The oncogenic landscape of the idiopathic pulmonary fibrosis: a narrative review
Review Article

The oncogenic landscape of the idiopathic pulmonary fibrosis: a narrative review

Giulia Maria Stella1, Vito D’Agnano2, Davide Piloni1, Laura Saracino1, Sara Lettieri1, Francesca Mariani1, Andrea Lancia3, Chandra Bortolotto4, Pietro Rinaldi5, Francesco Falanga5, Cristiano Primiceri5, Angelo Guido Corsico1, Andrea Bianco2

1Department of Medical Sciences and Infective Diseases, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, Pavia, Italy; 2Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, Napoli, Italy; 3Department of Medical Sciences and Infective Diseases, Unit of Radiation Therapy, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, Pavia, Italy; 4Department of Intensive Medicine, Unit of Radiology, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, Pavia, Italy; 5Department of Intensive Medicine, Unit of Thoracic Surgery, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, Pavia, Italy

Contributions: (I) Conception and design: GM Stella, V D’Agnano, AG Corsico, D Piloni, A Bianco; (II) Administrative support: None; (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: GM Stella, D Piloni, L Saracino, S Lettieri, F Mariani, C Bortolotto, A Lancia, P Rinaldi, C Primiceri; (V) Data analysis and interpretation: GM Stella, D Piloni, L Saracino, S Lettieri, F Mariani, C Bortolotto, A Lancia, P Rinaldi, C Primiceri; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Giulia Maria Stella, MD, PhD. Department of Medical Sciences and Infective Diseases, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27100 Pavia, Italy. Email: g.stella@smatteo.pv.it.

Background and Objective: Translational research is a source of continuous innovation in medicine, more particularly for clinical research on new treatment modalities in idiopathic pulmonary fibrosis (IPF) patients. However, the heterogeneity of the disease is well recognized, and different pathological and molecular settings have been identified. The molecular mechanisms by which IPF proceeds in time and space remains poorly understood. Although some IPF features are reminiscent of cancer, the dynamics of malignant divergent clonal selective pressure and heterogeneity clearly differ from those occurring in IPF. This is reflected in the absence of patient proper selection and stratification to biological agents (pirfenidone, nintedanib) which limit therapeutic efficacy. Consequently, increased costs are related to the clinical management of advanced IPF patients. Steady collaboration and fluid communication between pneumo-oncologists, radiologists and molecular biologists is a clear priority for the correct interpretation of tests and the definition of effective personalized strategies against this orphan disease. The present work aims at providing the most relevant hints shared by cancer and IPF.

Methods: A systematic literature review was performed to identify all relevant data. The examined databases were Scopus, Web of Science, Cochrane, Google Scholar, and PubMed. The last search was run on January 5, 2022. We have primarily conducted separated research for lung cancer, IPF, genetics, epigenetics, surgery in IPF and cancer.

Key Content and Findings: The data here presented mainly focus on gene mutations, epigenetics and novel therapeutic approaches. Moreover, epidemiology, prognostic variables and in new treatment strategies adopted in patients with IPF and lung cancer are discussed as well.

Conclusions: Overall, the findings of this narrative review will be of help in defining the key molecular features that could applied in IPF setting with promising rationale to improve therapy and to better manage those cases carrying IPF and cancer concomitantly.

Keywords: Idiopathic pulmonary fibrosis (IPF); cancer; genetics; immunotherapy; personalized medicine


Submitted Nov 06, 2021. Accepted for publication Feb 16, 2022.

doi: 10.21037/tlcr-21-880


Introduction

Idiopathic pulmonary fibrosis (IPF) is characterized by a proliferative landscape, which recalls—under several aspects—that of cancer. This critical issue has been already exploited for therapeutic purposes taking advantage from know-how and expertise from cancer pharmacology. Moreover, IPF diagnosis is associated to a significantly higher risk of lung cancer development (1,2). Notably the coexistence of IPF is associated to a more unfavourable prognosis in lung cancer patients who generally experience severe disease exacerbation during antineoplastic therapy (3-5). Others and we already described bio-molecular similarities and differences between IPF and cancer (6-10) (Figure 1), however some points need deeper clarification and update.

Figure 1 IPF and cancer. The two diseases share common pathogenic pathways that should be exploited for novel therapeutic approaches. The oncogenic gain behaves as main driver of proliferative and invasive phenotypes. Heterogeneity which characterizes both diseases, refers to clonal selection (cancer) and histology (IPF). The specific IPF context impacts on the therapeutic exploitation of targeting oncogenes. IPF, idiopathic pulmonary fibrosis. IPF, idiopathic pulmonary fibrosis.

The concept that interstitial lung diseases (ILDs) represent a relevant risk factor for lung cancer development is well documented and known (11-20). Within respect to IPF, reports indicated a cumulative incidence of cancer in IPF patients varying from 3.3%, 15.4%, and 54.7% after 1, 5, and 10 years of follow-up for IPF (21) to 41% and 82% at 1 and 3 years, respectively (5). Age and smoking habit act as known confounding variables since they impact on both lung cancer and IPF onset (19,20,22,23). Moreover, many occupational and environmental exposure toxics are common risks for the development of both the diseases. Notably, IPF patients are at higher risk of cancer development if compared to those affected by chronic obstructive pulmonary disease (COPD), another cancer predisposing pathologic entity (24). The Japanese Hokkaido registry data reports an unadjusted risk ratio of 7.8 for lung cancer in IPF patients vs. COPD ones (25,26). Most often tumors in IPF context arise in peripheral lung (27,28), although these data need further confirmation (19). The mechanistic explanation and the association between IPF and cancer are discussed in detail in the next sections of the manuscript. However, several issues deserve to be here underline. It is conceivable that the pro-proliferative landscape that characterizes IPF, should promote the selection of those cells carrying oncogenic mutations (29-31). Pirfenidone and nintedanib act as antifibrotic drugs through different mechanisms. The first essentially acts by deregulating a series of cytokines, including transforming growth factor (TGF)-β1, connective tissue growth factor (CTGF), platelet-derived growth factors (PDGF), and tumor necrosis factor (TNF)-α. Moreover, it behaves as scavenger of reactive oxygen species (ROS) and downregulate angiotensin-converting enzyme (ACE) expression (32,33). Nintedanib is a multikinase inhibitor which also down-regulates protein and mRNA expression of extracellular matrix (ECM) proteins, fibronectin, and collagen 1a1 and inhibits (TGF)-β1-induced myofibroblast differentiate (34). Notably, both drugs inhibited collagen I fibril formation (35). It should be underlined that a relationship exists between these main two treatments for IPF, namely pirfenidone and nintedanib, and lung as well. Several recent studies have shown a prophylactic effect of the use of pirfenidone perioperative setting against postoperative acute IPF exacerbations in patients with lung cancer (36-40). Notably therapy with pirfenidone seems to be associated to lower incidence of lung cancer in IPF patients if compared to non-pirfenidone treated cases (41), although this observation should be confirmed by more extensive analysis. Some recent observation also underlined a potential therapeutic role of pirfenidone against lung cancer. In detail, it has been reported in vitro and in vivo that it could suppressed activation of non-small-cell lung carcinoma (NSCLC) associated myofibroblasts (42), which are known to be involved in tumor progression (43-45) and impairs epithelial–mesenchymal transition (EMT) by acting on exogenous TGF-β1 and on paracrine TGF-β produced from NSCLC cells (46). Pirfenidone seems to play a synergic effect with conventional chemotherapy such as carboplatin (47), whereas studies evaluating effects of combination with immune checkpoint (IC) inhibitors are ongoing (the NCT04467723 trial evaluating the combination of pirfenidone with the programmed death-ligand 1 (PD-L1) and programmed cell death protein 1 (PD-1) inhibitor atezolizumab in second-line and beyond NSCLC, website at www.clinicaltrials.gov). The antiproliferative effect of nintedanib derives to its ability to block the vascular endothelial growth factor (VEGF), PDGF and the fibroblast growth factor receptor (FGFR). Nintedanib in combination with docetaxel is approved as second-line therapy for advanced NSCLC (48). It also promotes antitumor immunity and antitumor activity in combination with PD-1 blockade in mice by targeting cancer-associated fibroblasts (CAF) thus attenuating the immunosuppressive tumor microenvironment on one hand and promoting intratumoural activation of antitumor CD8+ T cells (49). Although some reports suggesting a positive effect (50-52), it is still unclear if nintedanib could play an effective role against lung cancer aroused in IPF patients. When associated with corticosteroids, it seems to be able to attenuate targeted drug (53) and IC inhibitor-related pneumonitis in cancer patients (54,55). Thus, we—here—report and discuss more recent advances from multidisciplinary contexts that will result in significant changes in the diagnosis and treatment of IPF patients. We present the following article in accordance with the Narrative Review reporting checklist (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-21-880/rc).


Methods

A systematic literature review was performed to identify all relevant data. The examined databases were Scopus, Web of Science, Cochrane, Google Scholar, and PubMed. The last search was run on January 5, 2022. Table 1 summarizes the search strategy.

Table 1

The search strategy summary

Items Specification
Date of Search (specified to date, month and year) Last search January 5, 2022
Databases and other sources searched Scopus, Web of Science, Cochrane, Google Scholar, and PubMed. For the Google Scholar database, due to the excessive amount of data obtained, only the first 200 results for each search were considered, because further results rapidly lost relevance
Search terms used (including MeSH and free text search terms and filters) Lung cancer, non-small cell lung carcinoma (NSCLC), idiopathic pulmonary fibrosis (IPF), epigenetic, genetic, surgery + IPF, ionizing radiation + IPF
Timeframe 5 years
Inclusion and exclusion criteria (study type, language restrictions, etc.) To obtain the highest search sensitivity, the keywords used to identify relevant articles were mainly: lung cancer OR NSCLC AND IPF AND genetics OR IPF AND NSCLC AND epigenetics; imaging AND IPF OR IPF AND CT AND ionizing radiation; IPF OR AND lung cancer AND surgery
Selection process (who conducted the selection, whether it was conducted independently, how consensus was obtained, etc.) Two authors (GMS and SL) independently screened the titles of the identified studies. GMS, AGC and AB independently screened the titles and the abstracts of the studies; then, they read the full text of selected studies. Any disagreement was analyzed and overcome by discussion and reaching a mutual agreement

Any additional considerations, if applicable.


Advances on pathogenic mechanisms of IPF: what we could learn from molecular epidemiology linking lung cancer and fibrosis

