BMS-986020, a lysophosphatidic acid receptor antago- 2007;56(1):57–65.
nist for the treatment of idiopathic pulmonary fibrosis. Chest. 36. Cullinane AR, Yeager C, Dorward H, Carmona-Rivera C, Wu
2018;154(5):1061–9. HP, Moss J, et al. Dysregulation of galectin-3. Implications for
21. Misharin AV, Morales-Nebreda L, Reyfman PA, Cuda CM, Walter Hermansky-Pudlak syndrome pulmonary fibrosis. Am J Respir
JM, McQuattie-Pimentel AC, et al. Monocyte-derived alveolar Cell Mol Biol. 2014;50(3):605–13.
macrophages drive lung fibrosis and persist in the lung over the 37. Ho JE, Gao W, Levy D, Santhanakrishnan R, Araki T, Rosas
life span. J Exp Med. 2017;214(8):2387–404. IO, et al. Galectin-3 is associated with restrictive lung disease
22. Castano AP, Lin SL, Surowy T, Nowlin BT, Turlapati SA, Patel and interstitial lung abnormalities. Am J Respir Crit Care Med.
T, et al. Serum amyloid P inhibits fibrosis through Fc gamma 2016;194(1):77–83.
R-dependent monocyte-macrophage regulation in vivo. Sci Transl 38. Hirani N, Mackinnon A, Nicol L, Walker J, Ford P, Schambye
Med. 2009;1(5):5ra13. H, et al. TD139, A novel inhaled galectin-3 inhibitor for the
23. Murray LA, Rosada R, Moreira AP, Joshi A, Kramer MS, Hesson treatment of idiopathic pulmonary fibrosis (IPF). Results from
DP, et al. Serum amyloid P therapeutically attenuates murine bleo- the first in (IPF) patients study. Am J Respir Crit Care Med.
mycin-induced pulmonary fibrosis via its effects on macrophages. 2017;195:A7560.
PLoS One. 2010;5(3):e9683. 39. Hynes RO. Integrins: bidirectional, allosteric signaling machines.
24. Murray LA, Chen Q, Kramer MS, Hesson DP, Argentieri RL, Cell. 2002;110(6):673–87.
Peng X, et  al. TGF-beta driven lung fibrosis is macrophage 40. Horan GS, Wood S, Ona V, Li DJ, Lukashev ME, Weinreb PH,
dependent and blocked by Serum amyloid P. Int J Biochem Cell et al. Partial inhibition of integrin alpha(v)beta6 prevents pulmo-
Biol. 2011;43(1):154–62. nary fibrosis without exacerbating inflammation. Am J Respir Crit
25. van den Blink B, Dillingh MR, Ginns LC, Morrison LD, Moerland Care Med. 2008;177(1):56–65.
M, Wijsenbeek M, et al. Recombinant human pentraxin-2 therapy 41. Saini G, Porte J, Weinreb PH, Violette SM, Wallace WA,
in patients with idiopathic pulmonary fibrosis: safety, pharmacoki- McKeever TM, et al. alphavbeta6 integrin may be a potential
netics and exploratory efficacy. Eur Respir J. 2016;47(3):889–97. prognostic biomarker in interstitial lung disease. Eur Respir J.
26. Raghu G, van den Blink B, Hamblin MJ, Brown AW, Golden 2015;46(2):486–94.
JA, Ho LA, et al. Effect of recombinant human pentraxin 2 vs 42. Munger JS, Huang X, Kawakatsu H, Griffiths MJ, Dalton SL, Wu
placebo on change in forced vital capacity in patients with idi- J, et al. The integrin alpha v beta 6 binds and activates latent TGF
opathic pulmonary fibrosis: a randomized clinical trial. JAMA. beta 1: a mechanism for regulating pulmonary inflammation and
2018;319(22):2299–307. fibrosis. Cell. 1999;96(3):319–28.
