Next Article in Journal
Natural Killer (NK) Cell-Based Therapies Have the Potential to Treat Ovarian Cancer Effectively by Targeting Diverse Tumor Populations and Reducing the Risk of Recurrence
Previous Article in Journal
The Immune Landscape of Canine Soft Tissue Sarcomas as a Model for Human Soft Tissue Sarcomas
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

From Fibrosis to Malignancy: Mechanistic Intersections Driving Lung Cancer Progression

1
School of Medicine, Kunming University, Kunming 650214, China
2
Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China
3
College of Life Sciences, Hebei Normal University, Shijiazhuang 050025, China
4
The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming 650011, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2025, 17(23), 3861; https://doi.org/10.3390/cancers17233861 (registering DOI)
Submission received: 13 October 2025 / Revised: 22 November 2025 / Accepted: 24 November 2025 / Published: 1 December 2025
(This article belongs to the Section Cancer Pathophysiology)

Simple Summary

Lung cancer often develops in patients with pulmonary fibrosis, but the link between the two diseases remains unclear. This review examines how inflammation, oxidative stress, and alterations in molecular signaling in fibrosis may contribute to the development of lung cancer. Understanding these shared mechanisms could help identify new targets for the early detection and treatment of lung cancer in patients with fibrotic lungs.

Abstract

Background/Objectives: Pulmonary fibrosis (PF) and lung cancer (LC) are major global health challenges that share several pathogenic mechanisms despite their distinct clinical features. PF leads to progressive fibrotic remodeling and respiratory decline, while LC is characterized by uncontrolled proliferation, invasion, and metastasis. Growing evidence shows that PF markedly increases the risk of LC development. This review aims to clarify the convergent molecular and cellular mechanisms that link fibrogenesis to tumorigenesis. Methods: Published studies exploring shared pathogenic pathways, molecular signaling networks, immune microenvironment alterations, and mitochondrial and genomic disturbances in PF and LC were systematically examined and integrated to identify common mechanisms contributing to fibrosis-associated carcinogenesis. Results: Findings highlight several overlapping processes between PF and LC, including oxidative stress, genomic instability, dysregulated DNA damage repair, immune microenvironment remodeling, mitochondrial dysfunction, and alterations in the ubiquitin–proteasome system. These aberrations drive chronic inflammation, epithelial–mesenchymal transition (EMT), extracellular matrix (ECM) remodeling, and other hallmarks shared by both diseases. Key signaling pathways—such as transforming growth factor-β (TGF-β), programmed cell death protein-1/programmed death-ligand 1 (PD-1/PD-L1), and tumor microenvironment–mediated immune evasion—further contribute to disease progression and increased LC risk in PF patients. Conclusions: Integrating molecular and pathological insights reveals a strong biological continuum between PF and LC. Understanding these convergent mechanisms may facilitate the identification of diagnostic biomarkers and therapeutic targets, ultimately helping to mitigate PF-associated lung carcinogenesis.

1. Introduction

Pulmonary fibrosis (PF) and lung cancer (LC) are among the most significant causes of morbidity and mortality worldwide. PF, a chronic and progressive interstitial lung disease, represents the end stage of various pulmonary disorders and is characterized by excessive deposition of fibrotic tissue, resulting in irreversible architectural distortion and functional decline. Idiopathic pulmonary fibrosis (IPF), the most prevalent form of PF, remains of unknown etiology but is associated with several risk factors, including genetic susceptibility, cigarette smoking, environmental exposure, microbial infection, gastroesophageal reflux, and metabolic disorders such as diabetes mellitus [1,2]. Epidemiological studies estimate the global incidence and prevalence of IPF to range from 0.09 to 1.30 and 0.33–4.51 cases per 10,000 individuals, respectively, with a rising trend in aging populations [3]. The relationship between PF and LC is critical, given LC’s status as the leading cause of cancer-related deaths worldwide, primarily classified as non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC), with SCLC and NSCLC accounting for approximately 15% and 85% of cases, respectively [4]. IPF patients have an approximately eightfold higher risk of developing LC compared to the general population, with reported prevalence rates ranging from 2.7% to 31.3% [5]. LC remains the leading cause of cancer-related mortality worldwide, encompassing two major histological types: non-small-cell lung cancer (NSCLC, ~85%) and small-cell lung cancer (SCLC, ~15%) [4]. The multifactorial etiology of LC involves tobacco exposure, environmental pollutants, genetic mutations, and socioeconomic determinants, all of which contribute to disease heterogeneity and therapeutic challenges [6,7]. The 2020 Global Cancer Statistics report confirmed lung cancer as one of the leading causes of cancer death, with an estimated 2.2 million fatalities [8]. While the epidemiological association between PF and LC is well established, this review focuses on elucidating the mechanistic interplay between these two diseases. We aim to clarify how shared molecular pathways drive lung carcinogenesis within fibrotic lung tissue and how the fibrotic microenvironment shapes tumor behavior. In support of this objective, findings from recent studies were incorporated to highlight the increased incidence and poor prognosis of lung cancer among patients with PF or IPF [9,10,11].
Notably, lung cancers arising in fibrotic lungs display distinct biological and clinical characteristics compared with those developing in non-fibrotic tissue. They frequently occur in the peripheral and lower lobes, are predominantly of the adenocarcinoma subtype with solid or sarcomatoid differentiation and often exhibit aggressive progression with limited therapeutic response. The fibrotic microenvironment enriched in TGF-β, IL-6, and characterized by increased extracellular matrix stiffness facilitates EMT, angiogenesis, and immune evasion, thereby fostering a tumor-promoting niche. Furthermore, chronic tissue injury and architectural remodeling restrict drug penetration and impair immune surveillance, suggesting that cancers developing in fibrotic lungs may follow distinct molecular trajectories and respond differently to targeted or immune-based therapies.
Accordingly, this review integrates current molecular, cellular, and translational evidence to provide a comprehensive perspective on the shared pathogenic mechanisms between PF and LC, offering insights into potential biomarkers and therapeutic targets for early detection and intervention.

2. Oxidative Stress, DNA Damage, and Repair

Oxidative stress, an imbalance between reactive oxygen species (ROS) production and antioxidant defenses, damages cellular components (DNA, proteins, lipids) [12]. Oxidative stress is a common pathogenic mechanism in both PF and LC, and it has been shown to activate fibroblasts, leading to excessive collagen deposition and fibrosis in PF [13]. It has been implicated in the development of lung cancer through various mechanisms, including DNA damage, activation of oncogenes, and inactivation of tumor suppressor genes [14]. Targeting oxidative stress is crucial for developing effective treatments.
PF myofibroblasts originate from diverse sources, including mesenchymal progenitor cells, resident lung fibroblasts, AECs undergoing EMT, and bone marrow-derived fibrocytes [15] and endothelial cells [16]. LC’s cellular origins are heterogeneous, including neuroendocrine cells [17] and alveolar epithelial type II cells (AECII) [18], exhibiting significant inter- and intra-tumoral variability [19]. Despite differing cellular origins, downstream consequences of inflammation and tissue remodeling share common oxidative stress-mediated pathways, creating a convergence point.
Environmental pollutants and tobacco smoke, along with iron overload (through genetic mutations, disease processes, or environmental factors via the Fenton reaction) [20], generate ROS-inducing DNA damage [21], and prolonged exposure leads to the accumulation of unpaired DNA lesions. DNA repair mechanisms (e.g., DNA-dependent protein kinase, Ku protein complex) attempt to mitigate damage [22]. However, unrepaired damage activates the DNA damage response (DDR) pathway. This involves the activation of ATM and ATR kinases, triggering p53-mediated cell cycle arrest and apoptosis. It is stated that p53-mediated cell cycle arrest and DNA damage repair may exert cytoprotective effects against cancer therapies [23,24]. Phase separation dysregulation in DNA damage response can further contribute to genomic instability [25]. Breast Cancer (BRCA1/2) and Poly (ADP-ribose) polymerase (PARP) are crucial for DNA repair (homologous recombination and base excision repair), respectively [24]. Deficient repair contributes to mutations and genomic instability, promoting carcinogenesis [26]. Mitochondrial ROS (mtROS) damage mitochondrial DNA, disrupt membrane integrity, reduce mitochondrial membrane potential, impair oxidative phosphorylation, decrease ATP, and further induce DNA damage and genomic instability, increasing the risk of both carcinogenesis and fibrosis [27,28]. However, not all studies support a purely pathogenic role for oxidative stress. Controlled ROS signaling has been shown to promote epithelial regeneration and activate antioxidant defense pathways through the Nrf2 pathway, suggesting that ROS may have context-dependent protective effects during early injury repair before chronic fibrosis develops [29,30]. The intersection of oxidative stress, DNA damage, and repair in PF and LC is shown in Figure 1.
This concept of DNA damage and repair is directly relevant to the genetic alterations observed in LC and PF. PF, characterized by fibrotic and inflammatory responses, frequently involves loss-of-function mutations, e.g., MUC5B promoter variant rs35705950 in IPF, leading to increased MUC5B expression and disease exacerbation [31]. And telomere shortening (TERT and TERC mutations in familial PF) in familial PF [32]. LC, conversely, often exhibits genomic instability (chromosomal rearrangements, oncogenic gain-of-function mutation), disrupting cell cycle control and inactivating tumor suppressor gene anaplastic lymphoma kinase (ALK) rearrangements in NSCLC [33].
Clinical studies show that oxidative stress plays a major role in both lung cancer and pulmonary fibrosis. In NSCLC, elevated levels of 8-oxo-dG and malondialdehyde (MDA) have been consistently reported, reflecting increased oxidative DNA damage and lipid peroxidation [34]. These markers correlate with disease severity and reduced antioxidant enzyme activities, such as superoxide dismutase (SOD) and glutathione peroxidase (GPx), indicating an overwhelmed antioxidant defense system [34]. In IPF, persistent oxidative stress and mitochondrial dysfunction contribute to DNA damage and reduced antioxidant capacity [35,36]. The same imbalance between oxidative injury (8-oxo-dG, MDA) and antioxidant defense (SOD, GPx) suggests shared molecular mechanisms between fibrosis and tumor progression. Collectively, elevated 8-oxo-dG and MDA, alongside reduced SOD and GPx, serve as reliable indicators of oxidative stress and disease advancement in lung cancer and fibrosis [35,36,37]. Overall, oxidative stress connects pulmonary fibrosis and lung cancer through DNA damage, impaired repair, and genomic instability, emphasizing the need for balanced redox control in disease prevention.