Genetics

Several already published data from genome-wide association and linkage studies have identified common genetic variants that are associated to increased risk of IPF onset and progression. Moreover, these IPF-related signature activation define a biologic context which essentially support clonal selection of transformed cells. Consequently IPF-related gene variants can be defined as: (I) IPF private; (II) favouring malignant transformation and (III) shared by IPF and lung cancer. The first gene set mainly affect genes encoding for proteins related to inflammatory and immune response, such as TGF-β1 (56,57), interleukin-1 receptor alpha (IL1RN) (58,59), interleukin 8 (IL8) (60), toll-like receptor 3 (TLR3) (61) human leukocyte antigen (HLA) DRB1*1501 (62), the cell-cycle progression related genes Cyclin Dependent Kinase Inhibitor 1A (CDKN1A) and tumor protein 53 (TP53) (63) and the Telomerase Reverse Transcriptase (TERT) genes. All these genes, known to confer a risk for IPF, are known to be associated to cancer as well. However, except for the tumor suppressor TP53, these genes do not behave as oncogenic drivers but rather their activation by genetic changes cooperates in sustaining malignant transformation. Telomerases are an enzyme that catalyse the addition of nucleotides on the ends of chromosomes and IPF is characterized by shortening of telomere lengths and consequent exhaustion of lung stem cells. Mutations in the genes encoding telomerase, TERT and telomerase RNA component (TERC), are pathogenetically associated to IPF. Mutational frequency affecting each gene is rare, but—overall—TERT mutations are the most common genetic defect found in FPF. The overall penetrance of pulmonary fibrosis in TERT mutation carriers is 40% in subjects with a mean age of 51 years (64-68). It could be hypothesized that the biologic landscape, which is linked to IPF, defined by pro-invasive, anti-apoptotic and pro-angiogenic features and properties of associated genes and molecules, could be exploited by tumour cells (already transformed) to optimize their malignant propensity, according to the biologic phenomenon already defined by “oncogene expedience” (69). In other words, the IPF genetic asset impacts on the risk of lung-cancer development although IPF-associated lung cancer does not derive from transformation due to mutation accumulation of IPF-related cells (70). With the expansion of genome-wide association studies (GWAS) novel biomarkers and actionable targets have been unveiled and new insights have been specifically derived by the integration of molecular techniques and conventional epidemiology, namely molecular epidemiology (71). This approach has been widely exploited in cancer, whereas few data are available for IPF (72). The identification of novel diagnostic and therapeutic endpoints, quantification of genetic damages, definition of genetic susceptibility for IPF could potentially derive from comparative studies on IPF-associated lung cancer. According to standard epidemiologic approach the two diseases share relevant environmental and occupational risk factors, such as tobacco smoke, dusts, and particulates, as well as some therapeutic agents. Moreover, IPF and lung cancer present lineage specifiers which underline a common cell-fate specification. The thyroid transcription factor-1 (TTF-1) also known as NK2 Homeobox 1 (NKX2.1) is a homeodomain-containing transcription factor, that is essential for the morphogenesis and differentiation of the thyroid, lung, and ventral forebrain. It controls the expression of select genes in the thyroid, lung, and the central nervous system. In the lung, TTF-1 is a critical regulator of the expression of surfactant proteins that are essential for lung morphogenesis, homeostasis, and host defence (73). TTF-1 is expressed in type II epithelial cells, less abundantly in non-ciliated respiratory epithelial cells and basal cells whereas it is not expressed in type I cells (29). TTF-1 is expressed lung after injury, and it may play a role in epithelial cell proliferation and differentiation during the repair processes, as fibrosis and cancer. In transgenic mice, increased TTF-1 expression caused severe inflammation, pulmonary fibrosis, respiratory failure, and death, associated with eosinophil infiltration and increased expression of eotaxin and interleukin 6 (IL-6). Increased expression of TTF-1 altered alveolarization and caused chronic pulmonary inflammation. In adults, TTF-1 is almost exclusively expressed in thyroid and pulmonary epithelial cells. Its expression, determined by immunohistochemistry, is a highly specific marker for primary lung adenocarcinomas (ADCs) and it must be used for the differential diagnosis between primary and metastatic ADCs (74,75). TTF-1 gene amplifications can be detected in about 2–4% of primary lung and 13% of metastatic lesions (76-78); activated TTF-1 promotes epidermal growth factor receptor (EGFR) driven transformation (79,80). Overall TTF-1 behave as oncogene in a lineage specific (ADC) context (81). However, opposing, and paradoxical effects have been reported in animal models carrying TTF-1 haploinsufficiency being associated with reduction of invasive and metastatic potentials because of a suppressive modulation on the genes implicated in cytoskeletal regulation, cell-cell organization, and motility. Interestingly, it is also associated to the enhancement of kirsten rat sarcoma (KRAS)-driven adenocarcinogenesis (82-84). TTF-1 is known to repress TGF-beta EMT by reducing TGF-beta and the TGF-beta related activation of Snail and Slug. On the contrary TGF beta represses TTF-1 through miR-365 (85). Lung cancer (LC) is genetically characterized by the presence of somatic mutations which are known to be selected by a variety of environmental exposures (among which tobacco smoke) on the background of specific germline mutations. Accumulation of somatic mutations affecting the RAS-RAF cascade has been reported with significantly higher prevalence of v-raf murine sarcoma viral oncogene homolog B1 (BRAF) mutations which define a novel potentially actionable target (86). Interestingly, growing evidence suggests that the occurrence of some germline mutations might predispose subjects to the development of IPF and to LC as well. The most intriguing changes that have been reported in lung tumors associated to IPF familial clusters refer to heterozygous missense mutations in pulmonary surfactant-associated proteins genes. It is well known that variants in the genes encoding for proteins A2 (SFTPA2) (87) and A1 (SFTPA1) (88) display pathogenic role and predispose to IPF (89) by impairing secretion of surfactant A proteins (SP-A) thus leading to protein instability and dysfunction of endoplasmic reticulum (ER). The latter induces stress and alter differentiation of resident alveolar type II (ATII) cells (45,90). Genetic variants of SP-A can, thus, interfere with intracellular protein trafficking and promoting cell proliferation in both lung fibrosis and malignant transformation (87). Similarly, mutations affecting genes encoding surfactant protein C (SFTPC) (91,92) are linked to lung fibrosis whereas protein-D gene variants (SFTPD) have been reported in paediatric cases of diffuse ILD (93). Inherited lung fibrosis is also associated to the occurrence of mutations affecting the gene encoding for the member A3 of the ATP-binding cassette family. Withing respect to the context of cancer, the ATP transporters are known to mediate chemoresistance (94) and, more recently, their role in all phases of disease onset and progression has been considered and documented (95). A comprehensive analysis retrieved from Gene Expression Omnibus (GEO) database reported the that genes encoding for peroxisome proliferator-activated receptor (PPAR) signalling pathway transducers were enriched in IPF associated to LC (96). The PPARs define a group of three nuclear receptor isoforms, PPAR-γ, PPAR-α, and PPAR-δ, encoded by different genes. They act as ligand-regulated transcription factors that control gene expression by binding to specific response elements (PPREs); they are known to play a critical physiological role as lipid sensors and regulators of lipid metabolism and are involved in cell proliferation. Deregulation of PPAR signalling has been reported in several disease including atherosclerosis, inflammation, cancer, infertility, and demyelination (97-99). Although the exact mechanism of PPARs in lung fibrosis and LC remains largely unknown, a PPARγ agonist, including a constitutively active PPAR-γ construct (VP16-PPAR-γ), has been found to exert antitumorigenic effects in both IPF and LC by inhibiting myofibroblast differentiation through the blockade of TGF-β and activating phosphatase and tensin homolog (PTEN) (100). The Acyl-CoA Dehydrogenase Long Chain (ACADL), cluster of differentiation 36 (CD36), Lipoprotein Lipase (LPL), and Matrix metalloproteinase-1 (MMP1) gene signatures in the PPAR pathway have been reported to be shared among IPF and LC (96). In vivo experiments demonstrated that silencing of CD36 resulted in the inhibition of TGF-β activation in a rat silicosis model, ultimately blocking silica-induced lung fibrogenesis (101). Moreover, a population enriched in CD36+ macrophages has been reported in in the lungs of patients affected by IPF (102). CD36 promotes adipocytes genesis and differentiation (103); in lung fibrosis its expression is involved in the transformation of latent TGF-β1 to active form (101) and a CD36 synthetic peptide reduces fibrotic tissue alteration and collagen accumulation in a mouse model of silicosis (104). Increased CD36 copy number has been related to tumor progression and increased risk of metastatic development (105). Interestingly, the IPF gene expression analysis indicates that the genes regulated by hypoxia are altered, suggesting a primary role of hypoxia-inducible factor 1-alpha (HIF-1α) in IPF onset (106). Overall, these data are coherent with most recent publications suggesting that a metabolic signature, linked to lipid mediators derived from phospholipids, sphingolipids, and polyunsaturated fatty acids play an important role in the pathogenesis of IPF (107-109). Coherently the ACADL gene, encoding for long-chain acyl-CoA dehydrogenase, an enzyme involved in fatty acid beta-oxidation and implicated in homeostasis of pulmonary surfactant (110), is known to be deregulated in IPF and to be a core signature gene that differentiates NSCLC from normal tissue. In cancer context ACADL seems to behave as tumor suppressor (111) and its expression correlates with aggressive tumor phenotype (112) and poor prognosis (113). The LPL gene has been reported to be deregulated in IPF where it is significantly upregulated in NSCLC if compared to surrounding healthy tissue (52). In this context the matrix metalloproteinases MMPs, a family of 25 secreted and cell surface-bound neutral proteinases, play a crucial role. The MMP1 seems to be the most significantly altered gene shared across IPF and LC. The human MMP1 consists of 10 exons and is located on human chromosome 11q22.2-22.3. This gene is tightly linked to a cluster of eight MMP genes, including MMP3, MMP7, MMP8, MMP10, MMP12, MMP13, MMP20, and MMP27, and two pseudogenes (114). MMP1 participates in several processes which characterize both cancer and IPF such as ECM remodelling, cell plasticity, cell migration, and angiogenesis (115-117). MMP1 mutations are associated with COPD (118). A large amount of data reported that MMP1 is highly expressed in interstitial collagenase degrading fibrillar collagens as well as in cancer tissues, where it acts by promoting invasive potential and distant spreading (119). Several single nucleotide polymorphisms (SNPs) were identified to be individually significantly associated with risk of early-onset LC (120); notably MMP1 expression, measured by immunohistochemical (IHC), was reported to be higher in those NSCLC tissues associated with IPF, even in early-stage diseases. Most recent data emerge regarding the common genetic signature between IPF and cancer. Ammar et al. analysed samples from Lung Tissue Research Consortium (LTRC) and National Jewish Health (NJH) cohorts, identified genetic signature able to predict the IPF condition. Some genes were already known to be related to the pathogenesis of IPF such as matrix metalloproteinases, some others identified potentially targetable pathways such as the frizzled-related protein 2 (FRP2), a WNT-signalling (121) or identify predictive disease biomarkers as the Glutathione Peroxidase-3 (GPX3) gene, expressed in epithelial cell from bleomycin-induced fibrosis models and upregulated in IPF (122). Interestingly, maternal smoking and e-smoking negatively affects WNT signalling cascade in mouse models. It affects mRNA expression of the WNT transducers frizzled7 (Fzd7) and Ctnnb1 (gene symbol of β-catenin) as well as the WNT target gene Fn (fibronectin) with significant implication lung development and homeostasis which could generate a favourable substrate for future onset of interstitial fibrosis (123-125). Moreover, a novel set of 12 disease-relevant translational gene markers (C6, CTHRC1, CTSE, FHL2, GAL, GREM1, LCN2, MMP7, NELL1, PCSK1, PLA2G2A, and SLC2A5) can split IPF vs. control patients (126). Another report on 35 matched tumor/IPF samples reported that somatic mutations occurred with predominant C/T transitions despite extensive smoking histories, thus suggesting more associations with APOBEC3B-related mutagenesis in the process of IPF-LC development, rather than smoking. TP53 (62.9%) and BRAF (17.1%) genes were found significantly mutated in IPF-LC. Recurrent focal amplifications in 3 chromosomal loci (3q26.33, 7q31.2, and 12q14.3), and 9p21.3 deletion were identified, and genes associated with JAK-STAT signalling pathway were significantly amplified in IPF-LC (P¼0.012). Moreover, one case report on laser-assisted microdissected samples of IPF associated to lung cancer identified five mutations (KDR, EPHA5, APC, CREBBP, and ERBB2) proper of IPF, four mutations (EPHA5, PKHD1, RB1, and KEAP1) proper of IPF-associated cancer whereas the only mutated gene shared by both the diseases was EPHA5 (118,127). The latter belongs to the ephrin receptor subfamily of the protein-tyrosine kinase family which are known to play a role in several developmental events and in cancer (128) as well as in modulating tumor surrounding microenvironment, being associated to enhanced infiltration of CD8+ T cells and M1 macrophages, reduced recruitment of immunosuppressive regulatory T cells (Tregs) into the tumor site, with prognostic and predictive value (129-131).

Epigenetics

The heterogeneous genetic background which characterized IPF cellular populations is and the possible presence of cells featuring characteristics of stemness define the genesis, maintenance, and plasticity of these cells. In this context the process of lung fibrogenesis is extensively regulated by epigenetic remodelling. Epigenetic mechanisms which modulate the expression of fibrotic genes are emerging as driver players in lung fibrogenesis (132-134). Indeed, several epigenetic regulators are deregulated in IPF: DNA methyltransferases, non-coding RNAs, histone demethylases, and histone acetyltransferases (135). Most studies aiming at analysing epigenetic profile of IPF have been conducted on DNA or mRNA extracted on fixed-formalin paraffin-embedded (FFPE) or frozen samples that are generally mixed before analysis. In this perspective it should be remarked that a relevant role is played not only by the epithelial cells and fibroblasts, but also by alveolar macrophages (AMs). Targeting epigenome represents a potential strategy for fibrosis treatment. Changes in the epigenome are associated with the development and function of AMsin the IPF lung (136,137). Most methylation changes have been identified outside of CpG islands and several gene expression signatures have been reported known (e.g., collagens, HDAC4, NOTCH1, PDGF, SERPINF1, and TOLLIP) and novel candidate (CASZ1) genes. Moreover, environmental stimuli contribute to epigenetic changes. Several components of cigarette smoke have been reported to affect epigenome not only in lung cancer but also in IPF (138,139). Exposure to cigarette smoking has been associated to increased global levels of histone methylation (140). Overall, genes with differentially methylated CpG islands in their promoters are involved in key biologic processes which are implicated in IPF and cancer onset, such as cell assembly, morphology and organization, cell growth, proliferation, signalling, and apoptosis. Some alterations involve the COL18A1 gene, known to be upregulated in IPF (141), genes that modulate myofibroblast differentiation and transition from pericytes to myofibroblasts as NOTCH1 (142-144) and progressive IPF as SMARCA4 (145) as well as the promoter of CXCL3, which is involved in bleomycin-induced fibrosis (146). Moreover the Serine/Threonine Kinase 17b (STK17B) and Serine/Threonine Kinase 3 (STK3) and the histone cluster 1 H2ah have been reported to be up-regulated in IPF, coherently with the hypo-methylated state of their promoter associated CpG islands (147). Moreover epigenetic regulation interferes in the capacity of fibroblasts from lung fibrosis to up-regulate cyclooxygenase-2 (COX-2) expression and COX-2-derived Prostaglandin E2 (PGE2) synthesis, through a mechanism involving hypermethylation of the transcriptional regulator, c8orf4 (148). Interestingly, hypermethylation of the Thy-1 gene promoter region causes the loss of this molecule, which in more invasive behaviour of cancer and the transformation of fibroblasts into myofibroblasts within fibroblast foci in IPF.

Although DNA hypomethylation is a hallmark of cancer and common epigenetic traits are shared between IPF and cancer, some reported data suggest that the similarities in the differentially methylated CpG islands are unexpectedly limited global meth being less extensive in IPF (104). This observation points out methylation occurs in both diseases but should occur through different mechanisms. Excessive histone deacetylation is involved in the progression of pulmonary fibrosis through a complex interaction between histone deacetylases (HDACs) or histone acetyltransferases and fibrosis modulators TGF-beta1, small mother against decapentaplegic (Smad) 3, Smad7 and Snail (149). At this regard histone acetyltransferase EP300, upregulated by TGF-beta1, accumulates in lung fibroblasts and, in turn, promotes SMAD-mediated TGF-beta signalling. EP300 also activates discoidin domain receptor 1 (DDR1), a collagen receptor kinase which triggers ECM deposition (150) and promotes transcription of profibrotic molecules such as alpha-smooth muscle actin (α-SMA), Collagen I (COL1) and tissue inhibitor of metalloprotein, which further promote production of ECM (151). EPP is also frequently mutated in small cell lung cancer (SCLC) (152). Similarly to IPF, indeed, histone deacetylation is associated with lung cancer progression, resistance to chemotherapy and targeted therapy, and is harboured by nickel, chromate, arsenite present in smoke tobacco. Histone deacetlylases I, I, III, VIII are the most common classes hyperexpressed in NSCLC and IPF and their deregulation is associated to poor prognosis in lung cancer (153). At higher level, histone acetylation is regulated by bromodomain and extra-terminal (BET) proteins, a family of chromatin readers (including BRD2, BRD3, BRD4, and BRDT) that bind acetylated histones, regulate gene transcription, and pass on epigenomic memory across cell divisions. BET proteins are implicated in cancer through activation of oncogenes like c-Myc, IL-7R, FOSL1, and E2F; among these, BRD4 has shown to exert profibrotic action in a variety of organs, included lung, by regulating multiple gene programs and biochemical pathways in various cell types, although precise mechanisms have still to be elucidated. In IPF BDR4 probably activates specific enhancers and promoters that regulate transcription of downstream genes encoding ECM proteins such as α-SMA, COL1A1, fibronectin, and factors that stimulate trans-differentiation of fibroblasts (154). Moreover, BDR4 upregulates the pro-oxidant enzyme NADPH oxidase 4 (Nox4), promoter of oxidative stress and cellular ageing (155). The similarity between cancer and IPF is higher if referred to microRNA expression such as in the case of let-7d and hsa-miR-21, which are found to deregulated in both diseases (156-159). Let-7d inhibits EMT through modulation of signalling mediators downstream TGF beta, key orchestrator of fibrogenesis (160). Low levels of Let7d have been found in bronchoalveolar lavage-derived exosomes of IPF murine models compared with normal mice (161). Let-7d downregulates FoxM1, a transcription factor previously known to promote cell proliferation and resistance to apoptosis in cancer cells by activation of Wnt/beta catenin and TGF-beta/SMAD3 pathways (162). Intriguingly, also in IPF high levels of FoxM1 have been found in lung fibroblasts where enhance differentiation of pericytes into myofibroblasts and collagen production, through activation of pathways common with carcinogenesis (70,163). The oncogene Mir-21 promotes cell proliferation, invasion, migration and radioresistance in various cancers, among which NSCLC, regulating nuclear factor kappa-light-chain-enhancer of activated B cells (NF-Kb), phosphatase and tensin homolog/akt murine thymoma viral oncogene homolog (PTEN/AKT), phosphoinositide 3-kinases (PI-3K)/AkT/mammalian target of rapamycin (mtor) pathways; moreover it induces EMT in cancer cells reducing the inhibitory effects of SMAD7 on fibrosis with upregulation of TGF-beta signalling (164). Similarly, in IPF, TGF-beta-induced overexpression of miR-21 in fibroblasts and myofibroblasts creates a positive loop mechanism in which negative modulation of SMAD7 and PTEN increases the expression of TGF-beta, promoting EMT and matrix collagen deposition (165,166). Both miR-29a and miR-185 have been found downregulated in bronchoalveolar lavage fluid (BALf) of IPF and lung cancer patients, probably in response to high levels of TGF beta. However, their hypoexpression produced different effects on the common target COL1A1, whose expression was increased in IPF but not in lung cancer, pointing out the peculiar fibrotic nature of IPF (167). Despite similarities, some microRNAs (miRNAs) are inversely expressed, suggesting the existence of disease-specific mechanisms, which complicate the identification of actionable targets effective in both conditions. For example, the hsa-miR-17-92 miRNA cluster, which encodes six miRNAs (hsa-miR-17, -18a, -19a, -19b, -20a, -92a), is overexpressed in various solid neoplasms, including lung cancer, where behaves as tumor promoter: its block by therapeutic agents, such as Docosahexaenoic acid, may limit cell proliferation, resistance to apoptosis and metastatization (168). However, miR-17-92 cluster has a crucial role for a balanced lung cell damage repair and for regulation of fibrotic genes such as COL1A1, COL1A3, CTGF, VEGF, TGF beta, MMP-1, MMP-7 and MMP-9. Levels of the miR 17-92 cluster appear reduced in IPF lung tissue and fibroblasts for hypermethylation of promotor CpG islands by DNA methyltransferases (DNMTs), enzymes that may seem an effective actionable target to reduce lung fibrosis, as shown by Dakhlallah et al. (169). Unlike miRNA, whose action has been extensively studied, regulatory function of long non-coding RNAs (lnc-RNAs) is still largely unknown. Lnc-RNAs are single-stranded RNA sequences longer than 200 nucleotides, classified in 5 classes (intergenic, antisense, intronic, enhancers, and pseudogenes) according to the positional relationship with protein-coding genes (170). Lnc-RNAs form complexes with DNA, RNA, and proteins, regulating cellular processes such as chromatin modification, transcription, post-transcriptional modifications, scaffolding. Transcriptome sequences analyses reveal more than 1,800 lnc-RNAs deregulated in IPF (171). They upregulate mTOR and TGF-β1/Smad2/3 pathways, impair telomerase functions, alter mitochondrial, activate oxidative stress-related genes (ROS, superoxide dismutase, and catalase) and apoptosis-related genes (cytochrome-c, caspase-9, and caspase-3); they can directly bind to miRNAs silencing their expression (172). The complex scenario of epigenome is further modulated by exosomes, microvesicles and extracellular vesicles, membrane-derived vesicles of various diameter, released in extracellular microenvironment by a variety of cells such as B-cells, T-cells, mast cells, stem cells, dendritic cells, platelets, endothelial cells, epithelial cells. They convey proteins, lipids, miRNAs, long non-coding RNAs, DNA, enzymes and other factors responsible of cell-to-cell communication, epigenetic modifications. Composition of exosomes is influenced by microenvironment and determines the maintenance or the rupture of cellular homeostasis (173,174).