27. Lipson KE, Wong C, Teng Y, Spong S. CTGF is a central media- 43. Raghu G, Mouded M, Culver DA, Hamblin MJ, Golden JA,
tor of tissue remodeling and fibrosis and its inhibition can reverse Veeraraghavan S, et al. Randomized, double-blind, placebo-con-
the process of fibrosis. Fibrogenesis Tissue Repair. 2012;5(Suppl trolled, multiple dose, dose-escalation study of BG00011 (For-
1):S24. merly STX-100) in patients with idiopathic pulmonary fibrosis
28. Allen JT, Knight RA, Bloor CA, Spiteri MA. Enhanced insulin- (IPF). Am J Respir Crit Care Med. 2018;197:A7785.
like growth factor binding protein-related protein 2 (Connective 44. Maden CH, Fairman D, Chalker M, Costa MJ, Fahy WA, Gar-
tissue growth factor) expression in patients with idiopathic pul- man N, et  al. Safety, tolerability and pharmacokinetics of
monary fibrosis and pulmonary sarcoidosis. Am J Respir Cell Mol GSK3008348, a novel integrin alphavbeta6 inhibitor, in healthy
Biol. 1999;21(6):693–700. participants. Eur J Clin Pharmacol. 2018;74(6):701–9.
29. Bickelhaupt S, Erbel C, Timke C, Wirkner U, Dadrich M, Flech- 45. Lee CG, Homer RJ, Zhu Z, Lanone S, Wang X, Koteliansky V,
sig P, Tietz A, Pföhler J, Gross W, Peschke P, Hoeltgen L, Katus et al. Interleukin-13 induces tissue fibrosis by selectively stimulat-
HA, Gröne HJ, Nicolay NH, Saffrich R, Debus J, Sternlicht MD, ing and activating transforming growth factor beta [1]. J Exp Med.
Seeley TW, Lipson KE, Huber PE. Effects of CTGF blockade on 2001;194(6):809–21.
attenuation and reversal of radiation-induced pulmonary fibrosis. 46. Belperio JA, Dy M, Burdick MD, Xue YY, Li K, Elias JA, et al.
J Natl Cancer Inst. 2017. https​://doi.org/10.1093/jnci/djw33​9. Interaction of IL-13 and C10 in the pathogenesis of bleomy-
30. Raghu G, Scholand MB, de Andrade J, Lancaster L, Mageto Y, cin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol.
Goldin J, et al. FG-3019 anti-connective tissue growth factor 2002;27(4):419–27.
monoclonal antibody: results of an open-label clinical trial in idi- 47. Swigris JJ, Ogura T, Scholand MB, Glaspole I, Maher TM, Kar-
opathic pulmonary fibrosis. Eur Respir J. 2016;47(5):1481–91. datzke D, et al. The RIFF study (Cohort A): A phase II, rand-
31. Gorina E, Goldin J, Hyun KG, Zhong M, Sekayan T, Yu P, et al. omized, double-blind, placebo-controlled trial of lebrikizumab as
Lung fibrosis measured by quantitative high resolution computed monotherapy in patients with idiopathic pulmonary fibrosis. Am
tomography (qHRCT) in idiopathic pulmonary fibrosis (IPF) J Respir Crit Care Med. 2018;197:A6167.
patients treated with pamrevlumab (FG-3019). Am J Respir Crit 48. Maher TM, Kondoh Y, Corte TJ, Glassberg MK, Costabel U,
Care Med. 2018;197:A7688. Lancaster LH, et al. The RIFF Study (Cohort B): a phase II, ran-
32. Henderson NC, Mackinnon AC, Farnworth SL, Poirier F, domized, double-blind, placebo-controlled trial of lebrikizumab
Russo FP, Iredale JP, et  al. Galectin-3 regulates myofibro- in combination with pirfenidone in patients with idiopathic pul-
blast activation and hepatic fibrosis. Proc Natl Acad Sci USA. monary fibrosis. Am J Respir Crit Care Med. 2018;197:A6168.