3. Immune Responses and Microenvironment

It is stated that cancer microenvironmental cells are immune cells that exert profound effects on cancer cells [38]. Immune response regulation is crucial for the development and progression of both PF and LC. In PF, inflammation is a key driver of excessive collagen deposition in the lungs, leading to scarring and impaired lung function [39]. Damaged lung epithelial cells, neutrophils, macrophages, and other cells release inflammatory factors such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and transforming growth factor-beta (TGF-β) [40]. These factors stimulate excessive ECM deposition and tissue stiffening by activating fibroblasts and myofibroblasts [41,42,43].
While inflammation is crucial in PF, immune dysregulation in cancer leads to immune evasion. LC development and progression are linked to immune evasion. Effective anti-tumor immunity requires T-cell infiltration, often hindered by cancer cells that inhibit T-cell receptor signaling, reduce T-cell proliferation and cytokine production, and suppress T-cell cytotoxicity through immune checkpoint molecules like PD-1 (encoded by PDCD1) and PD-L1 (encoded by CD274) [44]. In IPF, the chronic inflammatory microenvironment and persistent oxidative stress contribute to DNA damage, genomic instability, and activation of pro-oncogenic pathways. TGF-β signaling is a central pathway in fibrosis, inducing the transformation of fibroblasts into myofibroblasts, which are key effector cells in ECM deposition. High mitochondrial damage also leads to ER stress, both contributing to lung injury and IPF development [36]. These processes are indicative of a dysregulated cellular environment where immune responses are likely altered.
Chimeric Antigen Receptor T-cell (CTLA-4), another critical immune checkpoint molecule, inhibits T-cell activation and proliferation by competitively binding to B7 molecules [45]. NK cells are crucial in early-stage lung cancer, as they recognize and kill cancer cells; however, cancer cells can evade this by altering surface antigens or secreting inhibitory factors. Beyond classical NK cell receptors (KIRs, LIRs, and NKG2A), numerous other immune checkpoints contribute to NK cell dysfunction in various cancers and chronic infections, including CTLA-4, PD-1, B7-H3, as well as LAG-3, TIGIT & CD96, TIM-3, checkpoint members of the Siglecs family (Siglec-7/9), CD200, and CD47 [46]. Macrophage polarization within the tumor microenvironment (TME) significantly impacts disease progression; M1 macrophages exhibit anti-tumor effects, while M2 macrophages promote tumor angiogenesis through the secretion of VEGF and other pro-angiogenic factors, thereby supporting tumor growth and tissue repair [47]. Regulatory T cells (Tregs) secrete immunosuppressive cytokines like IL-10 and TGF-β; IL-10 and TGF-β suppress anti-tumor immunity and promote tumor growth [48]. The intersection of immune response and extracellular matrix in PF and LC is shown in Figure 2.
This immunosuppressive TME poses challenges for effective cancer therapies, including the use of chimeric antigen receptor (CAR) T cells to significantly inhibit tumor growth and improve survival by targeting tumor-associated antigens like EpCAM, EGFR, and ROR1 [49,50]. EpCAM-directed CAR-T cells effectively accumulate in tumors in the brain metastasis model, reducing tumor growth without the central nervous system or systemic toxicity [51]. In PF, CAR-T cell therapy targeting fibrosis-related antigens is under investigation [52]. However, the immunosuppressive TME and antigen heterogeneity in LC complicate CAR-T cell effectiveness [53]. Combination therapies, such as pairing CAR-T cells with immune checkpoint inhibitors (e.g., anti-PD-1), are being explored to enhance antitumor effects [54]. Although PD-1/PD-L1 and TGF-β pathways are commonly linked to immune evasion, several studies demonstrate that transient activation of these signals can also mitigate acute inflammation and tissue injury, acting as compensatory immune regulators. For instance, PD-L1 upregulation may serve as a feedback mechanism to prevent excessive T-cell activation during tissue repair [55,56,57].
In PF, TGF-β, a key regulator, activates fibroblasts via the SMAD signaling pathway, inducing myofibroblast differentiation and increasing collagen production [58]. Moreover, it was stated that TGF-β could promote EMT in tumor cells and facilitate invasion and metastasis of cancer through regulation of cell survival, angiogenesis, vascular integrity, and interaction with the TME [59]. Similar to TGF-β’s role in PF, certain cytokines contribute to immune suppression in cancer, fostering tumor growth and metastasis [43].
In addition, ECM is crucial in both PF and LC as it significantly influences TME. In PF, fibroblast activation and proliferation increase collagen and fibronectin (Fn1) levels in the ECM, leading to excessive accumulation and increased stiffness, promoting the TGF-β/SMAD2/3 signaling pathway in a LEMD3-dependent manner [60]. Moreover, the altered ECM disrupts the balance of MMPs and TIMPs, and the stiffened ECM activates mechanotransduction pathways, perpetuating the fibrotic cycle [61,62,63]. In cancer, ECM components (collagen, elastin, fibronectin, and laminin) reduce treatment efficacy and are associated with poor prognosis, growth, metastasis, and drug resistance [64]. ECM changes (stiffness, degradation) significantly affect tumor growth and progression [65]. In LC, cancer-associated fibroblasts (CAFs) are crucial components of the TME; they promote tumor growth, invasion, and metastasis, exhibiting distinct metabolic profiles compared to normal fibroblasts. CAFs primarily utilize glycolysis to produce lactate, which can then fuel cancer cells, a phenomenon known as the “reverse Warburg effect”. This metabolic interplay between CAFs and cancer cells highlights another avenue for in vitro investigation into therapeutic strategies [66,67]. It was stated that high expression of fibroblast activation protein correlates with poor prognosis in high-grade serous ovarian cancer [68]. Tumor-derived type III collagen maintains tumor dormancy; its disruption reactivates tumor cell proliferation via DDR1-mediated STAT1 signaling and facilitates metastasis [69]. The stiffness of the ECM also hinders drug penetration into tumor regions [65]. ECM also modulates the behavior of immune cells by creating an immunosuppressive microenvironment, thus reducing the effectiveness of immunotherapy [69,70]. Therefore, manipulating ECM stiffness and composition to modulate the TME is a potential anti-tumor strategy [62].

4. Cell Activation, Proliferation, Cell Cycle, and Anti-Apoptotic Mechanisms

Cellular activation, proliferation, cell cycle regulation, and anti-apoptotic mechanisms are critical in the pathogenesis of both PF and LC. In PF, Fibroblast activation, proliferation, and fibrotic differentiation are regulated by multiple signaling pathways, including TGF-β, the Hedgehog, Wnt/β-catenin, and the Notch [71]. These pathways also influence LC; For example, Wnt pathway activation stimulates LC cell proliferation and survival [71]. In addition, LC cells utilize various mechanisms to evade apoptosis and enhance proliferation [72]. Tyrosine kinases play pivotal roles in the signaling pathways that drive lung cancer progression, particularly in NSCLC, where their dysregulation leads to uncontrolled cell growth, proliferation, differentiation, metabolism, and survival [73]. These enzymes function by phosphorylating tyrosine residues on substrate proteins, acting as molecular switches that regulate various cellular processes. Aberrant activation or overexpression of these kinases is a hallmark of cancer, providing critical targets for therapeutic interventions [74]. Several signaling pathways promote cell proliferation and tumor growth, including PI3K/Akt/mTORC1 [75], Ras/Raf/ERK [76], EGFR [77], FGFR [78], HER2 [79], and CDK4/6 [80]. A critical factor in this process is telomerase, which serves as a nearly universal biomarker for advanced human cancers [81]. Telomerase activation maintains telomere length, preventing cellular senescence and death and enabling continuous proliferation [82]. Increased telomerase activity is notably observed in NSCLC and SCLC [83]. Most tumor cells maintain telomere length through telomerase, while a minority utilize the alternative lengthening of telomere mechanism [84]. High telomerase expression correlates with increased proliferation, invasiveness, and anti-apoptotic capacity, making it a potential therapeutic target [85]. Interestingly, telomerase induction is also increased in the lung fibroblasts of IPF patients [86], while alveolar epithelial type II (AECII) in PF patients exhibit significant telomere shortening and DNA damage [87]. This is consistent with fibroblast proliferation and epithelial cell damage in PF. Furthermore, TERT/TERC gene mutations increase the risk of IPF, indicating that telomere length and maintenance capacity are important prognostic factors and potential therapeutic targets [88].
Dysregulated cell cycle control plays a crucial role in the progression of both LC and PF in LC. In AECII of IPF patients, increased p21 expression induces cell cycle arrest and senescence, inhibiting cell differentiation. p21 overexpression promotes myofibroblast activation via profibrotic cytokine release, contributing to fibrosis [89]. Regulator of cell cycle (RGCC) overexpression delays early Smad2/3 phosphorylation induced by TGF-β, having a protective effect in PF [90]. In NSCLC and SCLC, abnormal expression of cell cycle genes (TP53, p21, and cyclin D1) is associated with high proliferative and invasive tumors [91]. The long noncoding RNA HOX Transcript Antisense RNA (HOTAIR) is a marker of cell cycle abnormalities, promoting G1-S phase transition via Rb-E2F pathway regulation and accelerating cell proliferation and invasion via EMT and β-catenin pathways [92]. Small molecule cell cycle regulators such as WEE1 [93] and aurora kinase B inhibitors [94] show potential in SCLC treatment.
Dysregulation of cell death pathways, including apoptosis, contributes to both PF and LC progression. In PF, the PARP-1 inhibitor HYDAMTIQ reduces fibrosis and inflammation in preclinical models by blocking the TGF-β/Smad pathway and reducing fibroblast activation [95]. Cancer pathogenesis also involves dysregulation of cell death pathways, mirroring the situation in PF. This dysregulation includes imbalances in Bcl-2 family proteins, various inhibitors of apoptosis proteins (IAPs), dysregulation of repair mechanisms, activation of P53, and death receptors [64]. The therapeutic exploitation of these dysregulations is evident in the use of PARP inhibitors like Olaparib and Niraparib. These drugs trap PARP-DNA complexes, hindering repair and inducing apoptosis, thereby enhancing cancer treatment efficacy [96]. Survivin overexpression promotes cancer progression and therapy resistance [97]. Targeting anti-apoptotic mechanisms represents a promising therapeutic avenue for both diseases [97]. Conversely, some studies highlight the dual nature of these signaling pathways. For instance, moderate activation of PI3K/Akt and Wnt/β-catenin can promote epithelial repair and protect against apoptosis during acute lung injury, suggesting that complete inhibition may impair regeneration [98]. In summary, immune dysregulation and extracellular matrix remodeling create a shared pathogenic microenvironment in pulmonary fibrosis and lung cancer, fostering chronic inflammation, immune evasion, and tumor progression.