It should be remarked that not only smoke exposure can affect DNA methylation in both cancer and IPF. This issue is even more relevant for the cancer and the rare fibrotic cases that arise in non-smoker subjects. Indeed environmental or occupational exposure, pathogen infection and persistent tissue damage. For instance, polymorphisms in CYP1A1 and GTSM1, xenobiotic metabolizing enzymes, have been reported to be associated to higher risk of lung cancer development whereas polymorphisms in MLH1, a mismatch pair enzyme should play a role in the onset of the disease in never smokers. Moreover, polymorphisms in genes involved in inflammatory cascade such as those encoding for interleukin (IL)-10, TNF, IL1-RN and IL-6 have been reported to be associated to lung cancer risk independently from cigarette smoke exposure (175). Within respect to IPF, previous reports suggested that SNPs in Mucin 5B (MUC5B) promotor region (rs35705950) are associated with prognosis of IPF and this fact may be related to the reduction of immune defense mechanism of MUC5B (176,177). Growing evidence suggests that lung microbiome plays a relevant role in maintaining lung immune homeostasis and that its alteration and disruption might be related to cancer onset by acting on epigenetic level such as by causing DNA damage, genomic instability, and inducing higher sensitivity to carcinogens (178). Environment factors that can alter lung microbiota might promote, mainly through production of bacterial toxins and other pro-inflammatory factors, cancer onset and progression (179,180). A number of recent observations suggests a role for lung bacteria in IPF onset as well (181-184). First observation regarded the fact that bacteria (most often Streptococcus pneumoniae and Moraxella catarrhalis) are frequently isolated from BALF from IPF patients (185) and that patients enrolled in clinical trials have better outcomes in those arms encompassing treatment with antibiotics (186-188). Next generation analysis approaches more recently reported that changes in the lung microbiome are associated to IPF progression and acute phases (not in those patients treated with interferon (189); however, these data are too preliminary to define their potential predictive or prognostic role (190,191).


Non-invasive diagnostic and monitoring tools

Is there a role for liquid biopsy in IPF setting?

Liquid biopsy is a minimally invasive procedure that has been developed in molecular oncology. It allows the identification of circulating tumor-derived DNA (ct-DNA) that is shed from tumor cell in body fluids. Serial analysis of circulating tumor DNA (ctDNA) during treatment will provide a dynamic picture of molecular disease changes and could be used to monitor the emergence of secondary resistance and to identify heterogeneous subclonal populations developing during targeted treatments (192). This non-invasive sampling issue is overall simple to collect, although should present quantity and quality problems and representation bias. There is a strong rationale for application of this technique in early diagnosis and monitoring of IPF patients, although some key differences should be underlined. IPF landscape is enriched in neoplastic potential expressed in a context of complex genomic polyclonality and cellular heterogeneity. Smoking is strongly associated with IPF (193) and is a strong negative predictor for the occurrence of EGFR activating mutations in lung cancer according to previous reports (194). However, no somatic changes in coding sequences of driver known oncogenes. The latter observation, in therapeutic perspective, results in the absence of oncogenic addiction phenomenon. Thus, the oncogenic shock phenomenon cannot be exploited for therapeutic purposes in IPF. In cancer setting, the application of liquid biopsy: (I) ctDNA; (II) circulating tumor cells (CTC) for genetic analysis; (III) CTC. Interestingly, the level of circulating cell-free double-stranded DNA fragments (ccf-dsDNA) is increased in those IPF patients featuring rapid progression of the diseases if compared to stable IPF and health subjects. Moreover, the high expression of ccf-dsDNA is associated with that of amino acid, energy, and lipid metabolism pathways (195). Very recently, Pallante and colleagues (196) demonstrated the concordance between ccfDNA and genomic DNA by analyzing and detecting the rs35705950 polymorphism of MUC5B gene promoter, known to be involved in IPF onset (197). Overall, IPF is associated to increased tumor mutational burden (TMB) which, in turn, significantly contributes to the development of lung adenocarcinoma (198). However, data from TMB analysis from IPF-associated lung cancer are still controversial, being significantly higher than in lung adenocarcinoma alone (86,198). However, data from TMB analysis on lung cancer and concomitant ILDs are more controversial since some results reported that squamous cell carcinoma and adenocarcinoma with ILD do not have high TMB values (199). Due to the implication for therapy with IC inhibitors, since based on these data patients should not be addressed to immunotherapy (200), these preliminary observations deserve further validation data. Circulating cells have been studied to evaluate their possible role as predictive and prognostic markers. Elevated number of circulating fibrocytes, sorted by flow cytometry, is reported to be associated to higher mortality (201), rather than as validated marker of disease progression (202). Levels of circulating endothelial cells and endothelial progenitor cells have been found to be reduced in patients with IPF and treatment with nintedanib and pirfenidone further reduced their levels (203). Further validations are required regarding the role of circulating monocytes in prediction of disease progression (204).

Imaging

The diagnostic algorithm for suspected lung tumor in the IPF setting is still unclear. The most recent ATS/ERS/JRS/ALAT guidelines, updated in 2015, do not address this issue (205). Also, more “radiologically-oriented” guidelines such as those from the Fleischner society do not specifically include management suggestions tailored for IPF patients. Since those patients can be considered at high-risk of developing a lung tumor the suggested management will therefore rely mostly on surgical lung biopsy and resection. These approaches can be way too aggressive for an IPF patients. For these reasons a recent editorial suggests the following approach: high resolution computed tomography (HRCT) once a year in all patients with IPF. For patients with nodules less than 8 mm in diameter an HRCT every 3–6 months can be performed. If HRCT shows progression of the nodule, a PET-CT scan is recommended. For nodules with diameter of at least 8 mm, PET-CT scan is highly recommended. If PET indicates that a significant uptake is present, minimally invasive diagnostic procedures such as transthoracic needle biopsy or endoscopic approach are mandatory. If the patient is not suitable for biopsy, a multidisciplinary discussion is suggested (2). Even if nowadays early diagnosis of lung cancer mostly relies on HRCT and PET scans, which represent the cornerstone for timely therapeutic interventions, some new options may be available in the future. Magnetic Resonance (MR) and, especially, diffusion weighted imaging (DWI) has already shown that active lung inflammatory tissue in the IPF setting could be assessed effectively (206). It also has shown a capability of distinguishing between malignant and benign pathologies thanks to apparent diffusion coefficient (ADC) values (207). A promising tool may be IVIM (Intravoxel Incoherent Motion)-DWI which can also give information about perfusion. This may lead in the future to put aside data from PET-CT with those from MR. Another possible tool to optimize the management of IPF patients with a suspected lung cancer can be Radiomics. Radiomics is a quantitative approach to medical imaging, which aims, through mathematical extraction of the spatial distribution of signal intensities and pixel interrelationships, to quantify textural information by using analysis methods from the field of artificial intelligence. Radiomics has progressively gained attention for nodule characterization and, since no data are available in the IPF setting, more time is needed to distinguish the hope from the hype (208).


Rationale for immunotherapy in fibrotic lung

It is well known that inflammation plays a relevant pathogenic role in IPF even though anti-inflammatory drugs as steroids do not impact significantly on disease progression (209). This observation points out that the role on inflammatory reactions might not be a driver of IPF, or more properly, the complex IPF context requires a deeper characterization of the inflammatory pathways involved to identify effective targets. The inflammatory profile of IPF is characterized by type 2 inflammation (210,211) involving the interleukin (IL)-13 and IL-4, produced by T helper 2 (Th2) cells and type 2 innate lymphocytes; both are suggested to play a prominent role in fibrosis development (212). Type 2 immune cascade is known to drive pathogenic events in allergic asthma and several inhibitory molecules have reached the clinical use. Among them anti IL-13 monoclonal antibody (mAb) lebrikizumab has been recently tested in the randomized, multicenter, double-blind, placebo-controlled, parallel-group study NCT01872689 trial aimed at evaluating its efficacy and safety as monotherapy or with pirfenidone in IPF subjects. Although the pharmacodynamic biomarkers indicated a certain activity of lebrikizumab in association to the already known safety profile, lebrikizumab alone or in combination with pirfenidone showed no additional advantages since it was not able to improve functional parameters (213). Similar results have been reported using the anti-IL-13 mAb tralokinumab which safety profile resulted acceptable in absence of significant advantages (NCT01629667, NCT02036580) (214) and the study evaluating the mAb dectrekumab in IPF was discontinued in absence of significant results (215). These observations suggest that IL-13/type 2 immunity might not be the right target in IPF onset although a potential role of type 2-driven immune response is conceivable in acute exacerbation (AE) of disease. Growing evidence point out that many important fibrogenic steps should be orchestrated by both innate and adaptive immunity and that the innate response prevails (216) or, more properly, that the epithelial damage plays an important role in inducing immune system dysregulation which acts as critical driver for disease progression (217). This specific feature is a common denominator to cancer (218-220) and sustain a rationale for IC blockade therapeutic strategy. Immunotherapy has substantially changed the therapeutic strategies for cancers such as melanomas, lymphomas, and lung tumors. Unfortunately, only 20–50% of patients with advanced solid tumors respond to treatment. There is therefore a need for the development of methods to identify patients who are most likely to respond to immunotherapy. ICs are molecules located on the surface of cells that can send inhibitory stimuli to attenuate immune responses. Tumors express IC proteins on their cell surface to escape detection from the immune system. Thus, targeted inhibition towards these receptors enhances T cell response against the tumor. Tumor cells express checkpoint proteins on their surface to evade host immune response. Targeted inhibition towards these receptors enhances T cell response towards the tumor (221,222). Cytotoxic T-lymphocyte antigen 4 (CTLA-4), PD-1, and PD-L1 are key negative regulators of anti-tumor T cell reactivity. The development of IC inhibitors has revolutionized the treatment of a variety of cancers. Several studies have shown that pre-existing tumoral and peritumoral immune infiltration correlates with patient response to PD-1 and PD-L1 immunotherapy. Three distinct immune phenotypes have been described: immune-inflamed, immune-excluded, and immune-desert. Immune-inflamed tumors are characterized by dense, functional CD8 cell infiltration, increased interferon-γ signaling, expression of cell checkpoint markers (such as PD-L1), and a high mutational burden. These tumors tend to respond to immunotherapy. The detailed description of cancer microenvironment and sensitivity to IC inhibitors (ICIs) goes beyond the scope of this paper. It should be underlined that the cellular heterogeneity which characterizes IPF complicates data interpretation and can make elusive data interpretation when obtained from tissue homogenates. The recently completed study entitled “Immunopathologic Profiles and Blood Biomarkers in Patients With IPF” (NCT04187079) aimed at IPF tissue and blood profiling also investigating the cellular expression of ICs (namely PD-L1) in lung epithelial cells. It is known that PD1-PDL1 are expressed in IPF lymphocytes (223), AMs (224) and myofibroblasts (159) through IHC stain and RNA sequencing. The PD-1/PD-L1 axis is likely to contribute to lung fibrogenesis (225) anti PD-L1 Abs significantly reduces pulmonary fibrosis (226). PD-1 expression on CD4+ T cells is known to lead to activation of Signal Transducer and Activator of Transcription (STAT) 3 which, in turn, induces IL-17A and TGF-β expression (227). Ex vivo blockade of the PD-1/PD-L1 axis is associated to STAT3-mediated IL-17A and TGF-β production by CD4+ T cells (223). PD-L1 inhibitors should not be used in conjunction with mesenchymal stromal cell (MSC) therapy (228). CTLA-4 is strongly overexpressed in IPF CD4- and CD28 null IPF lymphocytes if compared to health cells and anti-CTLA-4 antibody treatment was shown to aggravate fibrosis in a humanized IPF model (173,225,229). Notably, a high level of hypoxia and immune activity is associated to worst prognosis in IPF, whereas those patients featuring high level of oxygen and low immunogenic reactions the best prognosis (230). These preliminary findings point out a novel strategy to effectively select patients for immunotherapy. Overall, the expression of IC molecules in lung fibrotic tissues sustain a rationale for a deeper investigation of their pathogenic role and as actionable targets. Durable responses to nivolumab in a lung cancer patient withs idiopathic pulmonary fibrosis (231-233). This observation suggests that in those cases, ICI treatment should be considered a potentially effective option even though the occurrence of ILD has been identified as a rare but potentially severe event induced by immunotherapy (234). In this perspective the already reported activation of the MET oncogene in IPF should become relevant (9). IPF resembles cancer in two critical MET-associated behaviors: invasive phenotype and pro-coagulant status. In cancer, MET activation occurs as a late event, consequently to transcriptional up-regulation driven by unfavorable microenvironmental conditions MET (mainly hypoxia) amplified cancer clones are selected under therapeutic pressure in a context of molecularly heterogeneous lesions exposed to targeted therapies or radiotherapy (235,236). Thus, this oncogenic expedient (69) can be exploited for therapeutic purposes in IPF. Moreover, it has been already reported that, in lung cancer mutations occurring in several oncogenes among which MET, modulate tumor microenvironment and a positive correlation between MET amplification and PDL1 overexpression has been already reported (237,238). Thus, in a context-specific regulation of its expression, MET might become a functional marker of IPF and an actionable target, positively associated to response to ICIs (Figure 2).