2006;103(13):5060–5. 49. Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB. Exploiting
33. Henderson NC, Mackinnon AC, Farnworth SL, Kipari T, Has- the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug
lett C, Iredale JP, et al. Galectin-3 expression and secretion links Discov. 2005;4(12):988–1004.
macrophages to the promotion of renal fibrosis. Am J Pathol. 50. Mercer PF, Woodcock HV, Eley JD, Platé M, Sulikowski MG,
2008;172(2):288–98. Durrenberger PF, et  al. Exploration of a potent PI3 kinase/
R. J. Hewitt, T. M. Maher

mTOR inhibitor as a novel anti-fibrotic agent in IPF. Thorax. 56. Chambers DC, Enever D, Ilic N, Sparks L, Whitelaw K, Ayres J,
2016;71(8):701–11. et al. A phase 1b study of placenta-derived mesenchymal stromal
51. Lukey PT, Harrison SA, Yang S, Man Y, Holman BF, Rashidna- cells in patients with idiopathic pulmonary fibrosis. Respirology.
sab A, Azzopardi G, Grayer M, Simpson JK, Bareille P, Paul L, 2014;19(7):1013–8.
Woodcock HV, Toshner R, Saunders P, Molyneaux PL, Thiele- 57. Glassberg MK, Minkiewicz J, Toonkel RL, Simonet ES, Rubio
mans K, Wilson FJ, Mercer PF, Chambers RC, Groves AM, Fahy GA, DiFede D, et  al. Allogeneic human mesenchymal stem
WA, Marshall RP, Maher TM. A randomised, placebo-controlled cells in patients with idiopathic pulmonary fibrosis via intrave-
study of omipalisib (PI3K/mTOR) in idiopathic pulmonary Fibro- nous delivery (AETHER): a phase I safety clinical trial. Chest.
sis. Eur Respir J. 2019. https​://doi.org/10.1183/13993​003.01992​ 2017;151(5):971–81.
-2018. 58. Tzouvelekis A, Paspaliaris V, Koliakos G, Ntolios P, Bouros E,
52. Gagnon L, Leduc M, Thibodeau J-F, Zhang M-Z, Grouix B, Oikonomou A, et al. A prospective, non-randomized, no placebo-
Sarra-Bournet F, et al. A newly discovered antifibrotic pathway controlled, phase Ib clinical trial to study the safety of the adipose
regulated by two fatty acid receptors: GPR40 and GPR84. Am J derived stromal cells-stromal vascular fraction in idiopathic pul-
Pathol. 2018;188(5):1132–48. monary fibrosis. J Transl Med. 2013;11(1):171.
53. Khalil N, Manganas H, Ryerson CJ, Shapera S, Cantin AM, Her- 59. Ntolios P, Manoloudi E, Tzouvelekis A, Bouros E, Steiropoulos P,
nandez P, Turcotte EE, Parker JM, Moran JE, Albert GR, Sawtell Anevlavis S, et al. Longitudinal outcomes of patients enrolled in a
R, Hagerimana A, Laurin P, Gagnon L, Cesari F, Kolb M. Phase phase Ib clinical trial of the adipose-derived stromal cells-stromal
2 clinical trial of PBI-4050 in patients with idiopathic pulmo- vascular fraction in idiopathic pulmonary fibrosis. Clin Repir J.
nary fibrosis. Eur Respir J. 2018. https​://doi.org/10.1183/13993​ 2018;12(6):2084–9.
003.00663​-2018. 60. Bonniaud P, Fabre A, Frossard N, Guignabert C, Inman M, Kue-
54. Rojas M, Xu J, Woods CR, Mora AL, Spears W, Roman J, et al. bler WM, et al. Optimising experimental research in respiratory
Bone marrow-derived mesenchymal stem cells in repair of the diseases: an ERS statement. Eur Respir J. 2018;51(5):1702133.
injured lung. Am J Respir Cell Mol Biol. 2005;33(2):145–52. 61. Raghu G, Amatto VC, Behr J, Stowasser S. Comorbidities in idi-
55. van Laar JM, Farge D, Sont JK, Naraghi K, Marjanovic Z, Lar- opathic pulmonary fibrosis patients: a systematic literature review.
ghero J, et al. Autologous hematopoietic stem cell transplanta- Eur Respir J. 2015;46(4):1113–30.
tion vs intravenous pulse cyclophosphamide in diffuse cuta- 62. Mora AL, Rojas M, Pardo A, Selman M. Emerging therapies for
neous systemic sclerosis: a randomized clinical trial. JAMA. idiopathic pulmonary fibrosis, a progressive age-related disease.
2014;311(24):2490–8. Nat Rev Drug Discov. 2017;16(11):755–72.

Вам также может понравиться