5. Mitochondrial Metabolism

Mitochondrial dysfunction, often a consequence of oxidative stress, is a crucial factor in both PF and LC [99]. Mitochondria are a major source of ROS, particularly during oxidative phosphorylation. Mitochondrial dysfunction often leads to increased ROS production, contributing to oxidative stress and lipid peroxidation, both key drivers of ferroptosis [100]. Moreover, mitochondrial DAMPs, particularly mtDNA and cardiolipin, released as a result of mitochondrial injury in both IPF and lung cancer, can activate innate immune pathways such as TLR9 and cGAS–STING, initiating proinflammatory signaling that further amplifies tissue injury and tumor-promoting inflammation [101].
Ferroptosis, an iron-dependent form of cell death driven by lipid peroxidation and ROS accumulation and closely linked to mitochondrial function, is emerging as a promising therapeutic target in both LC and PF [102,103]. In LC, inducing ferroptosis inhibits cell proliferation and growth [102]. Ferroptosis regulators, such as glutathione peroxidase 4 (GPX4), can promote chemoresistance by suppressing ferroptosis. Conversely, GPX4 inhibitors enhance the efficacy of platinum-based anti-cancer drugs [104]. In PF, increased iron deposition and ferroptosis are observed in IPF lung tissue. Iron exacerbates fibrosis by inducing AECII death and mitochondrial dysfunction [103] and ferroptosis accelerates PF pathology by promoting EMT and ECM accumulation [105].
Mitochondrial homeostasis significantly impacts cellular function in both LC and PF. In LC, mitochondrial dynamics, regulated by proteins like dynamin-related protein 1 (Drp1) that promote mitochondrial fission, influence cell proliferation, invasion, and metastasis [106]. Mitophagy, the selective removal of damaged mitochondria, is essential for mitochondrial quality control. PTEN-Induced Kinase 1 (PINK1) and Parkin proteins mediate the autophagic degradation of damaged mitochondria, maintaining cellular energy metabolism and functional stability and homeostasis [107]. In LC, mitophagy’s role varies under different stress conditions; methyltransferase-like 3 (METTL3) promotes cisplatin and etoposide chemoresistance in SCLC by upregulating PINK1-Parkin-mediated mitophagy [108]. In PF, reduced PINK1 expression in AECII leads to abnormal mitochondrial morphology and increased ROS generation; Decreased PARK2 expression in IPF myofibroblasts results in insufficient mitophagy, leading to mtROS generation and Platelet-derived growth factor receptor (PDGFR)/mTOR signaling activation, and increased myofibroblast differentiation and proliferation [109,110]. Nevertheless, emerging evidence indicates that mitophagy can have both profibrotic and antifibrotic outcomes, depending on cellular context. Excessive mitophagy may deplete functional mitochondria and impair energy metabolism, while insufficient mitophagy promotes oxidative damage [111]. So, mitochondrial dysfunction and ferroptosis are key shared mechanisms in pulmonary fibrosis and lung cancer, linking oxidative stress, iron overload, and impaired mitophagy to fibrosis progression, tumor growth, and therapy resistance. The function of mitochondrial metabolism in PF and LC is shown in Figure 3.

6. The Ubiquitin–Proteasome System in LC and PF

The ubiquitin–proteasome system (UPS) plays a critical role in protein degradation and influences various cellular processes [112]. The UPS plays a crucial role in cancer development, with an imbalance between ubiquitin E3 ligases and deubiquitinases frequently contributing to tumorigenesis and therapeutic resistance [113].
In NSCLC, abnormal ubiquitin modifications are implicated in oncogenesis [114]. High expression of ubiquitin-specific protease USP39 is associated with increased proliferation and drug resistance in LC cells [115]. The 26S proteasome promotes tumorigenesis by regulating the stability and function of tumor suppressor proteins such as p53 [116]. Proteasome inhibitors, such as MG132, induce apoptosis in lung cancer cells by increasing oxidative stress and depleting glutathione, enhancing chemotherapy effectiveness and reducing drug resistance [117].
In PF, 26S proteasome activity is significantly increased, particularly during TGF-β-induced myofibroblast differentiation, with Rpn6 subunit regulation playing a key role [118]. The UPS significantly impacts PF pathogenesis by regulating EMT and intracellular protein homeostasis [16]. The FDA-approved proteasome inhibitor Bortezomib inhibits TGF-β1-induced gene expression in fibroblasts by preventing Smad-mediated transcription and collagen deposition, and it inhibits BLM-induced fibrosis in mice lungs [119]. However, the less toxic, more selective proteasome inhibitor augmented BLM-induced PF in a mouse model following local lung application, highlighting the need for further investigation into the safety and efficacy of proteasome inhibitors in clinical [119]. Interestingly, proteasome inhibition does not uniformly suppress fibrosis or cancer progression. Certain studies indicate that partial proteasome inhibition may disrupt cellular homeostasis and exacerbate stress responses, particularly in lung tissue with high metabolic demand [120].
The UPS represents a promising therapeutic target for both LC and PF. Modulating UPS activity, either through proteasome inhibition or targeting specific deubiquitinases, could offer novel therapeutic strategies. However, the complex roles of the UPS necessitate careful consideration of potential off-target effects and the development of highly specific inhibitors to maximize therapeutic benefit and minimize adverse effects.

7. Biomarkers in Lung Fibrosis and Lung Cancer

Given the shared molecular landscape between pulmonary fibrosis and lung cancer, several classes of biomarkers are being actively investigated for early cancer detection in individuals with fibrotic lungs. Genetic and epigenetic biomarkers play a pivotal role in this regard. Bioinformatics analyses have identified numerous differentially expressed genes (DEGs) and hub genes in IPF that are also implicated in lung cancer [74]. For instance, a study reported 1888 DEGs in IPF—comprising 1105 upregulated and 783 downregulated genes—and identified 10 hub genes potentially crucial for the pathogenesis of IPF and its association with lung cancer [74]. These genes could serve as valuable indicators of increased cancer risk in patients with fibrotic lungs. RNA modifications, particularly N6-methyladenosine (m6A), have also emerged as promising epigenetic biomarkers for both lung cancer and pulmonary fibrosis, suggesting potential utility in identifying early cancer progression within fibrotic lungs [121].
Noncoding RNAs (ncRNAs) have drawn significant attention as diagnostic tools. Exosomal miRNAs are highly stable and easily measurable in biofluids, making them attractive non-invasive biomarkers. Dysregulated miRNA expression has been observed in various lung diseases, including silicosis-related lung fibrosis and NSCLC [122]. The detection of specific miRNA expression profiles in fibrotic individuals could thus provide early indications of malignant transformation. Likewise, circRNAs, which are also found in exosomes, have demonstrated potential as biomarkers for lung cancer due to their high stability and involvement in tumor progression and metastasis [121]. Monitoring the levels of particular circRNAs in fibrotic patients might help identify the onset of cancer development.
Protein biomarkers represent another major class of diagnostic targets. Extracellular heat shock proteins (eHSPs) are currently being investigated as biomarkers and therapeutic targets in fibrosing interstitial lung diseases (ILDs) [123], as their altered expression may indicate a shift toward malignant transformation. Furthermore, oncogenic and immune checkpoint proteins, such as EGFR, ALK, KRAS G12C, and PD-L1, which are well-established in lung cancer, could be monitored in fibrotic patients to detect early malignancy or to guide preventive therapies [124]. A broad range of cancer-associated proteins, including HER2, c-MET, MUC1, EGFR, and PD-L1, are also expressed in lung cancer [125]. Tracking their alterations in the context of lung fibrosis could facilitate early diagnosis and intervention.
The lung microbiome has also emerged as a potential source of diagnostic biomarkers. Alterations in the composition and function of the lung microbiota are increasingly linked to lung cancer development [121]. Analyzing circulating microbial DNA from sources such as bronchoalveolar lavage fluid (BALF), thoracentesis samples, or liquid biopsies could serve as a non-invasive means of detecting early cancerous changes in fibrotic lungs [121].
Together with these molecular and microbial indicators, many of the key mediators involved in fibrotic remodeling also participate in carcinogenic progression, reinforcing their value as dual-purpose biomarkers. Several of these molecules—such as TGF-β, IL-6, VEGF, and MMPs—are summarized in the Supplementary Table S1 and exemplify how inflammatory signaling, extracellular matrix remodeling, and cellular transitions like EMT intersect in both fibrosis and malignant transformation. These shared mechanisms are strengthened by common pathways, including PI3K/AKT and Wnt/β-catenin, which not only drive fibrotic progression but also facilitate tumor invasion and metastasis. Collectively, this convergence highlights promising therapeutic opportunities, with biomarkers such as CXCL12, MMPs, and EMT-related factors offering avenues to disrupt processes that contribute to both pulmonary fibrosis and lung cancer (Table S1).

8. Clinical Implications and Future Directions

Integrating advanced genomic and proteomic technologies, such as large-panel next-generation sequencing (NGS) in conjunction with Molecular Tumor Boards (MTB), is vital for identifying comprehensive molecular profiles that guide precision medicine, particularly in fibrotic lungs with cancer risk [126]. This personalized approach facilitates the discovery of both established and emerging biomarkers, enabling more tailored and effective treatment strategies [126]. The development of non-invasive biomarkers from exosomes in biofluids offers additional promise for early disease detection and longitudinal monitoring, particularly for fibrotic lung conditions such as silicosis [127]. Meanwhile, artificial intelligence (AI) and machine learning are being employed to analyze complex biological datasets, uncovering novel biomarkers and therapeutic targets while addressing the interpretability challenges inherent to these models [126]. Future investigations will likely leverage transcriptomic and systems biology approaches to unravel the mechanisms underlying lung diseases and to identify new therapeutic avenues—especially for conditions like ischemia–reperfusion injury in lung transplantation [126]. The overarching goal is to achieve early prediction, accurate prognosis, and effective intervention strategies that improve patient outcomes by targeting the shared and distinct molecular pathways underlying lung fibrosis and lung cancer.
The close crosstalk between the development of PF and LC highlights the shared pathogenic mechanisms and key signaling molecules involved. It summarizes how oxidative and mitochondrial stress trigger DNA damage, genomic instability, and altered cell cycle control, leading to fibroblast activation and ECM accumulation in PF and enhanced proliferation and metastasis in LC. Cytokines such as TGF-β, IL-6, TNF-α, and IL-10, along with signaling cascades like PI3K/Akt/mTOR, Wnt/β-catenin, Hippo/YAP, and Hedgehog/Notch, mediate inflammation, EMT, and immune dysregulation. Key regulators, including GPX4, HOTAIR, USP15, USP39, TP53 (p53), and CDKN1A (p21), further connect ferroptosis, apoptosis, and metabolism. Together, these interactions highlight the molecular convergence of PF and LC and reveal potential shared therapeutic targets (Figure 4).

9. Conclusions

PF and LC, while clinically distinct, exhibit a complex interplay at the molecular level, driven by shared mechanisms that create a microenvironment conducive to malignant transformation. This review delineated these convergent pathways, emphasizing the roles of oxidative stress, DNA damage and repair, immune microenvironment remodeling, mitochondrial metabolism, and the ubiquitin–proteasome system in the pathogenesis of both diseases. We highlighted how persistent inflammation, genomic instability, and aberrant signaling pathways, such as TGF-β, Wnt, and PI3K/Akt/mTOR, contribute to fibrogenesis and tumorigenesis alike. Furthermore, we explored the impact of ECM remodeling, immune evasion, and altered cellular metabolism on disease progression and therapeutic resistance.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/cancers17233861/s1. Table S1: Key Molecular Mediators Linking Pulmonary Fibrosis and Lung Cancer [128,129,130,131,132,133,134,135,136,137,138,139,140,141].