Figure 2 Functional annotation of the MET oncogene as an actionable target of IPF. MET-mediated events in IPF rely on qualitative differences among physiological signals. No driver genetic lesions, causally implicated in the disease can be clearly demonstrated (“Fibrogenic Expedience”). The MET blockage falls among those therapeutic strategies aimed at impairing the “aberrant recapitulation of developmental programs”. The hypoxia-induced MET up-regulation might cooperate in triggering IPF regenerative/reparative processes. HGF, hepatocyte growth factor; IPF, idiopathic pulmonary fibrosis.

Radiotherapy in lung cancer with IPF

Radiation induced lung injury (RILI) represents one of the major issues in the setting of thoracic radiotherapy (239); it generally corresponds to radiation-induced pneumonitis, an intermediate phase injury after exposure to ionising irradiation, which in most cases paves the way for the development of late fibrosis. Both pneumonitis and fibrosis are dose-limiting toxicities of great concern to the radiation oncologist, especially in the scenario of a combined chemoradiotherapy approach or in high dose hypo-fractionated radiotherapy.

Thoracic radiotherapy plays a role in enhancing the occurrence of AEs in IPF, even when baseline symptoms are trivial. Pre-existing IPF is a well-known risk factor for pulmonary toxicity after ionising irradiation (240); previous reports have shown that it can raise the risk of severe and even life-threatening pneumonitis, whose rates in such patients range between 6.3% and 18.2%, in relation to different radiotherapy techniques (241). Nevertheless, IPF does not constitute an absolute contraindication for thoracic radiotherapy, even if European Organisation for Research and Treatment of Cancer (EORTC) guidelines suggest avoiding irradiating lung cancer patients with IPF (242). While some encouraging data come from some preliminary experiences with proton therapy (241), the decision to offer radiotherapy to these patients should be made after a multidisciplinary approach in which patient’s individual risk is evaluated, especially in terms of his/her clinical status, disease specific survival and therapeutic index.

Surgery in lung cancer with IPF

Lung resection plays a role in the treatment of patients affected by IPF with resectable NSCLC. However, in this scenario, two major issues influence significantly the surgical procedure and the survival outcomes: the high risk of postoperative AEs of IPF in the short-term, and the death due to cancer in the long-term (243). Surgery is a defined risk factor for AE in IPF patients (16) and since its incidence in this group of patients is estimated to be approximately 9.3% (244) and no preventive measure is known, it is crucial to carefully select the patients to properly refer treat the patients. In a study by T. Sato and colleagues, a simple scoring system to identify high risk patients for AE was derived in order to help in the decision-making process for surgery selection and predict the patients requiring intensive observation postoperatively (245). Among the surgical procedures of lung resection, wedge resection is associated to the lowest risks of postoperative AE compared to segmentectomy, lobectomy, bilobectomy and pneumonectomy, since AE risk increases according to the resected lung parenchyma volume (244). Death due to cancer is the major concern in the long-term: it represents the main cause of death in lung cancer patients affected by IPF, mostly attributable to cancer recurrence after surgery. Contrary to AE risk, lobectomy shows better results for death due to cancer in patients with stage IA, while wedge resection and segmentectomy were associated to poor outcomes (244).

Lung resection in patients with IPF is challenging but required for several patients. The choice of surgical procedure must be tailored based on several criteria, such as pulmonary function, cancer stage and recurrence risk, postoperative AE risk, and the natural course of IPF.

Percutaneous thermal ablation in lung cancer with IPF

Alternative treatments such as radiofrequency ablation could be of therapeutic benefit with relatively minimal complications, particularly in patients who are not fit enough for surgical interventions. On the other hand, the risk of severe complications with stereotactic body radiation therapy (SBRT) when treating patients with IPF is widely recognized. For these reasons, in the IPF setting, thermal ablation procedure, generally performed under CT guidance, can be a viable therapeutic option. Radiofrequency ablation and SBRT in patients with inoperable stage I NSCLC had similar overall survival rates while local progression rates were higher for radiofrequency ablation (246). No specific comparison had been performed over different types of ablative procedures (radiofrequency, microwave, cryoablation) while the largest experience came from radiofrequency ablations. Every technique has its own advantages and disadvantages (e.g., cryoablation is safer near the airways while microwaves are powerful and faster than radiofrequency) that can be a further strength of minimally invasive procedure. At the same time this heterogeneity creates severe difficulties in obtaining large databases of procedures outcome and procedures performances. For these reasons a multidisciplinary advice and centre preferences and expertise are fundamental for alternative treatment choice and management.

Advanced cell therapies

Based on the U.S. Food and Drug Administration (FDA) cell therapy includes cellular products for immunotherapies, cancer vaccines, and other types of both autologous and allogeneic cells for certain therapeutic indications (www.fda.gov). According to this definition, the most clinical implications regard clearly cancer, but the recent progresses in the knowledge of molecular mechanisms responsible of IPF with the evidence of biologic similarities between IPF and malignant proliferation give a strong rationale for the investigation and development of cell therapeutic strategies and tissue engineering to impair fibrotic damages. MSCs feature the pluripotent capacity of and their ability to differentiate to important lineages that can modulate on immunity, impair inflammatory reactions, and promote epithelial tissue repair (247); the clinical application of MSC therapy has been shown to be feasible and safe in humans with IPF (www.clinicaltrial.gov) and several data have been already published (248-252). A schematic representation of the application of MSCs in lung fibrosis is reported in Figure 3. MSCs and fibrocytes can be generated from the bone marrow and home to the injured lungs in response to several secreted chemokines and growth factor receptors (253,254). Lung resident MSCs (LR-MSCs) and mainly myofibroblasts precursors have been detected as well (255,256). Allogenic MSCs derived from unrelated donors seems to be safe as homologous obtained cells when infused in patients carrying mild-moderate disease (257). MSCs communicate with their surrounding microenvironment and in particular, the alveolar niche cells promote alveolar epithelial progenitors to regenerate the damaged epithelium (258). Different strategies have been explored to the development of advanced cellular therapy in IPF. The MSCs quiescence or dormancy is a key feature of stem cells; thus, a potential target of therapeutic intervention is that of inducing stem cells into the cell cycle to start differentiation. In this perspective, the Wnt/β-catenin signalling is known to be implicated IPF pathogenesis since its activation inhibits MSCs to differentiate into epithelium. The pharmacological inhibition of the Wnt cascade might be exploited to impair myofibroblasts differentiation and proliferation (259,260). Moreover, MSCs in IPF become rapidly senescent (261-263) and strategies to ameliorate this process are beneficial in reducing disease progression. miRNAs are involved in mediating MSC senescence by modulating the expression of several pathways. Very recently, miR-200 family members (miR-200b-3p and miR-200c-3p) and miR-199a-5p has been reported to regulate MSC senescence in IPF patients with by acting on the Sirtuin 1/AMP-activated protein kinase signalling cascade; thus, they emerge as a novel potential target to rejuvenate IPF-MSCs and to prevent fibrotic damages and to restore proper differentiation (264,265). MSCs display immunomodulatory properties and can secrete anti-fibrotic factors (Figure 4). It has been reported that lung resident MSC can be in IPF lungs and their secretome is able to damage fibroblast proliferation while promoting enhanced epithelial wound repair via several growth factors, among which hepatocyte growth factor (HGF) (266,267). Interestingly, inhaled lung spheroid cell-secretome (LSC-Sec) and exosomes (LSC-Exo) have been shown to attenuate bleomycin and silica-induced fibrosis in experimental models in a more effective manner that those derived from resident MSCs. They seem block EMT acting on WNT/beta catenin, Rho/Rock and TGFbeta 1/SMAD pathways (268). Other therapeutic targets in the context of cell therapy in IPF are represented by MSC-derived growth factors, as HGF, which play relevant roles in the repair of alveolar epithelial cells, actively contrasts myofibroblasts activation and the abnormal deposition of ECM (269). Growing evidence indicates that the changes in ECM composition and mechanical properties which characterize IPF can be exploited for therapeutic purposes. Synthetic materials as polyacrylamide, hydrogels, highly cross-linked polymer networks as well as liposomes, polymeric nanoparticles represent engineered platforms which can be decorated with cell-adhesive ligands, signalling factors, drugs which can modulate lung remodelling (270-272).

Figure 3 Stem cells and their application in lung fibrosis. Stem cells can be classified into embryonal stem cells (ESC), adult stem cells (ASC) and induced pluripotent stem cells (IPSC) according to their origin. ESCs derive from embryo blastocysts, ASC can be isolated from various tissues, such as bone marrow, lung, adipose tissue, umbilical cord blood, umbilical cord tissue and amniotic fluid. IPSC are obtained from somatic cells using reprogramming factors (OCT3/4, SOX2, C-MYC, KLF4), responsible for re-programming to pluripotency. Stem cells can be administrated intravenously, intratracheally or intraperitoneally. They migrate to the injured sites of the lungs where they differentiate in alveolar type II cells and exert anti-inflammatory, antifibrotic and immunomodulant actions. IPF, idiopathic pulmonary fibrosis.
Figure 4 Stem cells and secretoma. Lung spheroid cells are round aggregates composed by stem cells and stromal cells. They produce a complex of proteins and growth factors, complexify named as secretoma, also including exosomes. Lung spheroid cell-secretome (LSC-Sec) and exosomes (LSC-Exo) reproduce a regenerative microenvironment and promote differentiation of stem cells towards epithelial phenotypes. EMT, epithelial mesenchymal transition.

Concluding remarks

IPF identifies a specific entity characterized by chronic, progressive fibrosing interstitial pneumonia of unknown cause, still lacking effective therapies. Growing evidence points out that aberrant proliferative events in IPF recall malignant transformation given a specific temporal and cellular heterogeneity. To look at IPF through cancer glass can help in stratifying and addressing patients to personalized approaches as well as in analyzing the mechanisms of abnormal cell/matrix interactions which characterize the disease. Moreover, the advances in cancer immunotherapy and the improvement in imaging and radiotherapy techniques open the way for treatment of those patients carrying both lung cancer and IPF that till now have represented a sort of therapeutically orphan population. Translational research is a source of continuous innovation in medicine more particularly for clinical research on new treatment modalities in IPF patients and will contribute to improve mechanistic explanation of disease onset and progression. To reach this goal, the real efficiency of next future studies and trials will depend on the integration of proper sample collection, gene expression analysis and functional and bio-informatic annotation as well as on the coordination of multidisciplinary know how and technical platforms.


Acknowledgments

GMS would like to thank Elena Morganti for fruitful discussion and critical reading and Benedetta Marchelli for the continuous and enthusiastic support.

Funding: This work was supported by Ricerca Corrente (No. #08050 to GMS) from IRCCS Policlinico San Matteo.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-21-880/rc

Peer Review File: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-21-880/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-21-880/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Ballester B, Milara J, Cortijo J. Idiopathic Pulmonary Fibrosis and Lung Cancer: Mechanisms and Molecular Targets. Int J Mol Sci 2019;20:593. [Crossref] [PubMed]
  2. Tzouvelekis A, Spagnolo P, Bonella F, et al. Patients with IPF and lung cancer: diagnosis and management. Lancet Respir Med 2018;6:86-8. [Crossref] [PubMed]
  3. Kim HC, Lee S, Song JW. Impact of idiopathic pulmonary fibrosis on clinical outcomes of lung cancer patients. Sci Rep 2021;11:8312. [Crossref] [PubMed]
  4. Han S, Lee YJ, Park JS, et al. Prognosis of non-small-cell lung cancer in patients with idiopathic pulmonary fibrosis. Sci Rep 2019;9:12561. [Crossref] [PubMed]
  5. Tomassetti S, Gurioli C, Ryu JH, et al. The impact of lung cancer on survival of idiopathic pulmonary fibrosis. Chest 2015;147:157-64. [Crossref] [PubMed]
  6. Tzouvelekis A, Gomatou G, Bouros E, et al. Common Pathogenic Mechanisms Between Idiopathic Pulmonary Fibrosis and Lung Cancer. Chest 2019;156:383-91. [Crossref] [PubMed]
  7. Vancheri C, Failla M, Crimi N, et al. Idiopathic pulmonary fibrosis: a disease with similarities and links to cancer biology. Eur Respir J 2010;35:496-504. [Crossref] [PubMed]
  8. Lettieri S, Oggionni T, Lancia A, et al. Immune Stroma in Lung Cancer and Idiopathic Pulmonary Fibrosis: A Common Biologic Landscape? Int J Mol Sci 2021;22:2882. [Crossref] [PubMed]
  9. Stella GM, Gentile A, Balderacchi A, et al. Ockham's razor for the MET-driven invasive growth linking idiopathic pulmonary fibrosis and cancer. J Transl Med. 2016;14:256. Erratum in: J Transl Med 2017;15:194. [Crossref] [PubMed]
  10. Stella GM, Inghilleri S, Pignochino Y, et al. Activation of oncogenic pathways in idiopathic pulmonary fibrosis. Transl Oncol 2014;7:650-5. [Crossref] [PubMed]
  11. Archontogeorgis K, Steiropoulos P, Tzouvelekis A, et al. Lung cancer and interstitial lung diseases: a systematic review. Pulm Med 2012;2012:315918. [Crossref] [PubMed]
  12. Bouros D, Hatzakis K, Labrakis H, et al. Association of malignancy with diseases causing interstitial pulmonary changes. Chest 2002;121:1278-89. [Crossref] [PubMed]
  13. Antoniou KM, Tomassetti S, Tsitoura E, et al. Idiopathic pulmonary fibrosis and lung cancer: a clinical and pathogenesis update. Curr Opin Pulm Med 2015;21:626-33. [Crossref] [PubMed]
  14. Agabiti N, Porretta MA, Bauleo L, et al. Idiopathic Pulmonary Fibrosis (IPF) incidence and prevalence in Italy. Sarcoidosis Vasc Diffuse Lung Dis 2014;31:191-7. [PubMed]
  15. Enomoto Y, Inui N, Yoshimura K, et al. Lung cancer development in patients with connective tissue disease-related interstitial lung disease: A retrospective observational study. Medicine (Baltimore) 2016;95:e5716. [Crossref] [PubMed]
  16. Kreuter M, Ehlers-Tenenbaum S, Schaaf M, et al. Treatment and outcome of lung cancer in idiopathic interstitial pneumonias. Sarcoidosis Vasc Diffuse Lung Dis 2015;31:266-74. [PubMed]
  17. Le Jeune I, Gribbin J, West J, et al. The incidence of cancer in patients with idiopathic pulmonary fibrosis and sarcoidosis in the UK. Respir Med 2007;101:2534-40. [Crossref] [PubMed]
  18. Karampitsakos T, Tzilas V, Tringidou R, et al. Lung cancer in patients with idiopathic pulmonary fibrosis. Pulm Pharmacol Ther 2017;45:1-10. [Crossref] [PubMed]
  19. Park J, Kim DS, Shim TS, et al. Lung cancer in patients with idiopathic pulmonary fibrosis. Eur Respir J 2001;17:1216-9. [Crossref] [PubMed]
  20. Nagai A, Chiyotani A, Nakadate T, et al. Lung cancer in patients with idiopathic pulmonary fibrosis. Tohoku J Exp Med 1992;167:231-7. [Crossref] [PubMed]
  21. Ozawa Y, Suda T, Naito T, et al. Cumulative incidence of and predictive factors for lung cancer in IPF. Respirology 2009;14:723-8. [Crossref] [PubMed]
  22. Yoo H, Jeong BH, Chung MJ, et al. Risk factors and clinical characteristics of lung cancer in idiopathic pulmonary fibrosis: a retrospective cohort study. BMC Pulm Med 2019;19:149. [Crossref] [PubMed]
  23. Aubry MC, Myers JL, Douglas WW, et al. Primary pulmonary carcinoma in patients with idiopathic pulmonary fibrosis. Mayo Clin Proc 2002;77:763-70. [Crossref] [PubMed]
  24. Samet JM. Does idiopathic pulmonary fibrosis increase lung cancer risk? Am J Respir Crit Care Med 2000;161:1-2. [Crossref] [PubMed]
  25. Ohtsuka Y, Ukita H, Masaki Y, et al. A prospective study of lung cancer in cases of idiopathic interstitial pneumonia (IIP). Nihon Kyobu Shikkan Gakkai Zasshi 1991;29:560-5. [PubMed]
  26. Munakata M, Asakawa M, Hamma Y, et al. Present status of idiopathic interstitial pneumonia--from epidemiology to etiology. Nihon Kyobu Shikkan Gakkai Zasshi 1994;32:187-92. [PubMed]
  27. Matsushita H, Tanaka S, Saiki Y, et al. Lung cancer associated with usual interstitial pneumonia. Pathol Int 1995;45:925-32. [Crossref] [PubMed]
  28. Hironaka M, Fukayama M. Pulmonary fibrosis and lung carcinoma: a comparative study of metaplastic epithelia in honeycombed areas of usual interstitial pneumonia with or without lung carcinoma. Pathol Int 1999;49:1060-6. [Crossref] [PubMed]
  29. Hata A, Nakajima T, Matsusaka K, et al. Genetic alterations in squamous cell lung cancer associated with idiopathic pulmonary fibrosis. Int J Cancer 2021;148:3008-18. [Crossref] [PubMed]
  30. Königshoff M. Lung cancer in pulmonary fibrosis: tales of epithelial cell plasticity. Respiration 2011;81:353-8. [Crossref] [PubMed]
  31. Raghu G, Nyberg F, Morgan G. The epidemiology of interstitial lung disease and its association with lung cancer. Br J Cancer 2004;91:S3-10. [Crossref] [PubMed]
  32. Bouros D. Pirfenidone for idiopathic pulmonary fibrosis. Lancet 2011;377:1727-9. [Crossref] [PubMed]
  33. Seifirad S. Pirfenidone: A novel hypothetical treatment for COVID-19. Med Hypotheses 2020;144:110005. [Crossref] [PubMed]
  34. Wollin L, Wex E, Pautsch A, et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J 2015;45:1434-45. [Crossref] [PubMed]
  35. Knüppel L, Ishikawa Y, Aichler M, et al. A Novel Antifibrotic Mechanism of Nintedanib and Pirfenidone. Inhibition of Collagen Fibril Assembly. Am J Respir Cell Mol Biol 2017;57:77-90. [Crossref] [PubMed]
  36. Saiki A, Mizobuchi T, Nagato K, et al. Uniportal video-assisted thoracic surgery and perioperative pirfenidone for lung cancer and idiopathic pulmonary fibrosis: a case report. J Int Med Res 2021;49:3000605211016998. [Crossref] [PubMed]
  37. Iwata T, Yoshino I, Yoshida S, et al. A phase II trial evaluating the efficacy and safety of perioperative pirfenidone for prevention of acute exacerbation of idiopathic pulmonary fibrosis in lung cancer patients undergoing pulmonary resection: West Japan Oncology Group 6711 L (PEOPLE Study). Respir Res 2016;17:90. [Crossref] [PubMed]
  38. Kanayama M, Mori M, Matsumiya H, et al. Perioperative pirfenidone treatment for lung cancer patients with idiopathic pulmonary fibrosis. Surg Today 2020;50:469-74. [Crossref] [PubMed]
  39. Sekihara K, Aokage K, Miyoshi T, et al. Perioperative pirfenidone treatment as prophylaxis against acute exacerbation of idiopathic pulmonary fibrosis: a single-center analysis. Surg Today 2020;50:905-11. [Crossref] [PubMed]
  40. Iwata T, Yoshida S, Nagato K, et al. Experience with perioperative pirfenidone for lung cancer surgery in patients with idiopathic pulmonary fibrosis. Surg Today 2015;45:1263-70. [Crossref] [PubMed]
  41. Miura Y, Saito T, Tanaka T, et al. Reduced incidence of lung cancer in patients with idiopathic pulmonary fibrosis treated with pirfenidone. Respir Investig 2018;56:72-9. [Crossref] [PubMed]
  42. Fujiwara A, Funaki S, Fukui E, et al. Effects of pirfenidone targeting the tumor microenvironment and tumor-stroma interaction as a novel treatment for non-small cell lung cancer. Sci Rep 2020;10:10900. [Crossref] [PubMed]
  43. Ping Q, Yan R, Cheng X, et al. Cancer-associated fibroblasts: overview, progress, challenges, and directions. Cancer Gene Ther 2021;28:984-99. [Crossref] [PubMed]
  44. Hao Y, Zhang L, He J, et al. Functional investigation of NCI-H460-inducible myofibroblasts on the chemoresistance to VP-16 with a microfluidic 3D co-culture device. PLoS One 2013;8:e61754. [Crossref] [PubMed]
  45. Chen X, Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discov 2019;18:99-115. [Crossref] [PubMed]
  46. Fujiwara A, Shintani Y, Funaki S, et al. Pirfenidone plays a biphasic role in inhibition of epithelial-mesenchymal transition in non-small cell lung cancer. Lung Cancer 2017;106:8-16. [Crossref] [PubMed]
  47. Yamamoto Y, Yano Y, Kuge T, et al. Safety and effectiveness of pirfenidone combined with carboplatin-based chemotherapy in patients with idiopathic pulmonary fibrosis and non-small cell lung cancer: A retrospective cohort study. Thorac Cancer 2020;11:3317-25. [Crossref] [PubMed]
  48. Reck M, Kaiser R, Mellemgaard A, et al. Docetaxel plus nintedanib versus docetaxel plus placebo in patients with previously treated non-small-cell lung cancer (LUME-Lung 1): a phase 3, double-blind, randomised controlled trial. Lancet Oncol 2014;15:143-55. [Crossref] [PubMed]
  49. Kato R, Haratani K, Hayashi H, et al. Nintedanib promotes antitumour immunity and shows antitumour activity in combination with PD-1 blockade in mice: potential role of cancer-associated fibroblasts. Br J Cancer 2021;124:914-24. [Crossref] [PubMed]
  50. Kai Y, Matsuda M, Fukuoka A, et al. Remarkable response of non-small cell lung cancer to nintedanib treatment in a patient with idiopathic pulmonary fibrosis. Thorac Cancer 2021;12:1457-60. [Crossref] [PubMed]
  51. Fukunaga K, Yokoe S, Kawashima S, et al. Nintedanib prevented fibrosis progression and lung cancer growth in idiopathic pulmonary fibrosis. Respirol Case Rep 2018;6:e00363. [Crossref] [PubMed]
  52. Shiratori T, Tanaka H, Tabe C, et al. Effect of nintedanib on non-small cell lung cancer in a patient with idiopathic pulmonary fibrosis: A case report and literature review. Thorac Cancer 2020;11:1720-3. [Crossref] [PubMed]
  53. Fang W, Huang Y, Gan J, et al. Nintedanib Effect in Osimertinib-Induced Interstitial Pneumonia. J Thorac Oncol 2020;15:e34-5. [Crossref] [PubMed]
  54. Yamakawa H, Oba T, Ohta H, et al. Nintedanib allows retreatment with atezolizumab of combined non-small cell lung cancer/idiopathic pulmonary fibrosis after atezolizumab-induced pneumonitis: a case report. BMC Pulm Med 2019;19:156. [Crossref] [PubMed]
  55. Xie XH, Deng HY, Lin XQ, et al. Case Report: Nintedanib for Pembrolizumab-Related Pneumonitis in a Patient With Non-Small Cell Lung Cancer. Front Oncol 2021;11:673877. [Crossref] [PubMed]
  56. Son JY, Kim SY, Cho SH, et al. TGF-β1 T869C polymorphism may affect susceptibility to idiopathic pulmonary fibrosis and disease severity. Lung 2013;191:199-205. [Crossref] [PubMed]
  57. Xaubet A, Marin-Arguedas A, Lario S, et al. Transforming growth factor-beta1 gene polymorphisms are associated with disease progression in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2003;168:431-5. [Crossref] [PubMed]
  58. Whyte M, Hubbard R, Meliconi R, et al. Increased risk of fibrosing alveolitis associated with interleukin-1 receptor antagonist and tumor necrosis factor-alpha gene polymorphisms. Am J Respir Crit Care Med 2000;162:755-8. [Crossref] [PubMed]
  59. Korthagen NM, van Moorsel CH, Kazemier KM, et al. IL1RN genetic variations and risk of IPF: a meta-analysis and mRNA expression study. Immunogenetics 2012;64:371-7. [Crossref] [PubMed]
  60. Ahn MH, Park BL, Lee SH, et al. A promoter SNP rs4073T>A in the common allele of the interleukin 8 gene is associated with the development of idiopathic pulmonary fibrosis via the IL-8 protein enhancing mode. Respir Res 2011;12:73. [Crossref] [PubMed]
  61. O'Dwyer DN, Armstrong ME, Trujillo G, et al. The Toll-like receptor 3 L412F polymorphism and disease progression in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2013;188:1442-50. [Crossref] [PubMed]
  62. Xue J, Gochuico BR, Alawad AS, et al. The HLA class II Allele DRB1*1501 is over-represented in patients with idiopathic pulmonary fibrosis. PLoS One 2011;6:e14715. [Crossref] [PubMed]
  63. Korthagen NM, van Moorsel CH, Barlo NP, et al. Association between variations in cell cycle genes and idiopathic pulmonary fibrosis. PLoS One 2012;7:e30442. [Crossref] [PubMed]
  64. Diaz de Leon A, Cronkhite JT, Katzenstein AL, et al. Telomere lengths, pulmonary fibrosis and telomerase (TERT) mutations. PLoS One 2010;5:e10680. [Crossref] [PubMed]
  65. Stella GM, Balestro E. Idiopathic pulmonary fibrosis landscapes: looking glass from pathology to therapy. Minerva Med 2015;106:17-24. [PubMed]
  66. Garcia CK. Idiopathic pulmonary fibrosis: update on genetic discoveries. Proc Am Thorac Soc 2011;8:158-62. [Crossref] [PubMed]
  67. Kropski JA, Lawson WE, Young LR, et al. Genetic studies provide clues on the pathogenesis of idiopathic pulmonary fibrosis. Dis Model Mech 2013;6:9-17. [Crossref] [PubMed]