Author Contributions

Writing—original draft preparation, B.C., Y.F., J.S. and P.C.; writing—review and editing, B.C., X.Z., H.H. and P.C.; funding acquisition, B.C., X.Z., Y.F., and J.S.; Conceptualization, Y.F., J.S., and P.C.; Y.F., J.S., and P.C. are responsible for the integrity of the work as a whole. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (Grant Nos. 32260167, 82260924, 82274374) and Yunnan Fundamental Research Projects (Grant Nos. 2019FH001-046, 2024J0026).

Acknowledgments

The authors gratefully acknowledge the valuable contributions of researchers whose work has informed this review. We also thank our respective institutions for providing support and resources that facilitated this study.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

  1. Raghu, G.; Collard, H.R.; Egan, J.J.; Martinez, F.J.; Behr, J.; Brown, K.K.; Colby, T.V.; Cordier, J.F.; Flaherty, K.R.; Lasky, J.A.; 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. [Google Scholar] [CrossRef]
  2. Wang, Y.; Wang, X.; Du, C.; Wang, Z.; Wang, J.; Zhou, N.; Wang, B.; Tan, K.; Fan, Y.; Cao, P. Glycolysis and beyond in glucose metabolism: Exploring pulmonary fibrosis at the metabolic crossroads. Front. Endocrinol. 2024, 15, 1379521. [Google Scholar] [CrossRef]
  3. Maher, T.M.; Bendstrup, E.; Dron, L.; Langley, J.; Smith, G.; Khalid, J.M.; Patel, H.; Kreuter, M. Global incidence and prevalence of idiopathic pulmonary fibrosis. Respir. Res. 2021, 22, 197. [Google Scholar] [CrossRef]
  4. Inamura, K. Lung Cancer: Understanding Its Molecular Pathology and the 2015 WHO Classification. Front. Oncol. 2017, 7, 193. [Google Scholar] [CrossRef]
  5. Jang, H.J.; Park, M.S.; Kim, Y.S.; Chang, J.; Lee, J.H.; Lee, C.T.; Lee, S.H.; Yoon, H.I. The relationship between the severity of pulmonary fibrosis and the lung cancer stage. J. Cancer 2021, 12, 2807–2814. [Google Scholar] [CrossRef] [PubMed]
  6. Thandra, K.C.; Barsouk, A.; Saginala, K.; Aluru, J.S.; Barsouk, A. Epidemiology of lung cancer. Contemp. Oncol. 2021, 25, 45–52. [Google Scholar]
  7. de Groot, P.M.; Wu, C.C.; Carter, B.W.; Munden, R.F. The epidemiology of lung cancer. Transl. Lung Cancer Res. 2018, 7, 220–233. [Google Scholar] [CrossRef]
  8. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  9. Czyżak, B.; Majewski, S. Concomitant Idiopathic Pulmonary Fibrosis and Lung Cancer: An Updated Narrative Review. Adv. Respir. Med. 2025, 93, 31. [Google Scholar] [CrossRef] [PubMed]
  10. de Totero, D.; Barisione, E.; Clini, E. Editorial: Pulmonary fibrosis and lung carcinogenesis: Do myofibroblasts and cancer-associated fibroblasts share a common identity? Front. Oncol. 2024, 14, 1389532. [Google Scholar] [CrossRef] [PubMed]
  11. Abu Qubo, A.; Numan, J.; Snijder, J.; Padilla, M.; Austin, J.H.M.; Capaccione, K.M.; Pernia, M.; Bustamante, J.; O’Connor, T.; Salvatore, M.M. Idiopathic pulmonary fibrosis and lung cancer: Future directions and challenges. Breathe 2022, 18, 220147. [Google Scholar] [CrossRef]
  12. Kotha, R.R.; Tareq, F.S.; Yildiz, E.; Luthria, D.L. Oxidative Stress and Antioxidants—A Critical Review on In Vitro Antioxidant Assays. Antioxidants 2022, 11, 2388. [Google Scholar] [CrossRef]
  13. Tzouvelekis, A.; Gomatou, G.; Bouros, E.; Trigidou, R.; Tzilas, V.; Bouros, D. Common Pathogenic Mechanisms Between Idiopathic Pulmonary Fibrosis and Lung Cancer. Chest 2019, 156, 383–391. [Google Scholar] [CrossRef]
  14. Chan, H.P.; Lewis, C.; Thomas, P.S. Oxidative stress and exhaled breath analysis: A promising tool for detection of lung cancer. Cancers 2010, 2, 32–42. [Google Scholar] [CrossRef] [PubMed]
  15. Moore, B.B.; Thannickal, V.J.; Toews, G.B. Bone Marrow-Derived Cells in the Pathogenesis of Lung Fibrosis. Curr. Respir. Med. Rev. 2005, 1, 69–76. [Google Scholar] [CrossRef] [PubMed]
  16. Inui, N.; Sakai, S.; Kitagawa, M. Molecular Pathogenesis of Pulmonary Fibrosis, with Focus on Pathways Related to TGF-beta and the Ubiquitin-Proteasome Pathway. Int. J. Mol. Sci. 2021, 22, 6107. [Google Scholar] [CrossRef]
  17. Liang, J.; Guan, X.; Bao, G.; Yao, Y.; Zhong, X. Molecular subtyping of small cell lung cancer. Semin. Cancer Biol. 2022, 86 Pt 2, 450–462. [Google Scholar] [CrossRef]
  18. Sainz de Aja, J.; Dost, A.F.M.; Kim, C.F. Alveolar progenitor cells and the origin of lung cancer. J. Intern. Med. 2021, 289, 629–635. [Google Scholar] [CrossRef] [PubMed]
  19. de Sousa, V.M.L.; Carvalho, L. Heterogeneity in Lung Cancer. Pathobiology 2018, 85, 96–107. [Google Scholar] [CrossRef]
  20. Liu, J.; Kang, R.; Tang, D. Signaling pathways and defense mechanisms of ferroptosis. FEBS J. 2022, 289, 7038–7050. [Google Scholar] [CrossRef]
  21. Srinivas, U.S.; Tan, B.W.Q.; Vellayappan, B.A.; Jeyasekharan, A.D. ROS and the DNA damage response in cancer. Redox Biol. 2019, 25, 101084. [Google Scholar] [CrossRef]
  22. Pannunzio, N.R.; Watanabe, G.; Lieber, M.R. Nonhomologous DNA end-joining for repair of DNA double-strand breaks. J. Biol. Chem. 2018, 293, 10512–10523. [Google Scholar] [CrossRef] [PubMed]
  23. Xiong, C.; Ling, H.; Huang, Y.; Dong, H.; Xie, B.; Hao, Q.; Zhou, X. AZD1775 synergizes with SLC7A11 inhibition to promote ferroptosis. Sci. China Life Sci. 2025, 68, 204–218. [Google Scholar] [CrossRef]
  24. Jiang, M.; Jia, K.; Wang, L.; Li, W.; Chen, B.; Liu, Y.; Wang, H.; Zhao, S.; He, Y.; Zhou, C. Alterations of DNA damage response pathway: Biomarker and therapeutic strategy for cancer immunotherapy. Acta Pharm. Sin. B 2021, 11, 2983–2994. [Google Scholar] [CrossRef]
  25. Li, L.; Yao, L.; Wang, M.; Zhou, X.; Xu, Y. Phase separation in DNA damage response: New insights into cancer development and therapy. Biochim. Biophys. Acta (BBA)-Rev. Cancer 2024, 1879, 189206. [Google Scholar] [CrossRef] [PubMed]
  26. Lahiri, A.; Maji, A.; Potdar, P.D.; Singh, N.; Parikh, P.; Bisht, B.; Mukherjee, A.; Paul, M.K. Lung cancer immunotherapy: Progress, pitfalls, and promises. Mol. Cancer 2023, 22, 40. [Google Scholar] [CrossRef]
  27. Li, H.; Zimmerman, S.E.; Weyemi, U. Genomic instability and metabolism in cancer. Int. Rev. Cell Mol. Biol. 2021, 364, 241–265. [Google Scholar]
  28. Gniadecki, R.; Iyer, A.; Hennessey, D.; Khan, L.; O’Keefe, S.; Redmond, D.; Storek, J.; Durand, C.; Cohen-Tervaert, J.W.; Osman, M. Genomic instability in early systemic sclerosis. J. Autoimmun. 2022, 131, 102847. [Google Scholar] [CrossRef] [PubMed]
  29. Huynh, J.; Chand, A.; Gough, D.; Ernst, M. Therapeutically exploiting STAT3 activity in cancer—Using tissue repair as a road map. Nat. Rev. Cancer 2019, 19, 82–96. [Google Scholar] [CrossRef]
  30. Cloonan, S.M.; Choi, A.M. Mitochondria in lung disease. J. Clin. Investig. 2016, 126, 809–820. [Google Scholar] [CrossRef]
  31. Evans, C.M.; Fingerlin, T.E.; Schwarz, M.I.; Lynch, D.; Kurche, J.; Warg, L.; Yang, I.V.; Schwartz, D.A. Idiopathic Pulmonary Fibrosis: A Genetic Disease That Involves Mucociliary Dysfunction of the Peripheral Airways. Physiol. Rev. 2016, 96, 1567–1591. [Google Scholar] [CrossRef] [PubMed]
  32. Armanios, M.Y.; Chen, J.J.; Cogan, J.D.; Alder, J.K.; Ingersoll, R.G.; Markin, C.; Lawson, W.E.; Xie, M.; Vulto, I.; Phillips, J.A., III; et al. Telomerase mutations in families with idiopathic pulmonary fibrosis. N. Engl. J. Med. 2007, 356, 1317–1326. [Google Scholar] [CrossRef] [PubMed]
  33. Shaw, A.T.; Felip, E.; Bauer, T.M.; Besse, B.; Navarro, A.; Postel-Vinay, S.; Gainor, J.F.; Johnson, M.; Dietrich, J.; James, L.P.; et al. Lorlatinib in non-small-cell lung cancer with ALK or ROS1 rearrangement: An international, multicentre, open-label, single-arm first-in-man phase 1 trial. Lancet Oncol. 2017, 18, 1590–1599. [Google Scholar] [CrossRef]
  34. Peddireddy, V.; Siva Prasad, B.; Gundimeda, S.D.; Penagaluru, P.R.; Mundluru, H.P. Assessment of 8-oxo-7, 8-dihydro-2′-deoxyguanosine and malondialdehyde levels as oxidative stress markers and antioxidant status in non-small cell lung cancer. Biomarkers 2012, 17, 261–268. [Google Scholar] [CrossRef]
  35. Estornut, C.; Milara, J.; Bayarri, M.A.; Belhadj, N.; Cortijo, J. Targeting Oxidative Stress as a Therapeutic Approach for Idiopathic Pulmonary Fibrosis. Front. Pharmacol. 2022, 12, 794997. [Google Scholar] [CrossRef] [PubMed]
  36. Mohanan, A.; Washimkar, K.R.; Mugale, M.N. Unraveling the interplay between vital organelle stress and oxidative stress in idiopathic pulmonary fibrosis. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2024, 1871, 119676. [Google Scholar] [CrossRef]