  68. Stella GM, Balestro E, Lacedonia D, et al. Telomeropathies: an emerging spectrum of disorders with important implications for patients with interstitial lung disease. Minerva Med 2016;107:9-14. [PubMed]
  69. Comoglio PM, Giordano S, Trusolino L. Drug development of MET inhibitors: targeting oncogene addiction and expedience. Nat Rev Drug Discov 2008;7:504-16. [Crossref] [PubMed]
  70. Otsubo K, Iwama E, Ijichi K, et al. Paired genetic analysis by next-generation sequencing of lung cancer and associated idiopathic pulmonary fibrosis. Cancer Sci 2020;111:2482-7. [Crossref] [PubMed]
  71. Chen YC, Hunter DJ. Molecular epidemiology of cancer. CA Cancer J Clin 2005;55:45-54; quiz 57. [Crossref] [PubMed]
  72. Olson AL, Gifford AH, Inase N, et al. The epidemiology of idiopathic pulmonary fibrosis and interstitial lung diseases at risk of a progressive-fibrosing phenotype. Eur Respir Rev 2018;27:180077. [Crossref] [PubMed]
  73. Boggaram V. Thyroid transcription factor-1 (TTF-1/Nkx2.1/TITF1) gene regulation in the lung. Clin Sci (Lond) 2009;116:27-35. [Crossref] [PubMed]
  74. Stella GM, Kolling S, Benvenuti S, et al. Lung-Seeking Metastases. Cancers (Basel) 2019;11:1010. [Crossref] [PubMed]
  75. Shanzhi W, Yiping H, Ling H, et al. The relationship between TTF-1 expression and EGFR mutations in lung adenocarcinomas. PLoS One 2014;9:e95479. [Crossref] [PubMed]
  76. Tanaka H, Yanagisawa K, Shinjo K, et al. Lineage-specific dependency of lung adenocarcinomas on the lung development regulator TTF-1. Cancer Res 2007;67:6007-11. [Crossref] [PubMed]
  77. Kendall J, Liu Q, Bakleh A, et al. Oncogenic cooperation and coamplification of developmental transcription factor genes in lung cancer. Proc Natl Acad Sci U S A 2007;104:16663-8. [Crossref] [PubMed]
  78. Kwei KA, Kim YH, Girard L, et al. Genomic profiling identifies TITF1 as a lineage-specific oncogene amplified in lung cancer. Oncogene 2008;27:3635-40. [Crossref] [PubMed]
  79. Takeuchi T, Tomida S, Yatabe Y, et al. Expression profile-defined classification of lung adenocarcinoma shows close relationship with underlying major genetic changes and clinicopathologic behaviors. J Clin Oncol 2006;24:1679-88. [Crossref] [PubMed]
  80. Yatabe Y, Kosaka T, Takahashi T, et al. EGFR mutation is specific for terminal respiratory unit type adenocarcinoma. Am J Surg Pathol 2005;29:633-9. [Crossref] [PubMed]
  81. Yamaguchi T, Hosono Y, Yanagisawa K, et al. NKX2-1/TTF-1: an enigmatic oncogene that functions as a double-edged sword for cancer cell survival and progression. Cancer Cell 2013;23:718-23. [Crossref] [PubMed]
  82. Bai XY, Shen H. Mutational analysis of thyroid transcription factor-1 gene (TTF-1) in lung carcinomas. In Vitro Cell Dev Biol Anim 2008;44:17-25. [Crossref] [PubMed]
  83. Maeda Y, Tsuchiya T, Hao H, et al. Kras(G12D) and Nkx2-1 haploinsufficiency induce mucinous adenocarcinoma of the lung. J Clin Invest 2012;122:4388-400. [Crossref] [PubMed]
  84. Cancer Genome Atlas Research Network. Integrated genomic characterization of endometrial carcinoma. Nature 2013;497:67-73. [Crossref] [PubMed]
  85. Qi J, Rice SJ, Salzberg AC, et al. MiR-365 regulates lung cancer and developmental gene thyroid transcription factor 1. Cell Cycle 2012;11:177-86. [Crossref] [PubMed]
  86. Hwang JA, Kim D, Chun SM, et al. Genomic profiles of lung cancer associated with idiopathic pulmonary fibrosis. J Pathol 2018;244:25-35. [Crossref] [PubMed]
  87. Wang Y, Kuan PJ, Xing C, et al. Genetic defects in surfactant protein A2 are associated with pulmonary fibrosis and lung cancer. Am J Hum Genet 2009;84:52-9. [Crossref] [PubMed]
  88. Nathan N, Giraud V, Picard C, et al. Germline SFTPA1 mutation in familial idiopathic interstitial pneumonia and lung cancer. Hum Mol Genet 2016;25:1457-67. [Crossref] [PubMed]
  89. Selman M, Lin HM, Montaño M, et al. Surfactant protein A and B genetic variants predispose to idiopathic pulmonary fibrosis. Hum Genet 2003;113:542-50. [Crossref] [PubMed]
  90. Nogee LM, Dunbar AE 3rd, Wert SE, et al. A mutation in the surfactant protein C gene associated with familial interstitial lung disease. N Engl J Med 2001;344:573-9. [Crossref] [PubMed]
  91. Thomas AQ, Lane K, Phillips J 3rd, et al. Heterozygosity for a surfactant protein C gene mutation associated with usual interstitial pneumonitis and cellular nonspecific interstitial pneumonitis in one kindred. Am J Respir Crit Care Med 2002;165:1322-8. [Crossref] [PubMed]
  92. Blackwell TS. Lung injury and fibrosis induced by a mutant form of surfactant protein C. J Clin Invest 2018;128:3745-6. [Crossref] [PubMed]
  93. Gower WA, Nogee LM. Candidate gene analysis of the surfactant protein D gene in pediatric diffuse lung disease. J Pediatr 2013;163:1778-80. [Crossref] [PubMed]
  94. Mohammad IS, He W, Yin L. Understanding of human ATP binding cassette superfamily and novel multidrug resistance modulators to overcome MDR. Biomed Pharmacother 2018;100:335-48. [Crossref] [PubMed]
  95. Nobili S, Lapucci A, Landini I, et al. Role of ATP-binding cassette transporters in cancer initiation and progression. Semin Cancer Biol 2020;60:72-95. [Crossref] [PubMed]
  96. Leng D, Yi J, Xiang M, et al. Identification of common signatures in idiopathic pulmonary fibrosis and lung cancer using gene expression modelling. BMC Cancer 2020;20:986. [Crossref] [PubMed]
  97. Berger J, Moller DE. The mechanisms of action of PPARs. Annu Rev Med 2002;53:409-35. [Crossref] [PubMed]
  98. Berger J, Wagner JA. Physiological and therapeutic roles of peroxisome proliferator-activated receptors. Diabetes Technol Ther 2002;4:163-74. [Crossref] [PubMed]
  99. Schoonjans K, Staels B, Auwerx J. The peroxisome proliferator activated receptors (PPARS) and their effects on lipid metabolism and adipocyte differentiation. Biochim Biophys Acta 1996;1302:93-109. [Crossref] [PubMed]
  100. Milam JE, Keshamouni VG, Phan SH, et al. PPAR-gamma agonists inhibit profibrotic phenotypes in human lung fibroblasts and bleomycin-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2008;294:L891-901. [Crossref] [PubMed]
  101. Wang X, Chen Y, Lv L, et al. Silencing CD36 gene expression results in the inhibition of latent-TGF-beta1 activation and suppression of silica-induced lung fibrosis in the rat. Respir Res 2009;10:36. [Crossref] [PubMed]
  102. AyaubEAPoliSNgJSingle Cell RNA-seq and Mass Cytometry Reveals a Novel and a Targetable Population of Macrophages in Idiopathic Pulmonary FibrosisbioRxiv 2021.01.04.425268. 10.1101/2021.01.04.425268
  103. Christiaens V, Van Hul M, Lijnen HR, et al. CD36 promotes adipocyte differentiation and adipogenesis. Biochim Biophys Acta 2012;1820:949-56. [Crossref] [PubMed]
  104. Wang X, Lv L, Chen Y, et al. A CD36 synthetic peptide inhibits silica-induced lung fibrosis in the mice. Toxicol Ind Health 2010;26:47-53. [Crossref] [PubMed]
  105. Tanase C, Gheorghisan-Galateanu AA, Popescu ID, et al. CD36 and CD97 in Pancreatic Cancer versus Other Malignancies. Int J Mol Sci 2020;21:5656. [Crossref] [PubMed]
  106. Epstein Shochet G, Bardenstein-Wald B, McElroy M, et al. Hypoxia Inducible Factor 1A Supports a Pro-Fibrotic Phenotype Loop in Idiopathic Pulmonary Fibrosis. Int J Mol Sci 2021;22:3331. [Crossref] [PubMed]
  107. Suryadevara V, Ramchandran R, Kamp DW, et al. Lipid Mediators Regulate Pulmonary Fibrosis: Potential Mechanisms and Signaling Pathways. Int J Mol Sci 2020;21:4257. [Crossref] [PubMed]
  108. Sunaga H, Matsui H, Ueno M, et al. Deranged fatty acid composition causes pulmonary fibrosis in Elovl6-deficient mice. Nat Commun 2013;4:2563. [Crossref] [PubMed]
  109. Zhang J, Muise ES, Han S, et al. Molecular Profiling Reveals a Common Metabolic Signature of Tissue Fibrosis. Cell Rep Med 2020;1:100056. [Crossref] [PubMed]
  110. Goetzman ES, Alcorn JF, Bharathi SS, et al. Long-chain acyl-CoA dehydrogenase deficiency as a cause of pulmonary surfactant dysfunction. J Biol Chem 2014;289:10668-79. [Crossref] [PubMed]
  111. Zhao X, Qin W, Jiang Y, et al. ACADL plays a tumor-suppressor role by targeting Hippo/YAP signaling in hepatocellular carcinoma. NPJ Precis Oncol 2020;4:7. [Crossref] [PubMed]
  112. Xie BX, Zhang H, Wang J, et al. Analysis of differentially expressed genes in LNCaP prostate cancer progression model. J Androl 2011;32:170-82. [Crossref] [PubMed]
  113. Yu DL, Li HW, Wang Y, et al. Acyl-CoA dehydrogenase long chain expression is associated with esophageal squamous cell carcinoma progression and poor prognosis. Onco Targets Ther 2018;11:7643-53. [Crossref] [PubMed]
  114. Puente XS, Sánchez LM, Overall CM, et al. Human and mouse proteases: a comparative genomic approach. Nat Rev Genet 2003;4:544-58. [Crossref] [PubMed]
  115. Verma RP, Hansch C. Matrix metalloproteinases (MMPs): chemical-biological functions and (Q)SARs. Bioorg Med Chem 2007;15:2223-68. [Crossref] [PubMed]
  116. Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 2001;17:463-516. [Crossref] [PubMed]
  117. Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell 2010;141:52-67. [Crossref] [PubMed]
  118. Wood AM, Stockley RA. The genetics of chronic obstructive pulmonary disease. Respir Res 2006;7:130. [Crossref] [PubMed]
  119. Seiki M. Membrane-type 1 matrix metalloproteinase: a key enzyme for tumor invasion. Cancer Lett 2003;194:1-11. [Crossref] [PubMed]
  120. Sauter W, Rosenberger A, Beckmann L, et al. Matrix metalloproteinase 1 (MMP1) is associated with early-onset lung cancer. Cancer Epidemiol Biomarkers Prev 2008;17:1127-35. [Crossref] [PubMed]
  121. Ammar R, Sivakumar P, Jarai G, et al. A robust data-driven genomic signature for idiopathic pulmonary fibrosis with applications for translational model selection. PLoS One 2019;14:e0215565. [Crossref] [PubMed]
  122. Schamberger AC, Schiller HB, Fernandez IE, et al. Glutathione peroxidase 3 localizes to the epithelial lining fluid and the extracellular matrix in interstitial lung disease. Sci Rep 2016;6:29952. [Crossref] [PubMed]
  123. Blacquière MJ, Timens W, van den Berg A, et al. Maternal smoking during pregnancy decreases Wnt signalling in neonatal mice. Thorax 2010;65:553-4. [Crossref] [PubMed]
  124. Wang A, Zsengellér ZK, Hecht JL, et al. Excess placental secreted frizzled-related protein 1 in maternal smokers impairs fetal growth. J Clin Invest 2015;125:4021-5. [Crossref] [PubMed]
  125. Noël A, Hansen S, Zaman A, et al. In utero exposures to electronic-cigarette aerosols impair the Wnt signaling during mouse lung development. Am J Physiol Lung Cell Mol Physiol 2020;318:L705-22. [Crossref] [PubMed]
  126. Bauer Y, Tedrow J, de Bernard S, et al. A novel genomic signature with translational significance for human idiopathic pulmonary fibrosis. Am J Respir Cell Mol Biol 2015;52:217-31. [Crossref] [PubMed]
  127. Zhang X. The Expression Profile and Prognostic Values of EPHA Family Members in Breast Cancer. Front Oncol 2021;11:619949. [Crossref] [PubMed]
  128. Mathot L, Kundu S, Ljungström V, et al. Somatic Ephrin Receptor Mutations Are Associated with Metastasis in Primary Colorectal Cancer. Cancer Res 2017;77:1730-40. [Crossref] [PubMed]
  129. Huang W, Lin A, Luo P, et al. EPHA5 mutation predicts the durable clinical benefit of immune checkpoint inhibitors in patients with lung adenocarcinoma. Cancer Gene Ther 2021;28:864-74. [Crossref] [PubMed]
  130. Wang H, Shan Q, Guo J, et al. PDL1 high expression without TP53, KEAP1 and EPHA5 mutations could better predict survival for patients with NSCLC receiving atezolizumab. Lung Cancer 2021;151:76-83. [Crossref] [PubMed]
  131. Zhang J, Zhang Z, Song W, et al. EPHA5 mutation impairs natural killer cell-mediated cytotoxicity against non-small lung cancer cells and promotes cancer cell migration and invasion. Mol Cell Probes 2020;52:101566. [Crossref] [PubMed]
  132. Xue T, Qiu X, Liu H, et al. Epigenetic regulation in fibrosis progress. Pharmacol Res 2021;173:105910. [Crossref] [PubMed]
  133. Zeisberg EM, Zeisberg M. The role of promoter hypermethylation in fibroblast activation and fibrogenesis. J Pathol 2013;229:264-73. [Crossref] [PubMed]
  134. Mann J, Mann DA. Epigenetic regulation of wound healing and fibrosis. Curr Opin Rheumatol 2013;25:101-7. [Crossref] [PubMed]
  135. Hardy T, Mann DA. Epigenetics in liver disease: from biology to therapeutics. Gut 2016;65:1895-905. [Crossref] [PubMed]
  136. McErlean P, Bell CG, Hewitt RJ, et al. DNA Methylome Alterations Are Associated with Airway Macrophage Differentiation and Phenotype during Lung Fibrosis. Am J Respir Crit Care Med 2021;204:954-66. [Crossref] [PubMed]
  137. Ballinger MN, Mora AL. The Epigenomic Landscape: A Cornerstone of Macrophage Phenotype Regulation in the Fibrotic Lung. Am J Respir Crit Care Med 2021;204:881-3. [Crossref] [PubMed]
  138. Helling BA, Yang IV. Epigenetics in lung fibrosis: from pathobiology to treatment perspective. Curr Opin Pulm Med 2015;21:454-62. [Crossref] [PubMed]
  139. Yang IV, Schwartz DA. Epigenetics of idiopathic pulmonary fibrosis. Transl Res 2015;165:48-60. [Crossref] [PubMed]
  140. Sundar IK, Nevid MZ, Friedman AE, et al. Cigarette smoke induces distinct histone modifications in lung cells: implications for the pathogenesis of COPD and lung cancer. J Proteome Res 2014;13:982-96. [Crossref] [PubMed]
  141. Karatzas E, Kakouri AC, Kolios G, et al. Fibrotic expression profile analysis reveals repurposed drugs with potential anti-fibrotic mode of action. PLoS One 2021;16:e0249687. [Crossref] [PubMed]
  142. Wang YC, Chen Q, Luo JM, et al. Notch1 promotes the pericyte-myofibroblast transition in idiopathic pulmonary fibrosis through the PDGFR/ROCK1 signal pathway. Exp Mol Med 2019;51:1-11. [Crossref] [PubMed]
  143. Yin Q, Wang W, Cui G, et al. Potential role of the Jagged1/Notch1 signaling pathway in the endothelial-myofibroblast transition during BLM-induced pulmonary fibrosis. J Cell Physiol 2018;233:2451-63. [Crossref] [PubMed]
  144. Tsao PN, Matsuoka C, Wei SC, et al. Epithelial Notch signaling regulates lung alveolar morphogenesis and airway epithelial integrity. Proc Natl Acad Sci U S A 2016;113:8242-7. [Crossref] [PubMed]
  145. Peng D, Si D, Zhang R, et al. Deletion of SMARCA4 impairs alveolar epithelial type II cells proliferation and aggravates pulmonary fibrosis in mice. Genes Dis 2017;4:204-14. [Crossref] [PubMed]
  146. Russo RC, Garcia CC, Teixeira MM, et al. The CXCL8/IL-8 chemokine family and its receptors in inflammatory diseases. Expert Rev Clin Immunol 2014;10:593-619. [Crossref] [PubMed]
  147. Rabinovich EI, Kapetanaki MG, Steinfeld I, et al. Global methylation patterns in idiopathic pulmonary fibrosis. PLoS One 2012;7:e33770. [Crossref] [PubMed]
  148. Evans IC, Barnes JL, Garner IM, et al. Epigenetic regulation of cyclooxygenase-2 by methylation of c8orf4 in pulmonary fibrosis. Clin Sci (Lond) 2016;130:575-86. [Crossref] [PubMed]
  149. Li M, Zheng Y, Yuan H, et al. Effects of dynamic changes in histone acetylation and deacetylase activity on pulmonary fibrosis. Int Immunopharmacol 2017;52:272-80. [Crossref] [PubMed]
  150. Tao J, Zhang M, Wen Z, et al. Inhibition of EP300 and DDR1 synergistically alleviates pulmonary fibrosis in vitro and in vivo. Biomed Pharmacother 2018;106:1727-33. [Crossref] [PubMed]
  151. O'Reilly S. Epigenetics in fibrosis. Mol Aspects Med 2017;54:89-102. [Crossref] [PubMed]
  152. Peifer M, Fernández-Cuesta L, Sos ML, et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat Genet 2012;44:1104-10. [Crossref] [PubMed]
  153. Mamdani H, Jalal SI. Histone Deacetylase Inhibition in Non-small Cell Lung Cancer: Hype or Hope? Front Cell Dev Biol 2020;8:582370. [Crossref] [PubMed]
  154. Tang X, Peng R, Ren Y, et al. BET bromodomain proteins mediate downstream signaling events following growth factor stimulation in human lung fibroblasts and are involved in bleomycin-induced pulmonary fibrosis. Mol Pharmacol 2013;83:283-93. [Crossref] [PubMed]
  155. Sanders YY, Lyv X, Zhou QJ, et al. Brd4-p300 inhibition downregulates Nox4 and accelerates lung fibrosis resolution in aged mice. JCI Insight 2020;5:137127. [Crossref] [PubMed]
  156. Singh A, Singh AK, Giri R, et al. The role of microRNA-21 in the onset and progression of cancer. Future Med Chem 2021;13:1885-906. [Crossref] [PubMed]
  157. Liu Y, Nie H, Ding Y, et al. MiRNA, a New Treatment Strategy for Pulmonary Fibrosis. Curr Drug Targets 2021;22:793-802. [Crossref] [PubMed]
  158. Pandit KV, Milosevic J, Kaminski N. MicroRNAs in idiopathic pulmonary fibrosis. Transl Res 2011;157:191-9. [Crossref] [PubMed]
  159. Chirshev E, Oberg KC, Ioffe YJ, et al. Let-7 as biomarker, prognostic indicator, and therapy for precision medicine in cancer. Clin Transl Med 2019;8:24. [Crossref] [PubMed]
  160. Wang Y, Le Y, Xue JY, et al. Let-7d miRNA prevents TGF-β1-induced EMT and renal fibrogenesis through regulation of HMGA2 expression. Biochem Biophys Res Commun 2016;479:676-82. [Crossref] [PubMed]
  161. Xie H, Gao YM, Zhang YC, et al. Low let-7d exosomes from pulmonary vascular endothelial cells drive lung pericyte fibrosis through the TGFβRI/FoxM1/Smad/β-catenin pathway. J Cell Mol Med 2020;24:13913-26. [Crossref] [PubMed]
  162. Xue J, Lin X, Chiu WT, et al. Sustained activation of SMAD3/SMAD4 by FOXM1 promotes TGF-β-dependent cancer metastasis. J Clin Invest 2014;124:564-79. [Crossref] [PubMed]
  163. Penke LR, Speth JM, Dommeti VL, et al. FOXM1 is a critical driver of lung fibroblast activation and fibrogenesis. J Clin Invest 2018;128:2389-405. [Crossref] [PubMed]
  164. Bica-Pop C, Cojocneanu-Petric R, Magdo L, et al. Overview upon miR-21 in lung cancer: focus on NSCLC. Cell Mol Life Sci 2018;75:3539-51. [Crossref] [PubMed]
  165. Liu G, Friggeri A, Yang Y, et al. miR-21 mediates fibrogenic activation of pulmonary fibroblasts and lung fibrosis. J Exp Med 2010;207:1589-97. [Crossref] [PubMed]
  166. Roy S, Khanna S, Hussain SR, et al. MicroRNA expression in response to murine myocardial infarction: miR-21 regulates fibroblast metalloprotease-2 via phosphatase and tensin homologue. Cardiovasc Res 2009;82:21-9. [Crossref] [PubMed]
  167. Bibaki E, Tsitoura E, Vasarmidi E, et al. miR-185 and miR-29a are similarly expressed in the bronchoalveolar lavage cells in IPF and lung cancer but common targets DNMT1 and COL1A1 show disease specific patterns. Mol Med Rep 2018;17:7105-12. [Crossref] [PubMed]
  168. Zhang X, Li Y, Qi P, et al. Biology of MiR-17-92 Cluster and Its Progress in Lung Cancer. Int J Med Sci 2018;15:1443-8. [Crossref] [PubMed]
  169. Dakhlallah D, Batte K, Wang Y, et al. Epigenetic regulation of miR-17~92 contributes to the pathogenesis of pulmonary fibrosis. Am J Respir Crit Care Med 2013;187:397-405. [Crossref] [PubMed]
  170. Hadjicharalambous MR, Lindsay MA. Long Non-Coding RNAs and the Innate Immune Response. Noncoding RNA 2019;5:34. [Crossref] [PubMed]
  171. Hao X, Du Y, Qian L, et al. Upregulation of long noncoding RNA AP003419.16 predicts high risk of aging-associated idiopathic pulmonary fibrosis. Mol Med Rep 2017;16:8085-91. [Crossref] [PubMed]
  172. Zhang H, Song M, Guo J, et al. The function of non-coding RNAs in idiopathic pulmonary fibrosis. Open Med (Wars) 2021;16:481-90. [Crossref] [PubMed]
  173. Dinh PC, Paudel D, Brochu H, et al. Inhalation of lung spheroid cell secretome and exosomes promotes lung repair in pulmonary fibrosis. Nat Commun 2020;11:1064. [Crossref] [PubMed]
  174. Xie L, Zeng Y. Therapeutic Potential of Exosomes in Pulmonary Fibrosis. Front Pharmacol 2020;11:590972. [Crossref] [PubMed]
  175. Parris BA, O'Farrell HE, Fong KM, et al. Chronic obstructive pulmonary disease (COPD) and lung cancer: common pathways for pathogenesis. J Thorac Dis 2019;11:S2155-72. [Crossref] [PubMed]
  176. Molyneaux PL, Cox MJ, Wells AU, et al. Changes in the respiratory microbiome during acute exacerbations of idiopathic pulmonary fibrosis. Respir Res 2017;18:29. [Crossref] [PubMed]
  177. Roy MG, Livraghi-Butrico A, Fletcher AA, et al. Muc5b is required for airway defence. Nature 2014;505:412-6. [Crossref] [PubMed]
  178. Liu NN, Ma Q, Ge Y, et al. Microbiome dysbiosis in lung cancer: from composition to therapy. NPJ Precis Oncol 2020;4:33. [Crossref] [PubMed]
  179. Goto T. Airway Microbiota as a Modulator of Lung Cancer. Int J Mol Sci 2020;21:3044. [Crossref] [PubMed]
  180. Kovaleva OV, Romashin D, Zborovskaya IB, et al. Human Lung Microbiome on the Way to Cancer. J Immunol Res 2019;2019:1394191. [Crossref] [PubMed]
  181. Lipinski JH, Moore BB, O'Dwyer DN. The evolving role of the lung microbiome in pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2020;319:L675-82. [Crossref] [PubMed]
  182. Segal LN, Molyneaux PL. The Challenging Road of Moving from Association to Causation for Microbiome Research in Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2019;199:1054-6. [Crossref] [PubMed]
  183. Han MK, Zhou Y, Murray S, et al. Lung microbiome and disease progression in idiopathic pulmonary fibrosis: an analysis of the COMET study. Lancet Respir Med 2014;2:548-56. [Crossref] [PubMed]
  184. O'Dwyer DN, Ashley SL, Gurczynski SJ, et al. Lung Microbiota Contribute to Pulmonary Inflammation and Disease Progression in Pulmonary Fibrosis. Am J Respir Crit Care Med 2019;199:1127-38. [Crossref] [PubMed]
  185. Richter AG, Stockley RA, Harper L, et al. Pulmonary infection in Wegener granulomatosis and idiopathic pulmonary fibrosis. Thorax 2009;64:692-7. [Crossref] [PubMed]
  186. Idiopathic Pulmonary Fibrosis Clinical Research Network. Prednisone, azathioprine, and N-acetylcysteine for pulmonary fibrosis. N Engl J Med 2012;366:1968-77. [Crossref] [PubMed]
  187. Macaluso C, Maritano Furcada J, Alzaher O, et al. The potential impact of azithromycin in idiopathic pulmonary fibrosis. Eur Respir J 2019;53:1800628. [Crossref] [PubMed]
  188. Shulgina L, Cahn AP, Chilvers ER, et al. Treating idiopathic pulmonary fibrosis with the addition of co-trimoxazole: a randomised controlled trial. Thorax 2013;68:155-62. [Crossref] [PubMed]
  189. Wang J, Lesko M, Badri MH, et al. Lung microbiome and host immune tone in subjects with idiopathic pulmonary fibrosis treated with inhaled interferon-γ. ERJ Open Res 2017;3:e00008-2017. [Crossref] [PubMed]
  190. Kitsios GD, Rojas M, Kass DJ, et al. Microbiome in lung explants of idiopathic pulmonary fibrosis: a case-control study in patients with end-stage fibrosis. Thorax 2018;73:481-4. [Crossref] [PubMed]
  191. Yu G, Ibarra GH, Kaminski N. Fibrosis: Lessons from OMICS analyses of the human lung. Matrix Biol 2018;68-69:422-34. [Crossref] [PubMed]
  192. Siravegna G, Marsoni S, Siena S, et al. Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol 2017;14:531-48. [Crossref] [PubMed]
  193. Bae W, Lee CH, Lee J, et al. Impact of smoking on the development of idiopathic pulmonary fibrosis: results from a nationwide population-based cohort study. Thorax 2021; Epub ahead of print. [Crossref] [PubMed]
  194. Saarenheimo J, Eigeliene N, Andersen H, et al. The Value of Liquid Biopsies for Guiding Therapy Decisions in Non-small Cell Lung Cancer. Front Oncol 2019;9:129. [Crossref] [PubMed]
  195. Whalen W, Buyukozkan M, Moore B, et al. Association of circulating cell-free double-stranded DNA and metabolic derangements in idiopathic pulmonary fibrosis. Thorax 2022;77:186-90. [Crossref] [PubMed]
  196. Pallante P, Malapelle U, Nacchio M, et al. Liquid Biopsy Is a Promising Tool for Genetic Testing in Idiopathic Pulmonary Fibrosis. Diagnostics (Basel) 2021;11:1202. [Crossref] [PubMed]
  197. Dobrinskikh E, Estrella AM, Hennessy CE, et al. Genes, other than Muc5b, play a role in bleomycin-induced lung fibrosis. Am J Physiol Lung Cell Mol Physiol 2021;321:L440-50. [Crossref] [PubMed]
  198. Yoneshima Y, Iwama E, Matsumoto S, et al. Paired analysis of tumor mutation burden for lung adenocarcinoma and associated idiopathic pulmonary fibrosis. Sci Rep 2021;11:12732. [Crossref] [PubMed]
  199. Kobayashi H, Serizawa M, Naito T, et al. Characterization of tumour mutation burden in patients with non-small cell lung cancer and interstitial lung disease. Respirology 2020;25:850-4. [Crossref] [PubMed]
  200. Goodman AM, Kato S, Bazhenova L, et al. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol Cancer Ther 2017;16:2598-608. [Crossref] [PubMed]
  201. Stewart ID, Nanji H, Figueredo G, et al. Circulating fibrocytes are not disease-specific prognosticators in idiopathic pulmonary fibrosis. Eur Respir J 2021;58:2100172. [Crossref] [PubMed]
  202. Moeller A, Gilpin SE, Ask K, et al. Circulating fibrocytes are an indicator of poor prognosis in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2009;179:588-94. [Crossref] [PubMed]
  203. De Biasi S, Cerri S, Bianchini E, et al. Levels of circulating endothelial cells are low in idiopathic pulmonary fibrosis and are further reduced by anti-fibrotic treatments. BMC Med 2015;13:277. [Crossref] [PubMed]
  204. Fernandez IE, Kass DJ. Do Circulating Monocytes Promote and Predict Idiopathic Pulmonary Fibrosis Progression? Am J Respir Crit Care Med 2021;204:9-11. [Crossref] [PubMed]
  205. Raghu G, Collard HR, Egan JJ, et al. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management. Am J Respir Crit Care Med 2011;183:788-824. [Crossref] [PubMed]
  206. Lee CU, White DB, Sykes AM. Establishing a chest MRI practice and its clinical applications: our insight and protocols. J Clin Imaging Sci 2014;4:17. [Crossref] [PubMed]
  207. Shen G, Jia Z, Deng H. Apparent diffusion coefficient values of diffusion-weighted imaging for distinguishing focal pulmonary lesions and characterizing the subtype of lung cancer: a meta-analysis. Eur Radiol 2016;26:556-66. [Crossref] [PubMed]
  208. Lee G, Park H, Bak SH, et al. Radiomics in Lung Cancer from Basic to Advanced: Current Status and Future Directions. Korean J Radiol 2020;21:159-71. [Crossref] [PubMed]
  209. Heukels P, Moor CC, von der Thüsen JH, et al. Inflammation and immunity in IPF pathogenesis and treatment. Respir Med 2019;147:79-91. [Crossref] [PubMed]