  37. Wang, M.-C. Natural plant resource flavonoids as potential therapeutic drugs for pulmonary fibrosis. Heliyon 2023, 9, e19308. [Google Scholar] [CrossRef] [PubMed]
  38. Yuan, Y.; Li, H.; Pu, W.; Chen, L.; Guo, D.; Jiang, H.; He, B.; Qin, S.; Wang, K.; Li, N.; et al. Cancer metabolism and tumor microenvironment: Fostering each other? Sci. China Life Sci. 2022, 65, 236–279. [Google Scholar] [CrossRef]
  39. Wynn, T.A.; Ramalingam, T.R. Mechanisms of fibrosis: Therapeutic translation for fibrotic disease. Nat. Med. 2012, 18, 1028–1040. [Google Scholar] [CrossRef]
  40. Fan, H.; Wang, Y.; Zhao, K.; Su, L.; Deng, C.; Huang, J.; Chen, G. Incomplete Knockdown of MyD88 Inhibits LPS-Induced Lung Injury and Lung Fibrosis in a Mouse Model. Inflammation 2023, 46, 2276–2288. [Google Scholar] [CrossRef]
  41. Shao, S.; Qu, Z.; Liang, Y.; Xu, Y.; Zhou, D.; Li, D.; Zhang, Y.; Yin, S. Iguratimod decreases bleomycin-induced pulmonary fibrosis in association with inhibition of TNF-alpha in mice. Int. Immunopharmacol. 2021, 99, 107936. [Google Scholar] [CrossRef]
  42. Le, T.T.; Karmouty-Quintana, H.; Melicoff, E.; Le, T.T.; Weng, T.; Chen, N.Y.; Pedroza, M.; Zhou, Y.; Davies, J.; Philip, K.; et al. Blockade of IL-6 Trans signaling attenuates pulmonary fibrosis. J. Immunol. 2014, 193, 3755–3768. [Google Scholar] [CrossRef]
  43. Li, Y.; Qin, W.; Liang, Q.; Zeng, J.; Yang, Q.; Chen, Y.; Wang, J.; Lu, W. Bufei huoxue capsule alleviates bleomycin-induced pulmonary fibrosis in mice via TGF-beta1/Smad2/3 signaling. J. Ethnopharmacol. 2023, 316, 116733. [Google Scholar] [CrossRef]
  44. Li, Y.; Sharma, A.; Schmidt-Wolf, I.G.H. Evolving insights into the improvement of adoptive T-cell immunotherapy through PD-1/PD-L1 blockade in the clinical spectrum of lung cancer. Mol. Cancer 2024, 23, 80. [Google Scholar] [CrossRef]
  45. Zhang, H.; Dai, Z.; Wu, W.; Wang, Z.; Zhang, N.; Zhang, L.; Zeng, W.J.; Liu, Z.; Cheng, Q. Regulatory mechanisms of immune checkpoints PD-L1 and CTLA-4 in cancer. J. Exp. Clin. Cancer Res. 2021, 40, 184. [Google Scholar] [CrossRef] [PubMed]
  46. Khan, M.; Arooj, S.; Wang, H. NK Cell-Based Immune Checkpoint Inhibition. Front. Immunol. 2020, 11, 167. [Google Scholar] [CrossRef] [PubMed]
  47. Han, S.; Wang, W.; Wang, S.; Yang, T.; Zhang, G.; Wang, D.; Ju, R.; Lu, Y.; Wang, H.; Wang, L. Tumor microenvironment remodeling and tumor therapy based on M2-like tumor associated macrophage-targeting nano-complexes. Theranostics 2021, 11, 2892–2916. [Google Scholar] [CrossRef]
  48. Shevach, E.M.; Thornton, A.M. tTregs, pTregs, and iTregs: Similarities and differences. Immunol. Rev. 2014, 259, 88–102. [Google Scholar] [CrossRef] [PubMed]
  49. Ma, H.Y.; Das, J.; Prendergast, C.; De Jong, D.; Braumuller, B.; Paily, J.; Huang, S.; Liou, C.; Giarratana, A.; Hosseini, M.; et al. Advances in CAR T Cell Therapy for Non-Small Cell Lung Cancer. Curr. Issues Mol. Biol. 2023, 45, 9019–9038. [Google Scholar] [CrossRef]
  50. Zhang, Z.; Jiang, J.; Wu, X.; Zhang, M.; Luo, D.; Zhang, R.; Li, S.; He, Y.; Bian, H.; Chen, Z. Chimeric antigen receptor T cell targeting EGFRvIII for metastatic lung cancer therapy. Front. Med. 2019, 13, 57–68. [Google Scholar] [CrossRef]
  51. Xu, T.; Karschnia, P.; Cadilha, B.L.; Dede, S.; Lorenz, M.; Seewaldt, N.; Nikolaishvili, E.; Muller, K.; Blobner, J.; Teske, N.; et al. In vivo dynamics and anti-tumor effects of EpCAM-directed CAR T-cells against brain metastases from lung cancer. Oncoimmunology 2023, 12, 2163781. [Google Scholar] [CrossRef]
  52. Avouac, J.; Cauvet, A.; Orvain, C.; Boulch, M.; Tilotta, F.; Tu, L.; Thuillet, R.; Ottaviani, M.; Guignabert, C.; Bousso, P.; et al. Effects of B Cell Depletion by CD19-Targeted Chimeric Antigen Receptor T Cells in a Murine Model of Systemic Sclerosis. Arthritis Rheumatol. 2024, 76, 268–278. [Google Scholar] [CrossRef] [PubMed]
  53. Chocarro, L.; Arasanz, H.; Fernandez-Rubio, L.; Blanco, E.; Echaide, M.; Bocanegra, A.; Teijeira, L.; Garnica, M.; Morilla, I.; Martinez-Aguillo, M.; et al. CAR-T Cells for the Treatment of Lung Cancer. Life 2022, 12, 561. [Google Scholar] [CrossRef]
  54. Srivastava, S.; Furlan, S.N.; Jaeger-Ruckstuhl, C.A.; Sarvothama, M.; Berger, C.; Smythe, K.S.; Garrison, S.M.; Specht, J.M.; Lee, S.M.; Amezquita, R.A.; et al. Immunogenic Chemotherapy Enhances Recruitment of CAR-T Cells to Lung Tumors and Improves Antitumor Efficacy when Combined with Checkpoint Blockade. Cancer Cell 2021, 39, 193–208.e110. [Google Scholar] [CrossRef]
  55. Kythreotou, A.; Siddique, A.; Mauri, F.A.; Bower, M.; Pinato, D.J. PD-L1. J. Clin. Pathol. 2018, 71, 189–194. [Google Scholar] [CrossRef]
  56. Francisco, L.M.; Sage, P.T.; Sharpe, A.H. The PD-1 pathway in tolerance and autoimmunity. Immunol. Rev. 2010, 236, 219–242. [Google Scholar] [CrossRef]
  57. Xia, L.; Wang, H.; Sun, M.; Yang, Y.; Yao, C.; He, S.; Duan, H.; Xia, W.; Sun, R.; Yao, Y.; et al. Peripheral CD4+ T cell signatures in predicting the responses to anti-PD-1/PD-L1 monotherapy for Chinese advanced non-small cell lung cancer. Sci. China Life Sci. 2021, 64, 1590–1601. [Google Scholar] [CrossRef] [PubMed]
  58. Biernacka, A.; Dobaczewski, M.; Frangogiannis, N.G. TGF-β signaling in fibrosis. Growth Factors 2011, 29, 196–202. [Google Scholar] [CrossRef]
  59. Miyazono, K.; Ehata, S.; Koinuma, D. Tumor-promoting functions of transforming growth factor-β in progression of cancer. Upsala J. Med. Sci. 2012, 117, 143–152. [Google Scholar] [CrossRef] [PubMed]
  60. Chambers, D.M.; Moretti, L.; Zhang, J.J.; Cooper, S.W.; Chambers, D.M.; Santangelo, P.J.; Barker, T.H. LEM domain-containing protein 3 antagonizes TGFbeta-SMAD2/3 signaling in a stiffness-dependent manner in both the nucleus and cytosol. J. Biol. Chem. 2018, 293, 15867–15886. [Google Scholar] [CrossRef]
  61. Watson, W.H.; Ritzenthaler, J.D.; Roman, J. Lung extracellular matrix and redox regulation. Redox Biol. 2016, 8, 305–315. [Google Scholar] [CrossRef] [PubMed]
  62. Herrera, J.; Henke, C.A.; Bitterman, P.B. Extracellular matrix as a driver of progressive fibrosis. J. Clin. Investig. 2018, 128, 45–53. [Google Scholar] [CrossRef] [PubMed]
  63. Kristensen, J.H.; Karsdal Ma Genovese, F.; Johnson, S.; Svensson, B.; Jacobsen, S.; Hägglund, P.; Leeming, D.J. The role of extracellular matrix quality in pulmonary fibrosis. Respir. Int. Rev. Thorac. Dis. 2014, 88, 487–499. [Google Scholar] [CrossRef]
  64. Chaudhry, G.E.; Akim, A.M.; Sung, Y.Y.; Muhammad, T.S.T. Cancer and Apoptosis. Methods Mol. Biol. 2022, 2543, 191–210. [Google Scholar]
  65. Najafi, M.; Farhood, B.; Mortezaee, K. Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J. Cell Biochem. 2019, 120, 2782–2790. [Google Scholar] [CrossRef]
  66. Xia, B.; Qiu, L.; Yue, J.; Si, J.; Zhang, H. The metabolic crosstalk of cancer-associated fibroblasts and tumor cells: Recent advances and future perspectives. Biochim. Biophys. Acta Rev. Cancer 2024, 1879, 189190. [Google Scholar] [CrossRef]
  67. Du, H.; Che, G. Advancement of relationship between metabolic alteration in cancer-associated fibroblasts and tumor progression in lung cancer. Zhongguo Fei Ai Za Zhi 2014, 17, 679–684. [Google Scholar]
  68. Li, M.; Cheng, X.; Rong, R.; Gao, Y.; Tang, X.; Chen, Y. High expression of fibroblast activation protein (FAP) predicts poor outcome in high-grade serous ovarian cancer. BMC Cancer 2020, 20, 1032. [Google Scholar] [CrossRef]
  69. Di Martino, J.S.; Nobre, A.R.; Mondal, C.; Taha, I.; Farias, E.F.; Fertig, E.J.; Naba, A.; Aguirre-Ghiso, J.A.; Bravo-Cordero, J.J. A tumor-derived type III collagen-rich ECM niche regulates tumor cell dormancy. Nat. Cancer 2022, 3, 90–107. [Google Scholar] [CrossRef]
  70. Yuan, Z.; Li, Y.; Zhang, S.; Wang, X.; Dou, H.; Yu, X.; Zhang, Z.; Yang, S.; Xiao, M. Extracellular matrix remodeling in tumor progression and immune escape: From mechanisms to treatments. Mol. Cancer 2023, 22, 48. [Google Scholar] [CrossRef]
  71. Fang, Y.; Tian, J.; Fan, Y.; Cao, P. Latest progress on the molecular mechanisms of idiopathic pulmonary fibrosis. Mol. Biol. Rep. 2020, 47, 9811–9820. [Google Scholar] [CrossRef]
  72. Zhao, Y.; Yang, Z.Q.; Wang, Y.; Miao, Y.; Liu, Y.; Dai, S.D.; Han, Y.; Wang, E.H. Dishevelled-1 and dishevelled-3 affect cell invasion mainly through canonical and noncanonical Wnt pathway, respectively, and associate with poor prognosis in nonsmall cell lung cancer. Mol. Carcinog. 2010, 49, 760–770. [Google Scholar] [CrossRef]
  73. Alrumaihi, F.; Rahmani, A.H.; Prabhu, S.V.; Kumar, V.; Anwar, S. The Role of Plant-Derived Natural Products as a Regulator of the Tyrosine Kinase Pathway in the Management of Lung Cancer. Curr. Issues Mol. Biol. 2025, 47, 498. [Google Scholar] [CrossRef] [PubMed]
  74. Ningthoujam, R.; Chanu, N.B.; Anumala, V.; Babu, P.J.; Pradhan, S.; Panda, M.K.; Heisnam, P.; Singh, Y.D. Predicting Biomarkers and Therapeutic Targets in Cancer. Stud. Comput. Intell. 2022, 1016, 217–232. [Google Scholar]
  75. Buachan, P.; Namsa-Aid, M.; Sung, H.K.; Peng, C.; Sweeney, G.; Tanechpongtamb, W. Inhibitory effects of terrein on lung cancer cell metastasis and angiogenesis. Oncol. Rep. 2021, 45, 94. [Google Scholar] [CrossRef]
  76. Guy, S.; Geletu, M.; Arulanandam, R.; Raptis, L. Stat3 and gap junctions in normal and lung cancer cells. Cancers 2014, 6, 646–662. [Google Scholar] [CrossRef]
  77. Wang, T.H.; Huang, K.Y.; Chen, C.C.; Chang, Y.H.; Chen, H.Y.; Hsueh, C.; Liu, Y.T.; Yang, S.C.; Yang, P.C.; Chen, C.Y. PM2.5 promotes lung cancer progression through activation of the AhR-TMPRSS2-IL18 pathway. EMBO Mol. Med. 2023, 15, e17014. [Google Scholar] [CrossRef]
  78. Hamamoto, J.; Yasuda, H.; Nonaka, Y.; Fujiwara, M.; Nakamura, Y.; Soejima, K.; Betsuyaku, T. The FGF2 aptamer inhibits the growth of FGF2-FGFR pathway driven lung cancer cells. Biochem. Biophys. Res. Commun. 2018, 503, 1330–1334. [Google Scholar] [CrossRef] [PubMed]
  79. Iqbal, N.; Iqbal, N. Human Epidermal Growth Factor Receptor 2 (HER2) in Cancers: Overexpression and Therapeutic Implications. Mol. Biol. Int. 2014, 2014, 852748. [Google Scholar] [CrossRef]
  80. Lv, S.; Yang, J.; Lin, J.; Huang, X.; Zhao, H.; Zhao, C.; Yang, L. CDK4/6 inhibitors in lung cancer: Current practice and future directions. Eur. Respir. Rev. 2024, 33, 230145. [Google Scholar] [CrossRef]
  81. Mender, I.; LaRanger, R.; Luitel, K.; Peyton, M.; Girard, L.; Lai, T.P.; Batten, K.; Cornelius, C.; Dalvi, M.P.; Ramirez, M.; et al. Telomerase-Mediated Strategy for Overcoming Non-Small Cell Lung Cancer Targeted Therapy and Chemotherapy Resistance. Neoplasia 2018, 20, 826–837. [Google Scholar] [CrossRef]
  82. Ding, X.; Cheng, J.; Pang, Q.; Wei, X.; Zhang, X.; Wang, P.; Yuan, Z.; Qian, D. BIBR1532, a Selective Telomerase Inhibitor, Enhances Radiosensitivity of Non-Small Cell Lung Cancer Through Increasing Telomere Dysfunction and ATM/CHK1 Inhibition. Int. J. Radiat. Oncol. Biol. Phys. 2019, 105, 861–874. [Google Scholar] [CrossRef]
  83. Rotem, O.; Zer, A.; Yosef, L.; Beery, E.; Goldvaser, H.; Gutkin, A.; Levin, R.; Dudnik, E.; Berger, T.; Feinmesser, M.; et al. Blood-Derived Exosomal hTERT mRNA in Patients with Lung Cancer: Characterization and Correlation with Response to Therapy. Biomedicines 2023, 11, 1730. [Google Scholar] [CrossRef] [PubMed]
  84. De Vitis, M.; Berardinelli, F.; Sgura, A. Telomere Length Maintenance in Cancer: At the Crossroad between Telomerase and Alternative Lengthening of Telomeres (ALT). Int. J. Mol. Sci. 2018, 19, 606. [Google Scholar] [CrossRef] [PubMed]
  85. Guterres, A.N.; Villanueva, J. Targeting telomerase for cancer therapy. Oncogene 2020, 39, 5811–5824. [Google Scholar] [CrossRef] [PubMed]
  86. Liu, T.; Ullenbruch, M.; Young Choi, Y.; Yu, H.; Ding, L.; Xaubet, A.; Pereda, J.; Feghali-Bostwick, C.A.; Bitterman, P.B.; Henke, C.A.; et al. Telomerase and telomere length in pulmonary fibrosis. Am. J. Respir. Cell Mol. Biol. 2013, 49, 260–268. [Google Scholar] [CrossRef]
  87. van Batenburg, A.A.; Kazemier, K.M.; van Oosterhout, M.F.M.; van der Vis, J.J.; Grutters, J.C.; Goldschmeding, R.; van Moorsel, C.H.M. Telomere shortening and DNA damage in culprit cells of different types of progressive fibrosing interstitial lung disease. ERJ Open Res. 2021, 7, 00691-2020. [Google Scholar] [CrossRef]
  88. Arish, N.; Petukhov, D.; Wallach-Dayan, S.B. The Role of Telomerase and Telomeres in Interstitial Lung Diseases: From Molecules to Clinical Implications. Int. J. Mol. Sci. 2019, 20, 2996. [Google Scholar] [CrossRef]
  89. Lv, X.; Liu, C.; Liu, S.; Li, Y.; Wang, W.; Li, K.; Hua, F.; Cui, B.; Zhang, X.; Yu, J.; et al. The cell cycle inhibitor P21 promotes the development of pulmonary fibrosis by suppressing lung alveolar regeneration. Acta Pharm. Sin. B 2022, 12, 735–746. [Google Scholar] [CrossRef]
  90. Luzina, I.G.; Rus, V.; Lockatell, V.; Courneya, J.P.; Hampton, B.S.; Fishelevich, R.; Misharin, A.V.; Todd, N.W.; Badea, T.C.; Rus, H.; et al. Regulator of Cell Cycle Protein (RGCC/RGC-32) Protects against Pulmonary Fibrosis. Am. J. Respir. Cell Mol. Biol. 2022, 66, 146–157. [Google Scholar] [CrossRef]
  91. Lv, L.; Zhou, M.; Zhang, J.; Liu, F.; Qi, L.; Zhang, S.; Bi, Y.; Yu, Y. SOX6 suppresses the development of lung adenocarcinoma by regulating expression of p53, p21(CIPI), cyclin D1 and beta-catenin. FEBS Open Bio 2020, 10, 135–146. [Google Scholar] [CrossRef]
  92. Liu, M.; Zhang, H.; Li, Y.; Wang, R.; Li, Y.; Zhang, H.; Ren, D.; Liu, H.; Kang, C.; Chen, J. HOTAIR, a long noncoding RNA, is a marker of abnormal cell cycle regulation in lung cancer. Cancer Sci. 2018, 109, 2717–2733. [Google Scholar] [CrossRef] [PubMed]
  93. Sen, T.; Tong, P.; Diao, L.; Li, L.; Fan, Y.; Hoff, J.; Heymach, J.V.; Wang, J.; Byers, L.A. Targeting AXL and mTOR Pathway Overcomes Primary and Acquired Resistance to WEE1 Inhibition in Small-Cell Lung Cancer. Clin. Cancer Res. 2017, 23, 6239–6253. [Google Scholar] [CrossRef]
  94. Ramkumar, K.; Tanimoto, A.; Della Corte, C.M.; Stewart, C.A.; Wang, Q.; Shen, L.; Cardnell, R.J.; Wang, J.; Polanska, U.M.; Andersen, C.; et al. Targeting BCL2 Overcomes Resistance and Augments Response to Aurora Kinase B Inhibition by AZD2811 in Small Cell Lung Cancer. Clin. Cancer Res. 2023, 29, 3237–3249. [Google Scholar] [CrossRef]
  95. Lucarini, L.; Durante, M.; Lanzi, C.; Pini, A.; Boccalini, G.; Calosi, L.; Moroni, F.; Masini, E.; Mannaioni, G. HYDAMTIQ, a selective PARP-1 inhibitor, improves bleomycin-induced lung fibrosis by dampening the TGF-beta/SMAD signalling pathway. J. Cell Mol. Med. 2017, 21, 324–335. [Google Scholar] [CrossRef]
  96. Rose, M.; Burgess, J.T.; O’Byrne, K.; Richard, D.J.; Bolderson, E. PARP Inhibitors: Clinical Relevance, Mechanisms of Action and Tumor Resistance. Front. Cell Dev. Biol. 2020, 8, 564601. [Google Scholar] [CrossRef] [PubMed]
  97. Pachimatla, A.G.; Fenstermaker, R.; Ciesielski, M.; Yendamuri, S. Survivin in lung cancer: A potential target for therapy and prevention-a narrative review. Transl. Lung Cancer Res. 2024, 13, 362–374. [Google Scholar] [CrossRef]
  98. Chanda, D.; Otoupalova, E.; Smith, S.R.; Volckaert, T.; De Langhe, S.P.; Thannickal, V.J. Developmental pathways in the pathogenesis of lung fibrosis. Mol. Asp. Med. 2019, 65, 56–69. [Google Scholar] [CrossRef] [PubMed]
  99. ArulJothi, K.N.; Kumaran, K.; Senthil, S.; Nidhu, A.B.; Munaff, N.; Janitri, V.B.; Kirubakaran, R.; Singh, S.K.; Gupt, G.; Dua, K.; et al. Implications of reactive oxygen species in lung cancer and exploiting it for therapeutic interventions. Med. Oncol. 2022, 40, 43. [Google Scholar] [CrossRef]
  100. Javadov, S. Mitochondria and ferroptosis. Curr. Opin. Physiol. 2022, 25, 100483. [Google Scholar] [CrossRef]
  101. Liu, D.; He, W.; Yang, L.-L. Revitalizing antitumor immunity: Leveraging nucleic acid sensors as therapeutic targets. Cancer Lett. 2024, 588, 216729. [Google Scholar] [CrossRef]
  102. Xiong, R.; He, R.; Liu, B.; Jiang, W.; Wang, B.; Li, N.; Geng, Q. Ferroptosis: A New Promising Target for Lung Cancer Therapy. Oxid. Med. Cell Longev. 2021, 2021, 8457521. [Google Scholar] [CrossRef]
  103. Cheng, H.; Feng, D.; Li, X.; Gao, L.; Tang, S.; Liu, W.; Wu, X.; Yue, S.; Li, C.; Luo, Z. Iron deposition-induced ferroptosis in alveolar type II cells promotes the development of pulmonary fibrosis. Biochim. Biophys. Acta Mol. Basis Dis. 2021, 1867, 166204. [Google Scholar] [CrossRef]
  104. Liu, W.; Zhou, Y.; Duan, W.; Song, J.; Wei, S.; Xia, S.; Wang, Y.; Du, X.; Li, E.; Ren, C.; et al. Glutathione peroxidase 4-dependent glutathione high-consumption drives acquired platinum chemoresistance in lung cancer-derived brain metastasis. Clin. Transl. Med. 2021, 11, e517. [Google Scholar] [CrossRef]
  105. Ling, H.; Xiao, H.; Luo, T.; Lin, H.; Deng, J. Role of Ferroptosis in Regulating the Epithelial-Mesenchymal Transition in Pulmonary Fibrosis. Biomedicines 2023, 11, 163. [Google Scholar] [CrossRef] [PubMed]