  210. Zhu Z, Homer RJ, Wang Z, et al. Pulmonary expression of interleukin-13 causes inflammation, mucus hypersecretion, subepithelial fibrosis, physiologic abnormalities, and eotaxin production. J Clin Invest 1999;103:779-88. [Crossref] [PubMed]
  211. Kolodsick JE, Toews GB, Jakubzick C, et al. Protection from fluorescein isothiocyanate-induced fibrosis in IL-13-deficient, but not IL-4-deficient, mice results from impaired collagen synthesis by fibroblasts. J Immunol 2004;172:4068-76. [Crossref] [PubMed]
  212. Wijsenbeek MS, Kool M, Cottin V. Targeting interleukin-13 in idiopathic pulmonary fibrosis: from promising path to dead end. Eur Respir J 2018;52:1802111. [Crossref] [PubMed]
  213. Maher TM, Costabel U, Glassberg MK, et al. Phase 2 trial to assess lebrikizumab in patients with idiopathic pulmonary fibrosis. Eur Respir J 2021;57:1902442. [Crossref] [PubMed]
  214. Parker JM, Glaspole IN, Lancaster LH, et al. A Phase 2 Randomized Controlled Study of Tralokinumab in Subjects with Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2018;197:94-103. [Crossref] [PubMed]
  215. Ahluwalia N, Shea BS, Tager AM. New therapeutic targets in idiopathic pulmonary fibrosis. Aiming to rein in runaway wound-healing responses. Am J Respir Crit Care Med 2014;190:867-78. [Crossref] [PubMed]
  216. Burgoyne RA, Fisher AJ, Borthwick LA. The Role of Epithelial Damage in the Pulmonary Immune Response. Cells 2021;10:2763. [Crossref] [PubMed]
  217. Shenderov K, Collins SL, Powell JD, et al. Immune dysregulation as a driver of idiopathic pulmonary fibrosis. J Clin Invest 2021;131:143226. [Crossref] [PubMed]
  218. Kinoshita T, Goto T. Molecular Mechanisms of Pulmonary Fibrogenesis and Its Progression to Lung Cancer: A Review. Int J Mol Sci 2019;20:1461. [Crossref] [PubMed]
  219. van Geffen C, Deißler A, Quante M, et al. Regulatory Immune Cells in Idiopathic Pulmonary Fibrosis: Friends or Foes? Front Immunol 2021;12:663203. [Crossref] [PubMed]
  220. Nuovo GJ, Hagood JS, Magro CM, et al. The distribution of immunomodulatory cells in the lungs of patients with idiopathic pulmonary fibrosis. Mod Pathol 2012;25:416-33. [Crossref] [PubMed]
  221. Robert C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat Commun 2020;11:3801. [Crossref] [PubMed]
  222. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol 2020;20:651-68. [Crossref] [PubMed]
  223. Celada LJ, Kropski JA, Herazo-Maya JD, et al. PD-1 up-regulation on CD4+ T cells promotes pulmonary fibrosis through STAT3-mediated IL-17A and TGF-β1 production. Sci Transl Med 2018;10:eaar8356. [Crossref] [PubMed]
  224. Jovanovic D, Roksandic Milenkovic M, Kotur Stevuljevic J, et al. Membrane PD-L1 expression and soluble PD-L1 plasma levels in idiopathic pulmonary fibrosis-a pilot study. J Thorac Dis 2018;10:6660-9. [Crossref] [PubMed]
  225. Geng Y, Liu X, Liang J, et al. PD-L1 on invasive fibroblasts drives fibrosis in a humanized model of idiopathic pulmonary fibrosis. JCI Insight 2019;4:e125326. [Crossref] [PubMed]
  226. Duitman J, van den Ende T, Spek CA. Immune Checkpoints as Promising Targets for the Treatment of Idiopathic Pulmonary Fibrosis? J Clin Med 2019;8:1547. [Crossref] [PubMed]
  227. Zou S, Tong Q, Liu B, et al. Targeting STAT3 in Cancer Immunotherapy. Mol Cancer 2020;19:145. [Crossref] [PubMed]
  228. Antoniou KM, Karagiannis K, Tsitoura E, et al. Mesenchymal stem cell treatment for IPF-time for phase 2 trials? Lancet Respir Med 2017;5:472-3. [Crossref] [PubMed]
  229. Habiel DM, Espindola MS, Kitson C, et al. Characterization of CD28null T cells in idiopathic pulmonary fibrosis. Mucosal Immunol 2019;12:212-22. [Crossref] [PubMed]
  230. Li X, Cai H, Cai Y, et al. Investigation of a Hypoxia-Immune-Related Microenvironment Gene Signature and Prediction Model for Idiopathic Pulmonary Fibrosis. Front Immunol 2021;12:629854. [Crossref] [PubMed]
  231. Ide M, Tanaka K, Sunami S, et al. Durable response to nivolumab in a lung adenocarcinoma patient with idiopathic pulmonary fibrosis. Thorac Cancer 2018;9:1519-21. [Crossref] [PubMed]
  232. Khunger M, Velcheti V. A Case of a Patient with Idiopathic Pulmonary Fibrosis with Lung Squamous Cell Carcinoma Treated with Nivolumab. J Thorac Oncol 2017;12:e96-7. [Crossref] [PubMed]
  233. Dobre IA, Frank AJ, D'Silva KM, et al. Outcomes of Patients With Interstitial Lung Disease Receiving Programmed Cell Death 1 Inhibitors: A Retrospective Case Series. Clin Lung Cancer 2021;22:e738-44. [Crossref] [PubMed]
  234. Delaunay M, Cadranel J, Lusque A, et al. Immune-checkpoint inhibitors associated with interstitial lung disease in cancer patients. Eur Respir J 2017;50:1700050. [Crossref] [PubMed]
  235. Trusolino L, Bertotti A, Comoglio PM. MET signalling: principles and functions in development, organ regeneration and cancer. Nat Rev Mol Cell Biol 2010;11:834-48. [Crossref] [PubMed]
  236. Boccaccio C, Comoglio PM. MET, a driver of invasive growth and cancer clonal evolution under therapeutic pressure. Curr Opin Cell Biol 2014;31:98-105. [Crossref] [PubMed]
  237. Dantoing E, Piton N, Salaün M, et al. Anti-PD1/PD-L1 Immunotherapy for Non-Small Cell Lung Cancer with Actionable Oncogenic Driver Mutations. Int J Mol Sci 2021;22:6288. [Crossref] [PubMed]
  238. Domènech M, Muñoz Marmol AM, Mate JL, et al. Correlation between PD-L1 expression and MET gene amplification in patients with advanced non-small cell lung cancer and no other actionable oncogenic driver. Oncotarget 2021;12:1802-10. [Crossref] [PubMed]
  239. Giuranno L, Ient J, De Ruysscher D, et al. Radiation-Induced Lung Injury (RILI). Front Oncol 2019;9:877. [Crossref] [PubMed]
  240. Türkkan G, Willems Y, Hendriks LEL, et al. Idiopathic pulmonary fibrosis: Current knowledge, future perspectives and its importance in radiation oncology. Radiother Oncol 2021;155:269-77. [Crossref] [PubMed]
  241. Kim H, Pyo H, Noh JM, et al. Preliminary result of definitive radiotherapy in patients with non-small cell lung cancer who have underlying idiopathic pulmonary fibrosis: comparison between X-ray and proton therapy. Radiat Oncol 2019;14:19. [Crossref] [PubMed]
  242. De Ruysscher D, Faivre-Finn C, Moeller D, et al. European Organization for Research and Treatment of Cancer (EORTC) recommendations for planning and delivery of high-dose, high precision radiotherapy for lung cancer. Radiother Oncol 2017;124:1-10. [Crossref] [PubMed]
  243. Sato T, Watanabe A, Kondo H, et al. Long-term results and predictors of survival after surgical resection of patients with lung cancer and interstitial lung diseases. J Thorac Cardiovasc Surg 2015;149:64-9, 70.e1-2.
  244. Sato T, Teramukai S, Kondo H, et al. Impact and predictors of acute exacerbation of interstitial lung diseases after pulmonary resection for lung cancer. J Thorac Cardiovasc Surg 2014;147:1604-1611.e3. [Crossref] [PubMed]
  245. Sato T, Kondo H, Watanabe A, et al. A simple risk scoring system for predicting acute exacerbation of interstitial pneumonia after pulmonary resection in lung cancer patients. Gen Thorac Cardiovasc Surg 2015;63:164-72. [Crossref] [PubMed]
  246. Bi N, Shedden K, Zheng X, et al. Comparison of the Effectiveness of Radiofrequency Ablation With Stereotactic Body Radiation Therapy in Inoperable Stage I Non-Small Cell Lung Cancer: A Systemic Review and Pooled Analysis. Int J Radiat Oncol Biol Phys 2016;95:1378-90. [Crossref] [PubMed]
  247. Pittenger MF, Discher DE, Péault BM, et al. Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen Med 2019;4:22. [Crossref] [PubMed]
  248. Toonkel RL, Hare JM, Matthay MA, et al. Mesenchymal stem cells and idiopathic pulmonary fibrosis. Potential for clinical testing. Am J Respir Crit Care Med 2013;188:133-40. [Crossref] [PubMed]
  249. Chuang HM, Shih TE, Lu KY, et al. Mesenchymal Stem Cell Therapy of Pulmonary Fibrosis: Improvement with Target Combination. Cell Transplant 2018;27:1581-7. [Crossref] [PubMed]
  250. Yang S, Liu P, Jiang Y, et al. Therapeutic Applications of Mesenchymal Stem Cells in Idiopathic Pulmonary Fibrosis. Front Cell Dev Biol 2021;9:639657. [Crossref] [PubMed]
  251. Samarelli AV, Tonelli R, Heijink I, et al. Dissecting the Role of Mesenchymal Stem Cells in Idiopathic Pulmonary Fibrosis: Cause or Solution. Front Pharmacol 2021;12:692551. [Crossref] [PubMed]
  252. Ghadiri M, Young PM, Traini D. Cell-based therapies for the treatment of idiopathic pulmonary fibrosis (IPF) disease. Expert Opin Biol Ther 2016;16:375-87. [Crossref] [PubMed]
  253. Mehrad B, Burdick MD, Zisman DA, et al. Circulating peripheral blood fibrocytes in human fibrotic interstitial lung disease. Biochem Biophys Res Commun 2007;353:104-8. [Crossref] [PubMed]
  254. Hashimoto N, Jin H, Liu T, et al. Bone marrow-derived progenitor cells in pulmonary fibrosis. J Clin Invest 2004;113:243-52. [Crossref] [PubMed]
  255. Yoneshima Y, Iwama E, Matsumoto S, et al. Paired analysis of tumor mutation burden for lung adenocarcinoma and associated idiopathic pulmonary fibrosis. Sci Rep 2021;11:12732. [Crossref] [PubMed]
  256. Selman M, Pardo A. The leading role of epithelial cells in the pathogenesis of idiopathic pulmonary fibrosis. Cell Signal 2020;66:109482. [Crossref] [PubMed]
  257. Glassberg MK, Minkiewicz J, Toonkel RL, et al. Allogeneic Human Mesenchymal Stem Cells in Patients With Idiopathic Pulmonary Fibrosis via Intravenous Delivery (AETHER): A Phase I Safety Clinical Trial. Chest 2017;151:971-81. [Crossref] [PubMed]
  258. Ushakumary MG, Riccetti M, Perl AT. Resident interstitial lung fibroblasts and their role in alveolar stem cell niche development, homeostasis, injury, and regeneration. Stem Cells Transl Med 2021;10:1021-32. [Crossref] [PubMed]
  259. Ostrom RS. A two-pronged weapon in the fight against fibrosis. Focus on "Inhibition of Wnt/β-catenin signaling promotes epithelial differentiation of mesenchymal stem cells and repairs bleomycin-induced lung injury". Am J Physiol Cell Physiol 2014;307:C232-3. [Crossref] [PubMed]
  260. Wang C, Zhu H, Sun Z, et al. Inhibition of Wnt/β-catenin signaling promotes epithelial differentiation of mesenchymal stem cells and repairs bleomycin-induced lung injury. Am J Physiol Cell Physiol 2014;307:C234-44. [Crossref] [PubMed]
  261. Cárdenes N, Álvarez D, Sellarés J, et al. Senescence of bone marrow-derived mesenchymal stem cells from patients with idiopathic pulmonary fibrosis. Stem Cell Res Ther 2018;9:257. [Crossref] [PubMed]
  262. Shang J, Yao Y, Fan X, et al. miR-29c-3p promotes senescence of human mesenchymal stem cells by targeting CNOT6 through p53-p21 and p16-pRB pathways. Biochim Biophys Acta 2016;1863:520-32. [Crossref] [PubMed]
  263. Meng Y, Eirin A, Zhu XY, et al. Micro-RNAS Regulate Metabolic Syndrome-induced Senescence in Porcine Adipose Tissue-derived Mesenchymal Stem Cells through the P16/MAPK Pathway. Cell Transplant 2018;27:1495-503. [Crossref] [PubMed]
  264. Moimas S, Salton F, Kosmider B, et al. miR-200 family members reduce senescence and restore idiopathic pulmonary fibrosis type II alveolar epithelial cell transdifferentiation. ERJ Open Res 2019;5:e00138-2019. [Crossref] [PubMed]
  265. Shi L, Han Q, Hong Y, et al. Inhibition of miR-199a-5p rejuvenates aged mesenchymal stem cells derived from patients with idiopathic pulmonary fibrosis and improves their therapeutic efficacy in experimental pulmonary fibrosis. Stem Cell Res Ther 2021;12:147. [Crossref] [PubMed]
  266. Li X, Yue S, Luo Z. Mesenchymal stem cells in idiopathic pulmonary fibrosis. Oncotarget 2017;8:102600-16. [Crossref] [PubMed]
  267. Khan P, Gazdhar A, Fytianos K, et al. The secretome of lung resident mesenchymal stem cells is anti-fibrotic in vitro and in vivo. Eur Respir J 2018;52:PA594.
  268. Dinh PC, Paudel D, Brochu H, et al. Inhalation of lung spheroid cell secretome and exosomes promotes lung repair in pulmonary fibrosis. Nat Commun 2020;11:1064. [Crossref] [PubMed]
  269. Cahill EF, Kennelly H, Carty F, et al. Hepatocyte Growth Factor Is Required for Mesenchymal Stromal Cell Protection Against Bleomycin-Induced Pulmonary Fibrosis. Stem Cells Transl Med 2016;5:1307-18. [Crossref] [PubMed]
  270. Alsafadi HN, Uhl FE, Pineda RH, et al. Applications and Approaches for Three-Dimensional Precision-Cut Lung Slices. Disease Modeling and Drug Discovery. Am J Respir Cell Mol Biol 2020;62:681-91. [Crossref] [PubMed]
  271. Sundarakrishnan A, Chen Y, Black LD, et al. Engineered cell and tissue models of pulmonary fibrosis. Adv Drug Deliv Rev 2018;129:78-94. [Crossref] [PubMed]
  272. Skibba M, Drelich A, Poellmann M, et al. Nanoapproaches to Modifying Epigenetics of Epithelial Mesenchymal Transition for Treatment of Pulmonary Fibrosis. Front Pharmacol 2020;11:607689. [Crossref] [PubMed]
Cite this article as: Stella GM, D’Agnano V, Piloni D, Saracino L, Lettieri S, Mariani F, Lancia A, Bortolotto C, Rinaldi P, Falanga F, Primiceri C, Corsico AG, Bianco A. The oncogenic landscape of the idiopathic pulmonary fibrosis: a narrative review. Transl Lung Cancer Res 2022;11(3):472-496. doi: 10.21037/tlcr-21-880

Download Citation