  106. Ma, J.T.; Zhang, X.Y.; Cao, R.; Sun, L.; Jing, W.; Zhao, J.Z.; Zhang, S.L.; Huang, L.T.; Han, C.B. Effects of Dynamin-related Protein 1 Regulated Mitochondrial Dynamic Changes on Invasion and Metastasis of Lung Cancer Cells. J. Cancer 2019, 10, 4045–4053. [Google Scholar] [CrossRef]
  107. Eiyama, A.; Okamoto, K. PINK1/Parkin-mediated mitophagy in mammalian cells. Curr. Opin. Cell Biol. 2015, 33, 95–101. [Google Scholar] [CrossRef]
  108. Sun, Y.; Shen, W.; Hu, S.; Lyu, Q.; Wang, Q.; Wei, T.; Zhu, W.; Zhang, J. METTL3 promotes chemoresistance in small cell lung cancer by inducing mitophagy. J. Exp. Clin. Cancer Res. 2023, 42, 65. [Google Scholar] [CrossRef] [PubMed]
  109. Tsubouchi, K.; Araya, J.; Kuwano, K. PINK1-PARK2-mediated mitophagy in COPD and IPF pathogeneses. Inflamm. Regen. 2018, 38, 18. [Google Scholar] [CrossRef] [PubMed]
  110. Li, S.; Zhang, H.; Chang, J.; Li, D.; Cao, P. Iron overload and mitochondrial dysfunction orchestrate pulmonary fibrosis. Eur. J. Pharmacol. 2021, 912, 174613. [Google Scholar] [CrossRef]
  111. Bueno, M.; Lai, Y.C.; Romero, Y.; Brands, J.; St Croix, C.M.; Kamga, C.; Corey, C.; Herazo-Maya, J.D.; Sembrat, J.; Lee, J.S.; et al. PINK1 deficiency impairs mitochondrial homeostasis and promotes lung fibrosis. J. Clin. Investig. 2015, 125, 521–538. [Google Scholar] [CrossRef]
  112. Weiss, C.H.; Budinger, G.R.; Mutlu, G.M.; Jain, M. Proteasomal regulation of pulmonary fibrosis. Proc. Am. Thorac. Soc. 2010, 7, 77–83. [Google Scholar] [CrossRef]
  113. Fhu, C.W.; Ali, A.A. Dysregulation of the Ubiquitin Proteasome System in Human Malignancies: A Window for Therapeutic Intervention. Cancers 2021, 13, 1513. [Google Scholar] [CrossRef] [PubMed]
  114. Chen, W.; Ni, D.; Zhang, H.; Li, X.; Jiang, Y.; Wu, J.; Gu, Y.; Gao, M.; Shi, W.; Song, J.; et al. Over-expression of USP15/MMP3 predict poor prognosis and promote growth, migration in non-small cell lung cancer cells. Cancer Genet. 2023, 272–273, 9–15. [Google Scholar] [CrossRef] [PubMed]
  115. Yuan, J.; Zhang, G.; Li, X.; Ma, Q.; Cheng, W.; Wang, W.; Zhang, B.; Hu, T.; Song, G. Knocking down USP39 Inhibits the Growth and Metastasis of Non-Small-Cell Lung Cancer Cells through Activating the p53 Pathway. Int. J. Mol. Sci. 2020, 21, 8949. [Google Scholar] [CrossRef] [PubMed]
  116. Shkedy, D.; Gonen, H.; Bercovich, B.; Ciechanover, A. Complete reconstitution of conjugation and subsequent degradation of the tumor suppressor protein p53 by purified components of the ubiquitin proteolytic system. FEBS Lett. 1994, 348, 126–130. [Google Scholar] [CrossRef]
  117. Han, Y.H.; Park, W.H. MG132 as a proteasome inhibitor induces cell growth inhibition and cell death in A549 lung cancer cells via influencing reactive oxygen species and GSH level. Hum. Exp. Toxicol. 2010, 29, 607–614. [Google Scholar] [CrossRef]
  118. Semren, N.; Welk, V.; Korfei, M.; Keller, I.E.; Fernandez, I.E.; Adler, H.; Gunther, A.; Eickelberg, O.; Meiners, S. Regulation of 26S Proteasome Activity in Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2015, 192, 1089–1101. [Google Scholar] [CrossRef]
  119. Semren, N.; Habel-Ungewitter, N.C.; Fernandez, I.E.; Konigshoff, M.; Eickelberg, O.; Stoger, T.; Meiners, S. Validation of the 2nd Generation Proteasome Inhibitor Oprozomib for Local Therapy of Pulmonary Fibrosis. PLoS ONE 2015, 10, e0136188. [Google Scholar] [CrossRef]
  120. Schwartz, A.L.; Ciechanover, A. Targeting proteins for destruction by the ubiquitin system: Implications for human pathobiology. Annu. Rev. Pharmacol. Toxicol. 2009, 49, 73–96. [Google Scholar] [CrossRef]
  121. Papasotiriou, I.; Parsonidis, P.; Ntanovasilis, D.-A.; Iliopoulos, A.C.; Beis, G.; Apostolou, P. Identification of new targets and biomarkers in lung cancer. J. Clin. Oncol. 2019, 37, e14656. [Google Scholar] [CrossRef]
  122. Jeon, J.E.; Rajapaksa, Y.; Keshavjee, S.; Liu, M. Applications of transcriptomics in ischemia reperfusion research in lung transplantation. J. Heart Lung Transplant. 2024, 43, 1501–1513. [Google Scholar] [CrossRef]
  123. Liu, M.; Yu, X.; Qu, C.; Xu, S. Predictive Value of Gene Databases in Discovering New Biomarkers and New Therapeutic Targets in Lung Cancer. Medicina 2023, 59, 547. [Google Scholar] [CrossRef]
  124. Paul, S.; Konig, M.F.; Pardoll, D.M.; Bettegowda, C.; Papadopoulos, N.; Wright, K.M.; Gabelli, S.B.; Ho, M.; van Elsas, A.; Zhou, S. Cancer therapy with antibodies. Nat. Rev. Cancer 2024, 24, 399–426. [Google Scholar] [CrossRef]
  125. Kaushik, A.C.; Mehmood, A.; Wei, D.-Q.; Dai, X. Systems Biology Integration and Screening of Reliable Prognostic Markers to Create Synergies in the Control of Lung Cancer Patients. Front. Mol. Biosci. 2020, 7, 47. [Google Scholar] [CrossRef]
  126. D’Agnano, V.; Mariniello, D.F.; Ruotolo, M.; Quarcio, G.; Moriello, A.; Conte, S.; Sorrentino, A.; Sanduzzi Zamparelli, S.; Bianco, A.; Perrotta, F. Targeting Progression in Pulmonary Fibrosis: An Overview of Underlying Mechanisms, Molecular Biomarkers, and Therapeutic Intervention. Life 2024, 14, 229. [Google Scholar] [CrossRef]
  127. Gupta, G.; Goyal, A.; Ilma, B.; Rekha, M.M.; Nayak, P.P.; Kaur, M.; Khachi, A.; Goyal, K.; Rana, M.; Rekha, A.; et al. Exosomal miRNAs as biomarkers and therapeutic targets in silicosis-related lung fibrosis. Mol. Biol. Rep. 2025, 52, 585. [Google Scholar] [CrossRef]
  128. Moog, M.; Aschenbrenner, F.; Jonigk, D.; Prinz, I.; Welte, T.; Kolb, M.; Maus, U.A. Role of IL17 A/F in experimental lung fibrosis. Eur. Respir. J. 2022, 60, 1726. [Google Scholar] [CrossRef]
  129. Aversa, Z.; Atkinson, E.J.; Carmona, E.M.; White, T.A.; Heeren, A.A.; Jachim, S.K.; Zhang, X.; Cummings, S.R.; Chiarella, S.E.; Limper, A.H.; et al. Biomarkers of cellular senescence in idiopathic pulmonary fibrosis. Respir. Res. 2023, 24, 101. [Google Scholar] [CrossRef] [PubMed]
  130. Wahlund, C.J.E.; Eklund, A.; Grunewald, J.; Gabrielsson, S. Pulmonary Extracellular Vesicles as Mediators of Local and Systemic Inflammation. Front. Cell Dev. Biol. 2017, 5, 39. [Google Scholar] [CrossRef] [PubMed]
  131. Yang, L.; Zhou, F.; Zheng, D.; Wang, D.; Li, X.; Zhao, C.; Huang, X. FGF/FGFR signaling: From lung development to respiratory diseases. Cytokine Growth Factor Rev. 2021, 62, 94–104. [Google Scholar] [CrossRef]
  132. Zanoaga, O.; Braicu, C.; Jurj, A.; Rusu, A.; Buiga, R.; Berindan-Neagoe, I. Progress in Research on the Role of Flavonoids in Lung Cancer. Int. J. Mol. Sci. 2019, 20, 4291. [Google Scholar] [CrossRef]
  133. Suryadevara, V.; Ramchandran, R.; Kamp, D.W.; Natarajan, V. Lipid Mediators Regulate Pulmonary Fibrosis: Potential Mechanisms and Signaling Pathways. Int. J. Mol. Sci. 2020, 21, 4257. [Google Scholar] [CrossRef] [PubMed]
  134. Wu, T.; Dong, Y.; Yang, X.; Mo, L.; You, Y. Crosstalk between lncRNAs and Wnt/β-catenin signaling pathways in lung cancers: From cancer progression to therapeutic response. Non-Coding RNA Res. 2024, 9, 667–677. [Google Scholar] [CrossRef]
  135. Lei, Y.; Zhong, C.; Zhang, J.; Zheng, Q.; Xu, Y.; Li, Z.; Huang, C.; Ren, T. Senescent lung fibroblasts in idiopathic pulmonary fibrosis facilitate non-small cell lung cancer progression by secreting exosomal MMP1. Oncogene 2024, 44, 769–781. [Google Scholar] [CrossRef]
  136. Yi, J.-P.; Zeng, M.; He, X. Role of sphingolipids signaling in pathogenesis of pulmonary fibrosis. Chin. J. Pharmacol. Toxicol. 2016, 30, 158–164. [Google Scholar] [CrossRef]
  137. Zhang, M.; Wang, W.; Liu, K.; Jia, C.; Hou, Y.; Bai, G. Astragaloside IV protects against lung injury and pulmonary fibrosis in COPD by targeting GTP-GDP domain of RAS and downregulating the RAS/RAF/FoxO signaling pathway. Phytomedicine 2023, 120, 155066. [Google Scholar] [CrossRef]
  138. Daley, R.; Maddipatla, V.; Ghosh, S.; Chowdhury, O.; Hose, S.; Zigler, J.S.; Sinha, D.; Liu, H. Aberrant Akt2 signaling in the RPE may contribute to retinal fibrosis process in diabetic retinopathy. Cell Death Discov. 2023, 9, 243. [Google Scholar] [CrossRef] [PubMed]
  139. Anajafi, S.; Hadavi, R.; Valizadeh-Otaghsara, S.M.; Hemmati, M.; Hassani, M.; Mohammadi-Yeganeh, S.; Soleimani, M. A functional interplay between non-coding RNAs and cancer-associated fibroblasts in breast cancer. Gene Rep. 2024, 36, 101990. [Google Scholar] [CrossRef]
  140. Bodmer, N.K.; Choudhury, M.; Mirza, H.; Pradhan, S.; Yin, Y.; Mecham, R.P.; Brody, S.L.; Ornitz, D.M.; Koenitzer, J.R. Latent transforming growth factor binding protein-2 (LTBP2), an IPF biomarker of clinical decline, promotes TGF-beta signaling and lung fibrosis in mice. bioRxiv 2025. [Google Scholar] [CrossRef]
  141. Liao, Y.; Yang, Y.; Zhou, G.; Chen, L.; Yang, Y.; Guo, S.; Zuo, Q.; Zou, J. Anoikis and SPP1 in idiopathic pulmonary fibrosis: Integrating bioinformatics, cell, and animal studies to explore prognostic biomarkers and PI3K/AKT signaling regulation. Expert Rev. Clin. Immunol. 2024, 20, 679–693. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Conceptual overview of molecular mechanisms linking pulmonary fibrosis (PF) to lung cancer (LC). Chronic PF induces persistent oxidative stress, mitochondrial dysfunction, DNA damage, and remodeling of the extracellular matrix (ECM), which together create a pro-tumorigenic microenvironment. These processes may promote epithelial–mesenchymal transition (EMT), genomic instability, and chronic inflammation, facilitating LC initiation. Key signaling mediators include TGF-β, PD-1/PD-L1, and components of the tumor immune microenvironment, which contribute to immune evasion and tumor progression. References supporting each pathway are included in the main text.
Figure 1. Conceptual overview of molecular mechanisms linking pulmonary fibrosis (PF) to lung cancer (LC). Chronic PF induces persistent oxidative stress, mitochondrial dysfunction, DNA damage, and remodeling of the extracellular matrix (ECM), which together create a pro-tumorigenic microenvironment. These processes may promote epithelial–mesenchymal transition (EMT), genomic instability, and chronic inflammation, facilitating LC initiation. Key signaling mediators include TGF-β, PD-1/PD-L1, and components of the tumor immune microenvironment, which contribute to immune evasion and tumor progression. References supporting each pathway are included in the main text.
Cancers 17 03861 g001
Figure 2. Immune response and extracellular matrix (ECM) dysregulation in pulmonary fibrosis (PF) and lung cancer (LC). This schematic illustrates the interplay between immune responses and ECM remodeling in PF and LC. In PF, injured lung epithelial cells release inflammatory mediators, including TNF-α, IL-6, and TGF-β, which activate fibroblasts and myofibroblasts, leading to excessive ECM deposition and increased tissue stiffness. In LC, immune evasion mechanisms disrupt T cell function (reduced proliferation, cytokine production, and cytotoxicity) and impair natural killer (NK) cell recognition and killing through altered surface markers and inhibitory factor secretion. Macrophage polarization is skewed toward the M2 phenotype, further promoting a pro-tumorigenic microenvironment. This figure highlights the shared mechanisms by which immune dysregulation and ECM remodeling contribute to both fibrogenesis and tumor progression.
Figure 2. Immune response and extracellular matrix (ECM) dysregulation in pulmonary fibrosis (PF) and lung cancer (LC). This schematic illustrates the interplay between immune responses and ECM remodeling in PF and LC. In PF, injured lung epithelial cells release inflammatory mediators, including TNF-α, IL-6, and TGF-β, which activate fibroblasts and myofibroblasts, leading to excessive ECM deposition and increased tissue stiffness. In LC, immune evasion mechanisms disrupt T cell function (reduced proliferation, cytokine production, and cytotoxicity) and impair natural killer (NK) cell recognition and killing through altered surface markers and inhibitory factor secretion. Macrophage polarization is skewed toward the M2 phenotype, further promoting a pro-tumorigenic microenvironment. This figure highlights the shared mechanisms by which immune dysregulation and ECM remodeling contribute to both fibrogenesis and tumor progression.
Cancers 17 03861 g002
Figure 3. The role of mitochondrial metabolism in pulmonary fibrosis (PF) and lung cancer (LC). This schematic depicts the contribution of mitochondrial metabolism to PF and LC pathogenesis. In PF, iron accumulation (Fe2+) and ferroptosis drive alveolar epithelial type II (AECII) cell death, promoting extracellular matrix (ECM) deposition and tissue fibrosis. In LC, mitochondrial dysfunction is associated with decreased GPX4 expression, enhancing ferroptosis resistance and chemoresistance, while increased Drp1 expression promotes mitochondrial fission to support tumor growth. Dysregulated expression of PINK1 and PARK2 in both conditions contributes to abnormal mitochondrial morphology, impaired mitophagy, and elevated reactive oxygen species (ROS and mtROS) production. This figure highlights how mitochondrial dysregulation links cellular stress, ferroptosis, and disease progression in PF and LC.
Figure 3. The role of mitochondrial metabolism in pulmonary fibrosis (PF) and lung cancer (LC). This schematic depicts the contribution of mitochondrial metabolism to PF and LC pathogenesis. In PF, iron accumulation (Fe2+) and ferroptosis drive alveolar epithelial type II (AECII) cell death, promoting extracellular matrix (ECM) deposition and tissue fibrosis. In LC, mitochondrial dysfunction is associated with decreased GPX4 expression, enhancing ferroptosis resistance and chemoresistance, while increased Drp1 expression promotes mitochondrial fission to support tumor growth. Dysregulated expression of PINK1 and PARK2 in both conditions contributes to abnormal mitochondrial morphology, impaired mitophagy, and elevated reactive oxygen species (ROS and mtROS) production. This figure highlights how mitochondrial dysregulation links cellular stress, ferroptosis, and disease progression in PF and LC.
Cancers 17 03861 g003
Figure 4. Integrated Pathogenic Pathways Linking Mitochondrial Dysfunction, Epithelial Damage, Fibroblast Activation, and Tumor Progression in the Tumor Microenvironment. Mitochondrial stress increases mitoROS, triggering ATM/ATR–p53 signaling, cell-cycle arrest, senescence, and NF-κB-mediated chronic inflammation. In epithelial cells, DNA damage and p53 activation enhance EMT transcription factors (SMADs, SNAIL, TWIST, ZEB1), MMP production, and ECM degradation, driving invasion and metastasis. TGF-β-induced fibroblast activation generates α-SMA+ CAFs, which promote ECM remodeling and immunosuppressive cytokine release (IL-6, TGF-β, CXCL12). Tumor cells amplify NF-κB/STAT3 signaling, increase PD-L1 expression, recruit Tregs, and induce M2 macrophage polarization, collectively fostering an immunosuppressive, pro-tumorigenic microenvironment.
Figure 4. Integrated Pathogenic Pathways Linking Mitochondrial Dysfunction, Epithelial Damage, Fibroblast Activation, and Tumor Progression in the Tumor Microenvironment. Mitochondrial stress increases mitoROS, triggering ATM/ATR–p53 signaling, cell-cycle arrest, senescence, and NF-κB-mediated chronic inflammation. In epithelial cells, DNA damage and p53 activation enhance EMT transcription factors (SMADs, SNAIL, TWIST, ZEB1), MMP production, and ECM degradation, driving invasion and metastasis. TGF-β-induced fibroblast activation generates α-SMA+ CAFs, which promote ECM remodeling and immunosuppressive cytokine release (IL-6, TGF-β, CXCL12). Tumor cells amplify NF-κB/STAT3 signaling, increase PD-L1 expression, recruit Tregs, and induce M2 macrophage polarization, collectively fostering an immunosuppressive, pro-tumorigenic microenvironment.
Cancers 17 03861 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chen, B.; Hamdy, H.; Zhang, X.; Cao, P.; Fu, Y.; Shen, J. From Fibrosis to Malignancy: Mechanistic Intersections Driving Lung Cancer Progression. Cancers 2025, 17, 3861. https://doi.org/10.3390/cancers17233861

AMA Style

Chen B, Hamdy H, Zhang X, Cao P, Fu Y, Shen J. From Fibrosis to Malignancy: Mechanistic Intersections Driving Lung Cancer Progression. Cancers. 2025; 17(23):3861. https://doi.org/10.3390/cancers17233861

Chicago/Turabian Style

Chen, Bing, Hayam Hamdy, Xu Zhang, Pengxiu Cao, Yi Fu, and Junling Shen. 2025. "From Fibrosis to Malignancy: Mechanistic Intersections Driving Lung Cancer Progression" Cancers 17, no. 23: 3861. https://doi.org/10.3390/cancers17233861

APA Style

Chen, B., Hamdy, H., Zhang, X., Cao, P., Fu, Y., & Shen, J. (2025). From Fibrosis to Malignancy: Mechanistic Intersections Driving Lung Cancer Progression. Cancers, 17(23), 3861. https://doi.org/10.3390/cancers17233861

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop