Next Article in Journal
Integrated Analysis of mRNA and miRNA Associated with Reproduction in Female and Male Gonads in Abalone (Haliotis discus hannai)
Next Article in Special Issue
Molecular Landscape of Metastatic Lung Adenocarcinoma in Bulgarian Patients—A Prospective Study
Previous Article in Journal
Thermogenesis and Energy Metabolism in Brown Adipose Tissue in Animals Experiencing Cold Stress
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cancer-Associated Fibroblasts as the “Architect” of the Lung Cancer Immune Microenvironment: Multidimensional Roles and Synergistic Regulation with Radiotherapy

1
School of Biopharmaceutical and Engineering, Lanzhou Jiaotong University, Lanzhou 730070, China
2
Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
3
University of Chinese Academy of Sciences, Beijing 101408, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(7), 3234; https://doi.org/10.3390/ijms26073234
Submission received: 19 February 2025 / Revised: 20 March 2025 / Accepted: 25 March 2025 / Published: 31 March 2025
(This article belongs to the Special Issue Advances in Lung Cancer: From Genetic Landscape to Treatment)

Abstract

Cancer-associated fibroblasts (CAFs), as the “architect” of the immune microenvironment in lung cancer, play a multidimensional role in tumor progression and immune regulation. In this review, we summarize the heterogeneity of the origin and the molecular phenotype of CAFs in lung cancer, and explore the complex interactions between CAFs and multiple components of the tumor microenvironment, including the regulatory relationships with innate immune cells (e.g., tumor-associated macrophages, tumor-associated neutrophils), adaptive immune cells (e.g., T cells), and extracellular matrix (ECM). CAFs significantly influence tumor progression and immunomodulation through the secretion of cytokines, remodeling of the ECM, and the regulation of immune cell function significantly affects the immune escape and treatment resistance of tumors. In addition, this review also deeply explored the synergistic regulatory relationship between CAF and radiotherapy, revealing the key role of CAF in radiotherapy-induced remodeling of the immune microenvironment, which provides a new perspective for optimizing the comprehensive treatment strategy of lung cancer. By comprehensively analyzing the multidimensional roles of CAF and its interaction with radiotherapy, this review aims to provide a theoretical basis for the precise regulation of the immune microenvironment and clinical treatment of lung cancer.

1. Introduction

According to the latest statistics released by the International Agency for Research on Cancer (IARC) in 2022, lung cancer has emerged as the leading cause of both global cancer incidence and mortality [1]. Based on histological characteristics, lung cancer is categorized into two primary types: non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). NSCLC is further classified into adenocarcinoma and squamous cell carcinoma, which represent its most common subtypes [2]. Radiotherapy is one of the very important treatment methods for lung cancer. A total of 77% of all patients with lung cancer have an evidence-based indication for radiotherapy at some point in their cancer journey [3]. In recent years, the rapid advancement of genetic testing and drug development has underscored the increasing significance of two precision treatment modalities in lung cancer therapy. The first is targeted therapy, which involves inhibitors like epidermal growth factor receptor (EGFR), activin receptor-like kinase (ALK), and reactive oxygen species (ROS)1 inhibitors designed to address driver gene mutations in lung cancer. The second is immunotherapy, where immune checkpoint inhibitors harness the body’s immune system to combat tumor cells, exemplified by PD-1/PD-L1 monoclonal antibodies [4]. Despite remarkable progress in targeted therapy and immunotherapy, treatment resistance continues to pose a significant challenge in the clinical management of lung cancer [5]. Recent studies have emphasized the pivotal role of the tumor microenvironment (TME) in driving resistance mechanisms [6]. The availability of immune checkpoint inhibitors (ICIs) targeting PD-1/PD-L1 or cytotoxic T-lymphocyte antigen 4 (CTLA-4) has revolutionized the treatment of NSCLC in the last decade. However, the chronic inflammatory environment present in lung cancer [7] may alter or deviate the differentiation of immune cells, leading to an imbalance in anti-tumor activity that favors tumor evasion [8], and subsequent resistance to ICIs [9]. Increasing evidence suggests that TME may be a promising biomarker of sensitivity to ICI based on the presence of immunomodulatory cells such as Treg, myeloid-derived suppressor cells, and tumor-associated macrophages. Therefore, several clinical studies have aimed at influencing TME towards a pro-immunogenic state, thereby increasing ICI activity [10]. For example, it was demonstrated that macrophage infiltration in NSCLC may be a determinant of PD1/PD-L1 antibody sensitivity and tolerance [11]. Additionally, vascular endothelial growth factor D (VEGFD) was found to be associated with innate immunity in patients with LUAD, which may serve as a potential predictor of LUAD patients receiving anti-PD-1 therapy [12]. Therefore, various possible regimens capable of optimizing the efficacy of ICIs are being progressively developed to meet clinical needs. It was demonstrated that an innovative small molecule inhibitor of PD-L1, S4-1, was able to induce PD-L1 internalization and optimize the immune microenvironment [13]. For targeted therapies, although Tyrosine Kinase Inhibitors (TKIs) are directly effective in killing NSCLC cells by inducing apoptosis, an increasing number of studies have demonstrated a close relationship between the regulation of the tumor immune microenvironment and the outcome of targeted therapies in lung cancer. For EGFR-TKI and ALK-TKI therapy, studies have demonstrated the dynamics of the TME during the use of different TKIs for the treatment of NSCLC, with an anti-tumor immune or immunostimulatory effect initially produced by the use of the TKIs, but an immunosuppressive effect after prolonged use which is the main reason for the emergence of resistance to the targeted therapy [14]. In the past decade, nanoparticle-based photoconductive therapy has shown great promise in the treatment of cancer, including lung cancer [15,16], with encouraging efficacy [17,18]. However, hypoxia affects the efficacy of phototransduction therapy, and a hypoxic tumor microenvironment is considered one of the hostile features of solid tumors [19]. VEGF is not only an important angiogenic factor, but also an immunomodulator of the TME. VEGF inhibits antigen presentation and stimulates the activity of regulatory T (Treg) cells and tumor-associated macrophages, which in turn promotes the formation of an immunosuppressive microenvironment in NSCLC. There is growing evidence that VEGF/VEGFR-targeted compounds and immunotherapy can serve as a new TME-targeted approach for the treatment of NSCLC [20]. Therefore, a comprehensive understanding of the TME’s components and its regulatory influence on tumor behavior will be essential for addressing the critical issue of treatment resistance in lung cancer.
In recent years, the TME has received increasing attention due to its critical role in tumor immunosuppression, distant metastasis, and targeted therapy response [21,22,23,24]. The tumor microenvironment is a highly complex system composed mainly of tumor cells, infiltrating immune cells (e.g., macrophages, dendritic cells, and lymphocytes), tumor-associated stromal cells (e.g., tumor-associated fibroblasts (CAFs), endothelial cells, and lipocytes, as well as the extracellular matrix (ECM) and multiple signaling molecules [25,26]. The tumor immune microenvironment (TIME) is mainly composed of different immune cell populations in the tumor, including immune cells and cytokines. These components are classified into anti-tumor components and pro-tumor components, and their interactions determine the trend of anti-tumor immunity, which is closely related to the immune status in the TME and the clinical prognosis of tumor patients [27,28,29]. CAFs are the most important component of TME, and activated CAFs can promote tumor growth, angiogenesis, invasion, and metastasis, as well as ECM remodeling through multiple pathways. The interaction between CAFs and TIME is considered to be another key factor in promoting tumor progression [28].
CAFs are heterogeneous and diverse in terms of morphology, function and their role in the tumor microenvironment. Their characteristics change dynamically in interaction with other cell types as the cancer progresses. This heterogeneity may affect tumor progression, treatment response, and prognosis [30]. The heterogeneity of CAFs and their multifaceted roles in lung cancer treatment are increasingly recognized as a critical area of research. CAF heterogeneity is closely linked to the TIME, particularly in mediating immune escape and immune suppression [31,32]. These cells influence the TME by secreting cytokines, remodeling the extracellular matrix, and modulating immune cell functions, thereby impacting the efficacy of both immunotherapy and radiotherapy. Emerging evidence suggests that CAFs contribute to the TIME not only by regulating immune cell infiltration but also through the expression of immune checkpoint molecules and the secretion of immunosuppressive factors, which collectively promote immune tolerance [33]. Radiotherapy, as an important therapy for cancer, not only directly targets tumor cells but also has the potential to modify CAF phenotypes or enhance their interactions with the immune system. This can amplify immune responses and synergize with immune checkpoint inhibition, ultimately counteracting immune suppression [34].
A comprehensive understanding of the intricate interplay between CAF heterogeneity and the immune microenvironment offers novel insights for refining combined strategies involving radiotherapy and immunotherapy.

2. Heterogeneity of CAF in Lung Cancer

2.1. Heterogeneity of CAF Origin

2.1.1. Fibroblast

As shown in Figure 1, the primary precursor of CAFs is normal fibroblasts, and their transdifferentiating into CAFs is predominantly driven by cancer-derived cytokines, such as transforming growth factor-β (TGF-β) [35]. This process is often accompanied by the remodeling of cellular phenotype and function, such as the upregulation of α-smooth muscle actin (α-SMA) and fibroblast activation protein (FAP) expression. Traditionally, this type of CAF has been regarded as a major driver of tumor stroma remodeling and tumor invasion [36]. Navab et al. found that TGF-β expression in CAFs is upregulated compared to normal fibroblasts [37]. LIF initiates an epigenetic switch that leads to constitutive activation of JAK1/STAT3 signaling, which results in sustained pro-invasive activity of CAF; inhibition of this pathway can chronically reverse CAF-associated pro-invasive activity and restore wild-type fibroblast phenotype [38]. Zhou et al. demonstrated that the expression of vascular cell adhesion molecule-1 (VCAM-1) in CAF-conditioned medium (CAF-CM) is higher than that in normal fibroblast-conditioned medium (NF-CM). VCAM-1 enhances the growth and invasion of lung cancer cells through the AKT and MAPK signaling pathways. Additionally, the analysis showed a positive correlation between the expression of α-SMA and VCAM-1, which is associated with poor prognosis in lung cancer patients [39]. Procopio et al. demonstrated that CSL silencing induces senescence in primary fibroblasts derived from various tissues, including the lung, supporting a model where CSL-p53 regulates CAF activation. Enhanced CAF activation simultaneously promotes the expansion of stromal cells and cancer cells [40].

2.1.2. Epithelial-Mesenchymal Transition (EMT)

Epithelial cells can undergo transformation into CAFs through EMT, a process regulated by EMT-inducing factors such as transforming TGF-β, Snail, and Slug [41]. TGF-β, in particular, is the most potent inducer of EMT in NSCLC cells and is strongly associated with tumor invasion and metastasis [42]. Yang et al. isolated exosomes secreted by CAFs and normal fibroblasts and analyzed their effects on tumor volume and EMT. They demonstrated that miR-210 expression was significantly upregulated in CAF-derived exosomes (CAF-exo), which enhanced cell migration, proliferation, invasion, and EMT in NSCLC cells [43]. Similarly, You et al. showed that Snai1-dependent CAFs induce EMT in lung cancer cells through exosomes. The levels of Snai1 in CAF-derived exosomes correlated with Snai1 expression in CAFs, highlighting its critical role in promoting EMT in lung cancer cells [44]. Additionally, Wang et al. found that CAF-CM induces EMT by modulating the expression of EMT-related markers, such as E-cadherin and vimentin, and regulating metastasis-related genes, including MMP-2 and VEGF, both in vitro and in vivo [45]. In another study, Wang et al. demonstrated that CAFs isolated from lung adenocarcinoma enhance invasiveness and induce EMT by upregulating CXCR4, β-catenin, and PPARδ through the production of stromal cell-derived factor-1 (SDF-1) [46].

2.1.3. Endothelial-Mesenchymal Transition (EndMT)

Endothelial cells can transform into CAFs through the process of EndMT. EndMT is driven by factors such as TGF-β, IL-1β, and HIF-1α and is closely associated with tumor angiogenesis and stromal remodeling [47]. Zeisberg et al. demonstrated that endothelial cells can undergo EndMT and transform into fibroblast-like cells. This transition is associated with the emergence of mesenchymal markers, including fibroblast-specific protein-1 (FSP1), and the downregulation of CD31/PECAM [48]. Ciszewski et al. found that TGF-β2 induces the mesenchymal transition of human microvascular endothelial cells (HMEC-1) into CAF-like cells, which is associated with the ILK-MMP9-MRTF axis. Higher levels of α-SMA protein and activation of the RhoA and Rac-1 pathways are critical for this differentiation. Overexpression of integrin-linked kinase (ILK) enhances myocardin-related transcription factor (MRTF) activation via the RhoA and Rac-1-MMP9 pathway, thus, promoting differentiation [49]. In addition, Choi et al. demonstrated that endothelial HSPB1 regulates EndMT in both pulmonary fibrosis and lung cancer. In clinical specimens of non-small cell lung cancer and lung tumor mouse models, HSPB1 plays an inhibitory role in EndMT [50].

2.1.4. Bone Marrow-Derived Mesenchymal Stem Cells (BM-MSC)

BM-MSCs exhibit a propensity to migrate to tumor sites and differentiate into CAFs under the influence of tumor-derived factors, particularly TGF-β, thereby facilitating cancer progression [51]. These transformed CAFs are characterized by their robust secretion of pro-inflammatory cytokines and growth factors, which contribute to tumorigenesis. Notably, studies have demonstrated that bone marrow-derived macrophages can undergo transformation into CAF-like cells, thereby promoting pancreatic cancer progression, which underscores the remarkable plasticity of stromal cells within the TME [52]. The dynamic transdifferentiating of BM-MSCs into CAFs, influenced by wound fluid and tumor cell-secreted factors, further highlights the adaptive nature of stromal cell behavior in response to the TME [53]. Quante et al. provided compelling evidence that bone marrow-derived myofibroblasts not only contribute to the MSC niche but also actively promote tumor growth, suggesting a potential mechanistic link between MSCs and cancer progression [54]. Furthermore, Spaeth et al. demonstrated that CD44 expression in MSCs plays a crucial role in the activation of the fibroblast phenotype within the tumor microenvironment. Specifically, the attenuation of CD44 in MSCs was shown to significantly limit their expression of tumor-regulated CAF markers [55]. In a related study, Peng et al. revealed that tumor-derived glucose-regulated protein 78 (GRP78) treatment induces the expression of the CAF marker α-SMA in both human bone marrow mesenchymal stem cells (HBMSCs) and mouse bone marrow mesenchymal stem cells (BMMSCs). This differentiation of BMSCs into CAFs is mediated through the activation of the TGF-β/Smad signaling pathway [56].

2.1.5. Methods for Separating CAFs from TME

Isolation of fibroblasts from tumor samples is a crucial step for understanding the tumor microenvironment, their interactions with immune cells, and their impact on cancer progression. Various studies have focused on distinct methodologies for isolating fibroblasts from tumor specimens. Sha et al. employed a trypsin-collagenase I combined digestion method to process tumor tissues and non-tumorigenic tissue samples from cancer patients. After filtration through a 70 μm membrane, CAFs and normal fibroblasts were selected using anti-fibroblast microbead MACS technology [57]. Zawieracz et al. de-scribed methods for isolating primary cancer-associated fibroblasts and fibroblasts from omental tissue using mechanical dissociation and enzymatic digestion [58]. Sharon et al. developed a strategy based on the surface marker PDGFRα to isolate purified normal fibroblasts and CAFs from fresh mouse and human tissues (including breast, pancreas, lung, etc.) via fluorescence-activated cell sorting. This approach overcomes gene expression bias caused by in vitro culture and enables unbiased transcriptional analysis of heterogeneous CAF subpopulations. It also supports subsequent functional studies free from tumor contamination [59]. Jiang et al. introduced a rapid isolation method for circulating cancer-associated fibroblasts (cCAFs) from blood samples of cancer patients. The authors developed a label-free, high-throughput microfluidic technique based on microbubble acoustics, capable of separating circulating tumor cells, immune cells (e.g., leukocytes), and rare circulating populations such as cCAFs from clinically diagnosed patients [60].

2.2. Heterogeneity of Molecular Phenotype

2.2.1. Classical Molecular Markers of CAFs

Based on the available studies, the common markers of CAFs in lung cancer are summarized in Table 1. Many of the markers in the table are also suitable for the identification of CAFs in other tumor types [61,62]. Among them, the more important ones are FAP, α-SMA, and PDGFRβ, which will be specifically described below.
1.
FAP
FAP is a cell surface glycoprotein predominantly expressed on activated fibroblasts within tumor tissues, particularly in CAFs. As a type II transmembrane serine protease, FAP exhibits both collagenase and gelatinase activities, playing a pivotal role in ECM remodeling and facilitating cancer cell migration, invasion, and metastasis [72]. Elevated FAP expression has been observed in various cancers, including lung cancer, and is correlated with poor prognosis across multiple cancer types. Wang et al. investigated the metastatic potential of lung cancer cells influenced by CAFs, demonstrating that stromal fibroblasts isolated from lung cancer tissues displayed CAF characteristics, marked by high expression levels of α-SMA and FAP [45]. Kilvaer et al. explored the potential immune-adjuvant role of FAP-1-positive fibroblasts in NSCLC, suggesting a link between CAFs and immune markers within the tumor stroma [73]. Chen et al. examined the clinical significance of FAP-α in lung squamous cell carcinoma, revealing that higher CAF density and lymph node metastasis were negatively correlated with patient survival [74]. Fang et al. developed FAP-targeted chimeric antigen receptor NK-92 cells for NSCLC, demonstrating increased FAP expression in A549+CAF and H226+CAF cells in nude mouse models [75]. Mathieson et al. identified that specific CAF subpopulations expressing FAP and podoplanin in NSCLC serve as predictors of poor clinical outcomes, highlighting the importance of characterizing CAF subtypes in NSCLC [64]. Peltier et al. discovered that FAP is detectable on the surface of CAFs in numerous cancer types associated with poor prognosis. Consequently, they developed and validated 68Ga-labeled radiopharmaceutical FAP inhibitors (FAPIs) for whole-body imaging of FAP expression using positron emission tomography (PET/CT) [63].
2.
α-SMA
Wang et al. investigated the influence of CAFs on the metastatic potential of lung cancer cells, revealing that fibroblasts isolated from lung cancer tissue exhibited CAF characteristics, characterized by elevated levels of α-SMA [45]. Li et al. demonstrated that CAF markers, including α-SMA, vimentin, and CAF-specific factors such as CXC motif chemokine ligand 2 (CXCL12), FGF10, interleukin-6 (IL-6), and collagen type I alpha 1 chain (COL1A1), were highly expressed in senescent fibroblasts, which contributed to the promotion of lung cancer cell migration and invasion. These senescent fibroblasts acquired a CAF phenotype through a Stat3-dependent mechanism [76]. Additionally, Zhou et al. highlighted the role of VCAM-1 secreted by CAFs in enhancing lung cancer cell growth and invasion via the AKT and MAPK signaling pathways. Their studies revealed a positive correlation between the expression of CAF marker proteins, α-SMA and VCAM-1, which was associated with poor prognosis in lung cancer patients. These findings underscore the critical role of CAFs in driving lung cancer invasiveness [39]. In contrast, Lu et al. focused on SCLC and explored the dynamic phenotypic reprogramming induced by lung fibroblasts in SCLC. They identified a positive correlation between the expression levels of α-SMA, CAF markers, and REST (a protein typically expressed in non-neuroendocrine SCLC. This study emphasizes the importance of understanding the interplay between CAFs and non-neuroendocrine tumor cells within the tumor microenvironment [77].
3.
PDGFR-α/β
Platelet-derived growth factor receptor (PDGFR) is a receptor tyrosine kinase that plays a pivotal role in modulating the behavior of fibroblasts and other cellular components within the tumor microenvironment. PDGFR-α and PDGFR-β have been recognized as key markers for fibroblast identification and are frequently upregulated in various malignancies, including lung cancer [78]. Both the PDGFR-α homodimer and the PDGFR-α/β heterodimer are implicated in the functional activities of CAFs. Emerging evidence suggests that matrix reprogramming through dual PDGFRα/β inhibition holds potential for enhancing therapeutic outcomes in gastric and lung cancers [79]. Transgelin, a protein implicated in promoting lung cancer progression, has been linked to elevated levels of α-SMA and PDGFR-β, further underscoring the significance of these markers in the functional regulation of CAFs [80]. Additionally, Sugai et al. assessed the expression of CAF-related proteins, including PDGFR-α and PDGFR-β, in lung squamous cell carcinoma (LSCC) tissues using immunohistochemistry to evaluate their impact on clinical outcomes [64].

2.2.2. Subtypes of CAF Based on Molecular Phenotypes

1.
MyCAF
The heterogeneity of CAF subtypes is also briefly represented in Figure 1. Myofibroblast-like cancer-associated fibroblasts (myCAFs) represent a distinct subset of CAFs within the TME, prominently identified in malignancies such as pancreatic ductal adenocarcinoma (PDAC) and head and neck squamous cell carcinoma (HNSCC). Research has demonstrated that integrin α11β1 is expressed at moderate levels in myCAFs, while it is notably absent in other stromal cell types, including FSP1-positive and neuron-glial antigen 2 (NG2)-positive cells [81]. In lung adenocarcinoma (LUAD), myCAFs have been identified as the major subtype of CAFs [82]. myCAF is rich in ECM transcripts and can enhance the invasion and metastatic ability of lung cancer cells by remodeling the ECM and secreting chemokines. The secreted matrix metalloproteinases (MMPs) play a key role in tumor invasion and metastasis by promoting cell migration and ECM degradation [83].
2.
iCAF
Inflammatory CAFs (iCAFs) are also one of the more predominant subtypes of CAFs [84,85]. Xu et al. identified three distinct CAF subpopulations in NSCLC using single-cell sequencing. Their findings suggest that iCAFs may interact with NSCLC cells by activating molecules associated with invasion and metastasis, such as MET-hepatocyte growth factor (HGF) signaling pathway, thereby promoting brain metastasis in NSCLC [86]. In a spatially resolved single-cell imaging mass cytometry (IMC) analysis of CAFs from 1070 NSCLC patients, heterogeneity of CAFs was found to be an independent prognostic factor affecting patient survival. In particular, iCAF was associated with an inflammatory tumor microenvironment and higher patient survival [87]. Cords et al. discovered that the CAF phenotype is an independent prognostic factor for NSCLC, with iCAFs and interferon response CAFs being associated with an inflamed TME and improved patient survival [87]. Additionally, Zhao et al. revealed that the circRNA-mediated fibroblast niche plays a critical role in tumor metastasis. Specifically, the circNOX4/miR-329-5p/FAP axis activates the iCAFs niche by preferentially inducing IL-6, ultimately driving NSCLC progression [88].
3.
Other subtypes
Kim et al. identified a new subgroup of CAFs in lung adenocarcinoma, antigen-presenting CAFs (apCAF) via RAN-seq, and demonstrated that some of their markers, UBE2T and KPNA2, promote CAF aggressiveness [64]. Lu et al. demonstrated that crosstalk between CAFs and non-neuroendocrine (non-NE) SCLC cells promoted the presence of apCAFs, which may contribute to CD8+ T cell capture and Treg differentiation [89]. Chen et al. identified matCAF, suggesting that matCAF plays a crucial role in cancer development and progression [84]. In studies for NSCLC, it has been demonstrated that matrix CAF (mCAF) was associated with low immune infiltration and low patient survival [87].

2.2.3. Subtypes of CAFs in Different Lung Cancer Types

In NSCLC, CAFs exhibit significant heterogeneity, encompassing distinct subtypes such as myCAF, iCAF, apCAF, and matCAF [90]. Notably, CAFs with myofibroblast characteristics have been identified as a potential marker of malignancy in advanced NSCLC [91]. Hu et al. established an in vivo CAF biobank and identified three major functional CAF subtypes: Subtype I, which overcomes EGFR inhibitor resistance via the MET pathway and highly expresses HGF and FGF7; Subtype II, which mediates moderate resistance primarily through the FGFR pathway and also expresses FGF7; and Subtype III, which exhibits a weaker protective effect. These subtypes are regulated by the TGF-β pathway, and their functional differences significantly influence therapeutic responses to EGFR and ALK TKIs [92]. Hanley et al. utilized single-cell transcriptomic analysis to reveal an enriched myoCAF gene signature in NSCLC compared to normal lung fibroblasts, while iCAF-associated genes were more prominent in normal lung tissues [93].
In LUAD, one study identified five CAF subgroups in LUAD, three of which were associated with prognosis. Based on the prognostic significance of CAF subgroups, the study constructed a risk characterization model containing eight genes, including one protective gene (BTK) and three risk genes (CCNB1, CDC25C and EXO1). The CAF-based risk profile model can effectively predict the prognosis of LUAD patients and provide a new strategy for interpreting the response of LUAD to immunotherapy [94]. Another study found that Thy-1+ CAF (tCAF) localized in metastatic foci and formed tCAF-dominated invasive structures under ZEB1-driven REPULSIVE FORCES, supporting the notion that CAF facilitates invasion through mechanical forces [95].
In LUSC, single-cell RNA sequencing classified diverse CAF subtypes. Univariate Cox analysis identified prognostic CAF-related genes (CAFRGs), revealing correlations between CAFRGs and malignant features such as tumor angiogenesis, EMT, and cell cycle dysregulation [96]. Fang et al. understood the heterogeneity of the tumor microenvironment in lung adenocarcinoma and squamous carcinoma using single-cell transcriptomic analysis and found that infiltrating CAF were elevated in LUSC and up-regulated immunosuppressive extracellular matrix remodeling [97]. Notably, Ki67 exhibits divergent prognostic roles in LUAD and LUSC, reflecting tumor proliferation heterogeneity. High Ki67 expression predicts poor prognosis in LUAD, whereas low Ki67 levels correlate with elevated EMT potential and worse outcomes in LUSC [98].
SCLC is a highly malignant lung cancer type in which the role of CAF in TME remains incompletely defined. However, it has been shown that the non-neuroendocrine (non-NE) subtype of SCLC may be associated with the presence of apCAF. CAF regulates the immune microenvironment of the tumor through CD8+ T-cell capture and Treg differentiation, which in turn affects the therapeutic response of SCLC [89].

3. CAF and Tumor Immune Microenvironment in Lung Cancer

Much evidence has demonstrated that CAFs play an important role in the regulation of TIME. CAFs are able to regulate the antitumor activity of tumor-infiltrating immune cells [99], and are able to promote the regulation of ECM remodeling and immune checkpoint molecules, which indirectly affects the activity and recruitment of immune cells [100]. In addition, CAF are involved in cancer development and progression through the secretion of chemokines, cytokines, and other effector molecules, including TGF-β, CXCL2, collagen, MMPs, and laminin [101,102]. One study found that in NSCLC, the FBLIM1-positive CAF subtype is an aggressive subtype associated with increased TGF-β expression, elevated levels of mesenchymal markers and an immunosuppressive tumor microenvironment [103]. In a study of CAFs in NSCLC patients, CAFs with high expression of both HGF and FGF7, and CAFs with high expression of FGF7 were found to be differentially protective against tumors, and were associated with the immune microenvironment of tumors [92]. In short, CAF is closely related to the immune microenvironment of lung cancer, and can influence the recruitment and function of multiple immune cells, which leads to the regulation of the immune status. The relationships between CAF and innate immune cells, adaptive immune cells, and ECM are discussed separately below and summarized in Figure 2.

3.1. Connection Between CAF and Innate Immune Cells

3.1.1. CAF and TAMs

Tumor-infiltrating macrophages are known as tumor-associated macrophages (TAMs) and can be divided into two subtypes, M1 and M2 [104]. M1-type macrophages exhibit antitumor effects in TIME mainly by mediating the production of ROS, cytotoxicity, and tumor necrosis factor (TNF) [105], whereas M2 macrophages exhibit pro-tumor activity by activating cancer cell invasion and metastasis, tumor angiogenesis, immunosuppression, and ECM remodeling [106,107]. TAMs are the most prominent immune cells in proximity to CAFs, and there is a close interaction between the two cells [108]. It has been shown that CAFs can promote the recruitment of monocytes (precursors of macrophages) and differentiate them into M2-type TAMs through a variety of regulatory molecules, resulting in immunosuppression [109]. The same trend has been found in prostate cancer as well as breast cancer [110,111]. CAFs in lung adenocarcinoma promoting the polarization of TAMs to the M2 phenotype are associated with the FGF/FGFR signaling pathway [112]. For CAF subtypes, it has been found that PDPN+ CAFs exhibit higher TGF-β expression and are associated with the infiltration of CD204+ TAMs (M2 phenotype TAMs) in lung cancer, which in turn contributes to an immunosuppressive TME [113]. POSTN+ CAFs can bind to SPP1+ TAMs, which in turn contribute to the immunosuppression of NSCLC [114]. Furthermore, macrophage-myofibroblast transformation (MMT) is known to exist in lung cancer to support cancer progression, and this transformation may be Smad3-dependent [115]. The hematopoietic transcription factor Runx1, a key regulator downstream of Smad3 signaling, is positively associated with MMT-derived CAF abundance and mortality in NSCLC patients [116].

3.1.2. CAF and TANs

Tumor-associated neutrophils (TANs) are also one of the important constituents of TIME. Tumor-produced chemokines attract neutrophils in the blood circulation, which pass through the vessel wall into the tumor tissue and then form TANs. Once TANs are activated in the tumor microenvironment, they appear to increase the complexity of the inflammatory environment through a mechanism of attraction to other leukocytes [117]. TANs play an important role in many aspects of tumorigenesis and development, such as malignant transformation, tumor progression, extracellular matrix modification, angiogenesis, cell migration, and immunosuppression [118,119,120]. Similar to TAM, it has been shown that neutrophils can be categorized into N1 and N2 types, representing tumor suppressor and tumor promoter phenotypes, respectively [121]. Studies have demonstrated that CAFs promote the polarization of N2 TANs in lung cancer [122]. C-X-C chemokine receptor 2 (CXCR2), a cytokine receptor expressed by CAFs, has been shown to be a major factor involved in tumor neutrophil recruitment and migration [121,123]. It has been proved in lung cancer as well as in mesothelioma that TGF-β plays an important role in the polarization of the TANs phenotype [124]). Additionally, the transdifferentiating of fibroblasts to CAFs is largely driven by TGF-β [35,125]. Furthermore, studies on lung cancer have found that granulocyte colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) induce immune escape from lung cancer by regulating neutrophils [126].

3.1.3. CAF and NK Cells

Natural killer (NK) cells are members of the innate immune system that respond to tumor cells with a variety of effector functions, primarily cell killing and the production of pro-inflammatory cytokines [127,128,129] The cytotoxic activity of NK cells makes them most functionally similar to CD8+ T cells [129]. The activity of NK cells is dependent on the expression and stimulation of activating or inhibitory receptors on the cell surface [130,131]. A growing number of studies have shown that CAFs are able to exert effects on NK cells by direct or indirect means [132,133]. In lung cancer, CAFs are able to exert significant immunosuppressive effects on NK cells [134]. TGF-β is recognized as a key cytokine connecting CAFs and NK cells in tumors [135]. TGF-β secreted by CAFs significantly inhibits the activation and cytotoxic activity of NK cells [136]. The mechanism may be due to the fact that TGF-β reduces the production of interferon-γ (IFN-γ) and down-regulates activated receptors on the cell surface [137].

3.1.4. CAF and DCs

Tumor-infiltrating dendritic cells (DCs) are initiators and coordinators of the adaptive immune response and are essential for the anti-tumor immune response [138]. DCs are essential for T-cell-mediated cancer immunity; they transport tumor antigens to the draining lymph nodes and cross-present antigens to activate cytotoxic T lymphocytes [139]. In recent years, several studies have shown that CAFs can lead to the immune escape of tumor cells by blocking processes such as DC maturation and antigen presentation. In lung cancer, CAFs can induce a tolerogenic phenotype in DCs [140]. Galectin-1 derived from lung cancer is one of the culprits of DC cell incapacitation [141], and it has been demonstrated that this phenomenon is mediated by the galectin-1-CAF axis and mainly associated with the tryptophan 2,3-dioxygenase (TDO2)/kynurenine axis in CAFs [142]. Additionally, researchers have found that immunomodulation through inhibition of CAF function improves regional and systemic antitumor immune responses, thereby increasing the efficacy of DC-based vaccine immunotherapy [143].

3.1.5. CAF and MDSCs

Myeloid-derived suppressor cells (MDSCs) originate from the bone marrow and have potent immunosuppressive activity in TIME [144]. MDSCs are a heterogeneous population of myeloid cells, which are a mixture of myeloid cells with the phenotype of granulocytes and monocytes, but lack the expression of cell surface markers specific for monocytes, macrophages, or DC, and are effective in suppressing both innate and adaptive immune responses, especially in T cell proliferation and cytokine production [145]. In LSCC, CAF is able to induce the aggregation of mononuclear MDSCs (M-MDSCs) and the generation of large amounts of ROS, which ultimately leads to the inhibition of CD8+ T cell growth, as well as the restricted production of IFN-γ, resulting in immunosuppression [146]. Fibrinogen-like protein 2 (Fgl2), a pleiotropic cytokine affecting multiple cellular functions, was found to be able to increase the aggregation of MDSCs through CXCL12 in lung cancer, and also induced the activation and pro-tumorigenic phenotype of CAFs, which are the main source of CXCL12 in TME [147]. Moreover, CAFs produce the granulocyte chemokine CXCL1, which may also be involved in the recruitment of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) [148].

3.2. Connection Between CAF and Adaptive Immune Cells

T lymphocytes play a key role in regulating the adaptive immune response, and they consist of different subpopulations such as Treg cells, helper T (Th) cells, and cytotoxic T lymphocytes (CTL) [149]. The success of immune checkpoint therapy suggests that tumor-reactive T cells can destroy cancer cells but are constrained by immunosuppression in the TME. CAF are the major stromal cells in the TME and co-localize with T cells in lung cancer [150].

3.2.1. CAF and Treg Cells

Forkhead box P3 (Foxp3) highly expressed Treg cells have an important function in limiting anti-tumor immunity [151]. In lung cancer, it was demonstrated that Treg cells were adjacent to CAFs and that infiltration of Foxp3+ Tregs and CAFs in the tumor stroma correlated with poor prognosis [152]. Moreover, CAFs expressing immunomodulatory cytokines may induce Tregs in the stroma, resulting in a tumor-promoting microenvironment in lung adenocarcinoma that leads to a poor prognosis [152]. CAFs in NSCLC amplify a Treg phenotype in TME by driving the production of CXCL13 from activated T cells via TGF-β [153]. In a recent study targeting SCLC, apCAFs were shown to promote glycolysis in tumor cells and induce the formation of Tregs from naive CD4+ T cells in an antigen-specific manner [89].

3.2.2. CAF and Th Cells

The cell subsets mainly include Th1, Th2, and Th17 cells, which mostly differentiate from naive CD4+ T cells [154]. Th1 and Th2 cells are involved in cellular and humoral immunity, respectively, through the secretion of a variety of specific cytokines [155]. The presence of CAFs induced infiltration of B cells and CD69+CD4+ T cells, which was associated with increased expression of CCL13 and increased expression of CXCL16 [156]. Meflin-positive CAF enhanced the tumor response to immune checkpoint blockade (ICB) therapy, positively correlating with CD4+ T-cell infiltration and vascularization [157].

3.2.3. CAF and CTLs

CD8+ T cells, also known as CTLs, mediate cytotoxicity mainly by inducing apoptosis in tumor cells, which is the most critical component of anti-tumor immunity [155]. Numerous studies have reported interactions between CAFs and CD8+ T cells in lung cancer [158]. THBS2+ CAFs are capable of interacting with CD8+ T cells and are associated with immunosuppression in lung adenocarcinoma [159]. It was found that apCAFs may capture CD8+ T cells and trap them in the stromal region [89]. POSTN+ CAFs are able to bind to SPP1+ macrophages and cause inhibition of T-cell infiltration, and have been associated with a poor prognosis in NSCLC [114]. The CD8/Foxp3+ T-cell ratio tends to be lower in the region of PDPN+ CAFs, resulting in a suppressive immune microenvironment [160]. Two types of CAFs, MYH11+αSMA+ CAF and FAP+αSMA+ CAF, were also found to be associated with T-cell marginalization [161]. Hyewon et al. found that CAFs inhibit the function of CD4+ and CD8+ T-cells in NSCLC through their effects on COX2 and PD-L1, thereby promoting immune escape [162]. Another study demonstrated that CAF significantly increased CXCL13 production by CD4+ and CD8+ T cells via TGF-β, thereby altering tumor TIME [153]. Mao et al. demonstrated that the CD8+ T cell-derived exosome miR-2682 inhibited lung cancer tumor formation, whereas the CAF-derived exosome FOXD3-AS1 promoted lung cancer tumor formation [163]. In addition to this, the CD8+ T cell/CAF ratio (CFR) allows for prognostic stratification as well as response prediction to immunotherapy in multiple cancer types [164]. In addition to the regulatory effect of CAF on T cells, studies have demonstrated that there is also a negative feedback mechanism of T cells on CAF, promoting the expression of co-suppressor ligands, CD73 and IL-27 in NSCLC [150]. A recent study indicated that T-cell effector cytokines IFN-γ and TNF-α induced cytokine and chemokine secretion from NSCLC patient-derived CAFs in vitro, which may affect immune cell recruitment and activation and, ultimately, tumor progression and treatment response [165].

3.3. Interactions Between CAF and Other Factors in TIME

High expression of immune checkpoint molecules on the surface of T cells and tumor cells has been identified as a major cause of immunosuppression of T lymphocytes [166,167]. PD-L1 and PD-1 are well-known checkpoint molecules. The binding of PD-L1 to PD-1, a receptor on activated T cells, counteracts T cell activation signals and, thus, prevents anti-tumor immune response [168]. It was found that in lung cancer, CAF was able to inhibit T cells by activating immune checkpoints via PD-L2 and FASL [169]. Similarly, researchers found that CAF expressing PD-L1 and PD-L2 in NSCLC were affected by IFN-γ and influenced T cell function [170]. FAP was found to predict the efficacy of PD-1 blockade therapy in advanced non-small cell lung cancer [171]. Furthermore, WDR62 was significantly up-regulated in most tumors, and WDR62 expression was associated with the infiltration of a variety of immune cells, particularly CAF and Treg cells, and was significantly correlated with poor prognosis. Studies have shown that WDR62 predicts the efficacy of immunotherapy, particularly PD1/PD-L1 inhibitors [172]. In addition to upregulating molecules on their own surface, CAFs produce various cytokines and exosomes, which indirectly exert an inhibitory effect on T cell function and antitumor responses. By secreting factors such as CXCL2, CAFs can increase the expression of PD-L1 in lung adenocarcinoma cells, thereby affecting tumor immunity [173].
ECM is a complex network of different macromolecules such as collagen, fibronectin, glycoproteins, and proteoglycans, which are responsible for maintaining the structure, integrity, and homeostasis of normal tissues [174,175]. By secreting a variety of matrix proteins and producing a number of matrix metalloproteinases (MMPs), CAFs promote the degradation of normal ECM structures and increase matrix stiffness [175,176]. Dysregulation of the ECM has been found to be associated with activation of TGF-β signaling in CAFs and with immunosuppression of tumors [177]. This remodeled ECM acts as a physical barrier for immune cells, thereby inhibiting their recruitment to the cancer site and reducing the immune response [178,179]. The density of collagen in lung cancer has been found to influence anti-tumor immunity, and in areas of dense stroma, the ability of T cells to reach the cancer site is limited [180]. In addition, ECM remodeled by CAF modulates the activity and function of other immune cell populations. CAF-induced collagen-rich stroma is associated with TAM recruitment and function. In lung cancer, this aberrant collagen-rich ECM induces M2-type polarization of macrophages, and collagen monomer-induced innate immune responses may perpetuate lung fibrosis [181]. COL11A1+ CAF promotes the progression of lung adenocarcinoma and bladder cancer by regulating ECM remodeling and anti-tumor immune responses [182]. Conversely, it has been found that in lung adenocarcinoma, ADH1B+ CAF is associated with better survival. Reduced COL1A1 and COL3A1 expression and increased DCN, LAMA2, and ELN expression have been characterized in ADH1B+ CAF, and correlate with the function of the ECM [82].
In conclusion, the heterogeneity of CAF is capable of exerting diverse effects on immune checkpoint molecules and the ECM, and generating different responses to anti-tumor immune responses.

4. Relationship Between CAF and Radiotherapy

The treatment of lung cancer is complex, and radiotherapy (RT) can be used in all stages of the disease for curative or palliative care. With advances in technology, radiotherapy allows for better targeting of the tumor and reduces incidental irradiation of surrounding normal tissue. This has expanded the indications for radiotherapy in lung cancer and improved outcomes in terms of increased survival and reduced toxicity [3]. However, RT has been shown to be a double-edged sword that not only enhances systemic anti-tumor immune responses, but also leads to immunosuppression [183]. For example, radiotherapy promotes anti-tumor immunity, activates cytotoxic T cells, and enhances the killing effect on tumor cells [184]. In addition, radiotherapy induces the upregulation of immunosuppressive molecules, promotes the inactivation of immune effector cells, weakens the immunogenicity of tumor cells, and ultimately leads to the development of immune escape and tolerance [185]. Therefore, the alteration of TME by RT affects the anti-tumor immune response in multiple ways.
Many studies have shown that radiation can alter TME by affecting the phenotype of CAFs. For example, in NSCLC, the upregulation of some immune receptors such as CD73 and CD276 on the surface of irradiated CAFs contributes to the formation of an immunosuppressed TME after radiotherapy [183]. Radiotherapy-induced generation of senescence-like CAFs promotes the proliferation and radioresistance of NSCLC cells through the JAK/STAT pathway. Specific induction of senescence-like CAF apoptosis using the FOXO4-p53 interfering peptide FOXO4-DRI reduces both NSCLC radioresistance and incidence of radiation-induced pulmonary fibrosis (RIPF) [186]. Irradiation-induced elevation of immune receptors on the cell surface of CAFs may contribute to the establishment of immunosuppressive TME after radiotherapy [183]. It has been found that although much evidence suggests that high-dose (>5 Gy) radiotherapy regimens have a stronger pro-immunogenic effect than standard low-dose (2 Gy) regimens, CAFs have a potent immunosuppressive effect on activated T-cells, and this effect is maintained after high-dose radiotherapy (HD-RT). Importantly, CAFs do not initiate an immunogenic cell death response after exposure to HD-RT [187]. CAFs in irradiated NSCLC were able to maintain immunosuppressive effects on NK cells [134]. One study selected 68 patients with brain metastases from different primary cancer types, including 10 lung cancer patients, and evaluated the expression and clinical relevance of CAF-related biomarkers in brain metastases (BM), and showed that PDGFR-β and α-SMA were highly expressed in patients who had received chemotherapy or radiotherapy for their primary cancers, and that they were associated with a poor prognosis and recurrence in BM patients [188]. In another study, CAFs were isolated from lung tumor specimens of 16 NSCLC patients, and the results showed that radiation caused a sustained enhancement of the surface expression of integrins α2, β1 and α5, and that radiation had a favorable inhibitory effect on the proliferative, migratory and invasive capacities of CAFs in NSCLC [189]. However, conversely, it has also been shown that integrin β1 allows SCLC cells to survive in the presence of sustained DNA damage [190]. The fibroblast integrin α11β1 has been shown to induce myofibroblast activation [191]. Integrin αVβ6 has been reported to be upregulated prior to the onset of fibrosis in irradiated lungs, suggesting that integrins may also be involved in the development of radiation fibrosis [192]. In addition, radiotherapy enhances TGF-β signaling [193]. Elevated TGF-β levels may be a result of CAF activation and may also influence tumor progression by further modulating CAF activity [194].
In addition, molecules secreted by CAFs are able to influence the outcome of radiotherapy. It has been shown that the exosome miR-196a-5p from CAFs enhances the radiation resistance of lung cancer cells by down-regulating NFKBIA [195]. CAFs produce IGF1/2, CXCL12, and β-hydroxybutyrate that induce autophagy and promote the recovery of cancer cells from radiation damage after radiotherapy [196]. CAF-derived soluble factors mediate measurable changes in undifferentiated macrophages and down-regulate pro-inflammatory features of M1-polarized macrophages [197]. It has also been shown that CAFs are radioresistant, with significant changes in oxidative metabolic indices. CAFs that survive in radiation therapy may alter the fate of the associated cancer cells [198]. IL-22 produced by CAFs is able to increase the proliferation, invasion and migration of lung cancer cells [199]. It has been shown that in lung adenocarcinoma CAF is able to induce a tolerogenic phenotype in DC cells and enhance immunosuppression. However, ionizing radiation was able to reverse the CAF-mediated immunosuppressive effects [140].
Targeted radionuclide therapy (TRT) is a type of cancer therapy, and radionuclide therapy targeting FAP is a hot topic of current research. It has been shown that in preclinical lung cancer mouse models, [225Ac]Ac-DOTA-4AH29 and [131I]I-GMIB-4AH29 exhibited high and sustained tumor targeting, which prolonged the survival of mice with aggressive tumors; furthermore, the binding of PD-L1 ICB to [225Ac]Ac-DOTA-4AH29 TRT enhanced its efficacy [200]. In a clinical trial that included nine patients with advanced lung cancer, 177Lu-FAP-2286 treatment resulted in improvements in overall health, symptom response, and quality of life [201]. In addition to this, targeting FAP is more often used as a tracer. A clinical trial of [68Ga]Ga-FAPI-RGD, a dual-targeted heterodimer PET tracer that recognizes both FAP and integrin αvβ3, included 51 patients with lung malignancies. The results showed that [68Ga]Ga-FAPI-RGD PET/CT had a higher primary tumor detection rate, higher tracer uptake, and better metastasis detection rate compared with [18F]FDG PET/CT [202]. Another study developed a novel radiolabeled FAP inhibitor probe [18F]AlF-FAPI-74 for non-invasive monitoring of the effects of anti-FAP CAR T-cell therapy in solid tumors, demonstrating its potential for detecting changes in FAP expression in the tumor microenvironment and serving as a biomarker for predicting and monitoring response to FAP-targeted therapy [203]. In addition, a novel FAP-targeted diagnostic and therapeutic integrated probe, 68Ga/177Lu-labeled DOTA-FAPT, exhibited high stability, hydrophilicity, and strong targeting affinity, and showed excellent tumor uptake, diagnostic efficacy, and therapeutic potential in mouse models and lung cancer patients, with a promising future for application [204]. In an international multicenter retrospective analysis, 71 patients with various types of cancers, including 9 lung cancer patients, were treated with both 18F-FDG and 68Ga-labeled FAP inhibitors. Compared with 18F-FDG, 68Ga-FAPI had a higher diagnostic performance for cancer staging and restaging in various indications [205].
In conclusion, the importance of CAFs in the tumor microenvironment and their complexity profoundly affects the efficacy of radiotherapy. Radiotherapy can mainly produce immunosuppressive effects by altering the phenotype of CAF. However, the development of radionuclide therapy targeting CAF-specific markers, such as FAP, for diagnostic and therapeutic purposes offers new hope for overcoming tumor immunosuppression and improving the efficacy of radiotherapy.

5. Discussion

CAFs have emerged as key regulators in the immune microenvironment of lung cancer, where they orchestrate a complex network of interactions that have a major impact on tumor progression and treatment outcome. As the “architects” of the microenvironment, CAFs exhibit multifaceted roles, including ECM remodeling and immune regulation. Their ability to recruit and polarize immune cells highlights their central role in the formation of an immunosuppressive environment conducive to tumor immune evasion. This dynamic interaction between CAFs and immune cells highlights the potential of targeting CAFs to remodel the tumor immune microenvironment and improve efficacy.
The interaction between CAFs and radiotherapy adds another layer of complexity to their role in lung cancer. While the main goal of radiotherapy is to induce tumor cell death, it also has profound effects on the tumor microenvironment. As we discussed in the previous sections, CAFs can directly regulate the response to radiotherapy by promoting the radioresistance and recovery ability of cancer cells; they can also affect the tumor TME by secreting some cytokines, resulting in a microenvironment of immunosuppression and, thus, indirectly affecting the efficacy of radiotherapy. Secondly, radiotherapy can activate CAFs, leading to further remodeling and immunosuppression of the ECM, which may ultimately limit the efficacy of radiotherapy. However, we also addressed that new evidence suggests that combining CAFs as targets with radiotherapy can enhance anti-tumor immunity [206]. Therefore, strategies aimed at inhibiting CAF-derived signaling pathways or removing CAFs have shown promise in preclinical models, paving the way for novel combination therapies.
Despite these advances, a number of challenges remain. An important aspect of CAF biology that deserves further attention is its heterogeneity. CAFs are not a uniform population of cells, but rather consist of distinct subpopulations with different origins, functions, and molecular characteristics. However, the classification of CAF subtypes currently lacks a standardized framework, as its definition is largely based on individual study analyses and methods. This variation in classification criteria stems from differences in experimental methods, such as single-cell sequencing, immunohistochemistry, or functional assays, as well as the specific markers or pathways emphasized in each study. For example, some researchers classify CAFs based on their functional roles (e.g., myCAFs, iCAFs, and apCAFs), while others focus on molecular features or spatial distribution in the TME. This lack of consensus complicates cross-study comparisons and may hinder the development of universal therapeutic strategies for CAFs. Going forward, a unified classification system that integrates the molecular, functional, and spatial characteristics of CAFs is critical to advancing our understanding of the role of CAFs in cancer progression and improving the precision of CAF-targeted therapies.
CAFs play dual roles in promoting and inhibiting tumor progression, and this functional diversity is also influenced by spatial and temporal factors in the tumor microenvironment as well as by interactions with other stromal and immune cells. The heterogeneity of CAFs poses a great challenge for targeted therapies because indiscriminate depletion or inhibition of CAFs may inadvertently disrupt their potential antitumor functions. Secondly, although we have searched as much as possible for evidence to describe the role of CAFs with various immune cells in lung cancer, we have also found directions that have not been investigated so far, such as the relationship between mast cells (MCs) and CAFs. MCs also have two sides in tumors. As cancer promoters, they produce different pro-angiogenic molecules such as VEGF-A, VEGF-B, FGF, heparin, stem cell factor (SCF), etc. [207,208,209,210]; on the other hand, MCs have anti-tumor effects. By producing mediators such as chondroitin sulfate, TNF, IL-1, and IL-6, it increases inflammatory response and induces apoptosis in tumor cells [211,212]. In pancreatic cancer, secretions from activated MCs induce tumor proliferation and TME changes, especially transforming fibroblasts into an iCAF phenotype [213]. However, there are currently no relevant studies on lung cancer. Future studies should focus on elucidating the specific molecular mechanisms by which CAF interact with immune cells and their modulation by radiotherapy. In addition, the development of biomarkers to identify subpopulations of CAF with pro- or anti-carcinogenic properties is essential for patient stratification and personalized treatment strategies.
The applications and study of TME in the clinic are very important and wide-ranging. For example, specific components in TME can be used as therapeutic targets, and biomarkers in TME can also be used for tumor diagnosis, staging, and prognostic assessment. In addition, by analyzing the heterogeneity of TME, individualized treatment plans can be developed for patients. However, there are many limitations of TME research in the clinic. For example, TME is highly heterogeneous within tumors and among different patients, which makes the development of universal treatment regimens complex and difficult. The methods for assessing TME have not yet been fully standardized, resulting in limited reproducibility and comparability of results, and dynamic changes in TME over time during treatment add to the complexity of developing treatment strategies. Existing technologies still have limitations in resolution and sensitivity, making it difficult to fully capture the complexity of TME and its specific changes. Therefore, although TME shows great potential in basic research, its clinical translation still faces many challenges. CAFs, as key components in the TME, have not been fully elucidated in terms of their functional diversity and dynamic changes in the TME, which limits our comprehensive understanding of their roles in tumor biology as well as the development of relevant therapeutic strategies. The functional status and phenotype of CAFs change dynamically in response to tumor progression, therapeutic interventions, and changes in microenvironmental signals. As we have discussed above, radiotherapy affects the altered phenotype and function of CAFs, which in turn affects the function of other cells in the TME and the formation of an immunosuppressive environment. In future studies, in addition to systematically analyzing the subpopulation composition of CAFs and their functional characteristics with the help of high-throughput technologies such as single-cell sequencing, spatial transcriptomics, in order to clarify the specific mechanism of their role in the TME, we should also pay attention to the dynamic evolution of CAFs in the course of treatment, and monitor the changes of CAFs and their impact on the response to treatment. This will provide an important basis for developing more precise treatment strategies.
In conclusion, CAFs play a central role in shaping the immune microenvironment and modulating the response to radiotherapy in lung cancer. Their multidimensional roles and dynamic interactions with immune cells and therapeutic interventions highlight their potential as therapeutic targets. By unraveling the complexity of CAF biology and developing innovative strategies to disrupt their pro-tumorigenic functions, we can pave the way for more effective combination therapies that harness the immune system and enhance the efficacy of lung cancer radiotherapy.

Author Contributions

Conceptualization, C.S. and Q.L.; writing—original draft preparation, Z.S. and C.H.; writing—review and editing, C.S.; supervision, Q.L.; funding acquisition, Z.S., Q.L. and C.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was jointly supported by the Youth Science Foundation Project of Lanzhou Jiaotong University (Grant No. 2020006), the Top Talents Project of Gansu Province (Grant No. 2023-9), the Natural Science Foundation of Gansu Province (Grant No. 23JRRA677), the Longyuan Youth Talent Program of Gansu Province (Grant No. 2022-5), and the Top Talents Project of Gansu Province (Grant No. 2024-96).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CAFCancer-associated fibroblasts
ECMExtracellular matrix
NSCLCNon-small cell lung cancer
SCLCSmall cell lung cancer
BM-MSC Bone marrow-derived mesenchymal stem cells
EGFREpidermal growth factor receptor
ALKActivin receptor-like kinase
ROSReactive oxygen species
TMETumor microenvironment
TIMETumor immune microenvironment
TGF-βTransforming growth factor-β
α-SMAα-smooth muscle actin
FAPFibroblast activation protein
VCAM-1Vascular cell adhesion molecule-1
CAF-CMCAF-conditioned medium
NF-CMNormal fibroblast-conditioned medium
EMTEpithelial-mesenchymal transition
SDF-1stromal cell-derived factor-1
EndMTEndothelial-mesenchymal transition
FSP1Fibroblast-specific protein-1
HMEC-1Human microvascular endothelial cells
ILKIntegrin-linked kinase
MRTFMyocardin-related transcription factor
FAPIsFAP inhibitors
CXCL12C-X-C Motif Chemokine Ligand 12
IL-6Interleukin-6
COL1A1Collagen type I alpha 1 chain
PDGFRPlatelet-derived growth factor receptor
SCCLung squamous cell carcinoma
myCAFsMyofibroblast-like cancer-associated fibroblasts
PDACPancreatic ductal adenocarcinoma
HNSCCHead and neck squamous cell carcinoma
LUADLung adenocarcinoma
MMPsMatrix metalloproteinases
iCAFsInflammatory CAFs
HGFHepatocyte growth factor
IMCImaging mass cytometry
non-NENon-neuroendocrine
apCAFAntigen-presenting CAFs
mCAFMatrix CAF
TAMsTumor-associated macrophages
TNFTumor necrosis factor
MMTMacrophage-myofibroblast transformation
TANsTumor-associated neutrophils
CXCR2C-X-C chemokine receptor 2
G-CSFGranulocyte colony-stimulating factor
GM-CSFGranulocyte-macrophage colony-stimulating factor
NKNatural killer
IFN-γInterferon-γ
DCsDendritic cells
TDO2Tryptophan 2,3-dioxygenase
MDSCsMyeloid-derived suppressor cells
Fgl2Fibrinogen-like protein 2
PMN-MDSCPolymorphonuclear myeloid-derived suppressor cells
M-MDSCsMononuclear myeloid-derived suppressor cells
ThHelper T
CTLCytotoxic T lymphocytes
Foxp3Forkhead box P3
CFRCD8+ T cell/CAF ratio
MMPsMatrix metalloproteinases
RTRadiotherapy
RIPFRadiation-induced pulmonary fibrosis
HD-RTHigh-dose radiotherapy
MCsMast cells
SCFStem cell factor

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  2. Inamura, K. Lung Cancer: Understanding Its Molecular Pathology and the 2015 WHO Classification. Front. Oncol. 2017, 7, 193. [Google Scholar]
  3. Vinod, S.K.; Hau, E. Radiotherapy treatment for lung cancer: Current status and future directions. Respirology 2020, 25 (Suppl. S2), 61–71. [Google Scholar] [CrossRef]
  4. Duma, N.; Santana-Davila, R.; Molina, J.R. Non-Small Cell Lung Cancer: Epidemiology, Screening, Diagnosis, and Treatment. Mayo Clin. Proc. 2019, 94, 1623–1640. [Google Scholar] [CrossRef] [PubMed]
  5. Roszkowska, M. Multilevel Mechanisms of Cancer Drug Resistance. Int. J. Mol. Sci. 2024, 25, 12402. [Google Scholar] [CrossRef]
  6. Bejarano, L.; Jordāo, M.J.C.; Joyce, J.A. Therapeutic Targeting of the Tumor Microenvironment. Cancer Discov. 2021, 11, 933–959. [Google Scholar] [CrossRef]
  7. Tan, Z.; Xue, H.; Sun, Y.; Zhang, C.; Song, Y.; Qi, Y. The Role of Tumor Inflammatory Microenvironment in Lung Cancer. Front. Pharmacol. 2021, 12, 688625. [Google Scholar] [CrossRef]
  8. Murciano-Goroff, Y.R.; Warner, A.B.; Wolchok, J.D. The future of cancer immunotherapy: Microenvironment-targeting combinations. Cell Res. 2020, 30, 507–519. [Google Scholar] [CrossRef]
  9. O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef]
  10. Genova, C.; Dellepiane, C.; Carrega, P.; Sommariva, S.; Ferlazzo, G.; Pronzato, P.; Gangemi, R.; Filaci, G.; Coco, S.; Croce, M. Therapeutic Implications of Tumor Microenvironment in Lung Cancer: Focus on Immune Checkpoint Blockade. Front. Immunol. 2021, 12, 799455. [Google Scholar] [CrossRef]
  11. Larroquette, M.; Guegan, J.P.; Besse, B.; Cousin, S.; Brunet, M.; Le Moulec, S.; Le Loarer, F.; Rey, C.; Soria, J.C.; Barlesi, F.; et al. Spatial transcriptomics of macrophage infiltration in non-small cell lung cancer reveals determinants of sensitivity and resistance to anti-PD1/PD-L1 antibodies. J. Immunother. Cancer 2022, 10, e003890. [Google Scholar] [CrossRef]
  12. Du, X.; Yang, S.; Bian, J.; Zhang, Y.; Wang, Y.; Lv, Z. Role of vascular endothelial growth factor D in lung adenocarcinoma immunotherapy response. Am. J. Transl. Res. 2024, 16, 2263–2277. [Google Scholar] [CrossRef] [PubMed]
  13. Sun, C.; Yin, M.; Cheng, Y.; Kuang, Z.; Liu, X.; Wang, G.; Wang, X.; Yuan, K.; Min, W.; Dong, J.; et al. Novel Small-Molecule PD-L1 Inhibitor Induces PD-L1 Internalization and Optimizes the Immune Microenvironment. J. Med. Chem. 2023, 66, 2064–2083. [Google Scholar] [CrossRef] [PubMed]
  14. Lu, C.; Gao, Z.; Wu, D.; Zheng, J.; Hu, C.; Huang, D.; He, C.; Liu, Y.; Lin, C.; Peng, T.; et al. Understanding the dynamics of TKI-induced changes in the tumor immune microenvironment for improved therapeutic effect. J. Immunother. Cancer 2024, 12, e009165. [Google Scholar] [CrossRef]
  15. Gao, Y.H.; Lovreković, V.; Kussayeva, A.; Chen, D.Y.; Margetić, D.; Chen, Z.L. The photodynamic activities of dimethyl 13(1)-[2-(guanidinyl)ethylamino] chlorin e(6) photosensitizers in A549 tumor. Eur. J. Med. Chem. 2019, 177, 144–152. [Google Scholar] [CrossRef]
  16. Guo, D.; Xu, S.; Huang, Y.; Jiang, H.; Yasen, W.; Wang, N.; Su, Y.; Qian, J.; Li, J.; Zhang, C.; et al. Platinum(IV) complex-based two-in-one polyprodrug for a combinatorial chemo-photodynamic therapy. Biomaterials 2018, 177, 67–77. [Google Scholar] [CrossRef] [PubMed]
  17. Ai, X.; Mu, J.; Xing, B. Recent Advances of Light-Mediated Theranostics. Theranostics 2016, 6, 2439–2457. [Google Scholar] [CrossRef]
  18. Dai, J.; Li, Y.; Long, Z.; Jiang, R.; Zhuang, Z.; Wang, Z.; Zhao, Z.; Lou, X.; Xia, F.; Tang, B.Z. Efficient Near-Infrared Photosensitizer with Aggregation-Induced Emission for Imaging-Guided Photodynamic Therapy in Multiple Xenograft Tumor Models. ACS Nano 2020, 14, 854–866. [Google Scholar] [CrossRef]
  19. Zhang, Q.; Wu, L.; Liu, S.; Chen, Q.; Zeng, L.; Chen, X.; Zhang, Q. Targeted nanobody complex enhanced photodynamic therapy for lung cancer by overcoming tumor microenvironment. Cancer Cell Int. 2020, 20, 570. [Google Scholar] [CrossRef]
  20. Zhao, Y.; Guo, S.; Deng, J.; Shen, J.; Du, F.; Wu, X.; Chen, Y.; Li, M.; Chen, M.; Li, X.; et al. VEGF/VEGFR-Targeted Therapy and Immunotherapy in Non-small Cell Lung Cancer: Targeting the Tumor Microenvironment. Int. J. Biol. Sci. 2022, 18, 3845–3858. [Google Scholar] [CrossRef]
  21. Chen, F.; Zhuang, X.; Lin, L.; Yu, P.; Wang, Y.; Shi, Y.; Hu, G.; Sun, Y. New horizons in tumor microenvironment biology: Challenges and opportunities. BMC Med. 2015, 13, 45. [Google Scholar] [CrossRef]
  22. Paluskievicz, C.M.; Cao, X.; Abdi, R.; Zheng, P.; Liu, Y.; Bromberg, J.S. T Regulatory Cells and Priming the Suppressive Tumor Microenvironment. Front. Immunol. 2019, 10, 2453. [Google Scholar] [CrossRef]
  23. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef]
  24. Schulz, M.; Salamero-Boix, A.; Niesel, K.; Alekseeva, T.; Sevenich, L. Microenvironmental Regulation of Tumor Progression and Therapeutic Response in Brain Metastasis. Front. Immunol. 2019, 10, 1713. [Google Scholar] [CrossRef]
  25. Giraldo, N.A.; Sanchez-Salas, R.; Peske, J.D.; Vano, Y.; Becht, E.; Petitprez, F.; Validire, P.; Ingels, A.; Cathelineau, X.; Fridman, W.H.; et al. The clinical role of the TME in solid cancer. Br. J. Cancer 2019, 120, 45–53. [Google Scholar] [CrossRef] [PubMed]
  26. Li, H.; Fan, X.; Houghton, J. Tumor microenvironment: The role of the tumor stroma in cancer. J. Cell Biochem. 2007, 101, 805–815. [Google Scholar] [CrossRef]
  27. Chen, Z.; Zhou, L.; Liu, L.; Hou, Y.; Xiong, M.; Yang, Y.; Hu, J.; Chen, K. Single-cell RNA sequencing highlights the role of inflammatory cancer-associated fibroblasts in bladder urothelial carcinoma. Nat. Commun. 2020, 11, 5077. [Google Scholar] [CrossRef] [PubMed]
  28. Mao, X.; Xu, J.; Wang, W.; Liang, C.; Hua, J.; Liu, J.; Zhang, B.; Meng, Q.; Yu, X.; Shi, S. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Mol. Cancer 2021, 20, 131. [Google Scholar] [CrossRef]
  29. Lv, B.; Wang, Y.; Ma, D.; Cheng, W.; Liu, J.; Yong, T.; Chen, H.; Wang, C. Immunotherapy: Reshape the Tumor Immune Microenvironment. Front. Immunol. 2022, 13, 844142. [Google Scholar] [CrossRef]
  30. Chen, Y.; McAndrews, K.M.; Kalluri, R. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat. Rev. Clin. Oncol. 2021, 18, 792–804. [Google Scholar]
  31. Desbois, M.; Wang, Y. Cancer-associated fibroblasts: Key players in shaping the tumor immune microenvironment. Immunol. Rev. 2021, 302, 241–258. [Google Scholar] [CrossRef] [PubMed]
  32. Xu, Y.; Li, W.; Lin, S.; Liu, B.; Wu, P.; Li, L. Fibroblast diversity and plasticity in the tumor microenvironment: Roles in immunity and relevant therapies. Cell Commun. Signal 2023, 21, 234. [Google Scholar]
  33. Pei, L.; Liu, Y.; Liu, L.; Gao, S.; Gao, X.; Feng, Y.; Sun, Z.; Zhang, Y.; Wang, C. Roles of cancer-associated fibroblasts (CAFs) in anti- PD-1/PD-L1 immunotherapy for solid cancers. Mol. Cancer 2023, 22, 29. [Google Scholar] [PubMed]
  34. Raaijmakers, K.; Adema, G.J.; Bussink, J.; Ansems, M. Cancer-associated fibroblasts, tumor and radiotherapy: Interactions in the tumor micro-environment. J. Exp. Clin. Cancer Res. 2024, 43, 323. [Google Scholar]
  35. Bremnes, R.M.; Dønnem, T.; Al-Saad, S.; Al-Shibli, K.; Andersen, S.; Sirera, R.; Camps, C.; Marinez, I.; Busund, L.T. The role of tumor stroma in cancer progression and prognosis: Emphasis on carcinoma-associated fibroblasts and non-small cell lung cancer. J. Thorac. Oncol. 2011, 6, 209–217. [Google Scholar] [CrossRef] [PubMed]
  36. Öhlund, D.; Elyada, E.; Tuveson, D. Fibroblast heterogeneity in the cancer wound. J. Exp. Med. 2014, 211, 1503–1523. [Google Scholar]
  37. Navab, R.; Strumpf, D.; Bandarchi, B.; Zhu, C.Q.; Pintilie, M.; Ramnarine, V.R.; Ibrahimov, E.; Radulovich, N.; Leung, L.; Barczyk, M.; et al. Prognostic gene-expression signature of carcinoma-associated fibroblasts in non-small cell lung cancer. Proc. Natl. Acad. Sci. USA 2011, 108, 7160–7165. [Google Scholar]
  38. Albrengues, J.; Bertero, T.; Grasset, E.; Bonan, S.; Maiel, M.; Bourget, I.; Philippe, C.; Herraiz Serrano, C.; Benamar, S.; Croce, O.; et al. Epigenetic switch drives the conversion of fibroblasts into proinvasive cancer-associated fibroblasts. Nat. Commun. 2015, 6, 10204. [Google Scholar] [CrossRef]
  39. Zhou, Z.; Zhou, Q.; Wu, X.; Xu, S.; Hu, X.; Tao, X.; Li, B.; Peng, J.; Li, D.; Shen, L.; et al. VCAM-1 secreted from cancer-associated fibroblasts enhances the growth and invasion of lung cancer cells through AKT and MAPK signaling. Cancer Lett. 2020, 473, 62–73. [Google Scholar] [CrossRef]
  40. Procopio, M.G.; Laszlo, C.; Al Labban, D.; Kim, D.E.; Bordignon, P.; Jo, S.H.; Goruppi, S.; Menietti, E.; Ostano, P.; Ala, U.; et al. Combined CSL and p53 downregulation promotes cancer-associated fibroblast activation. Nat. Cell Biol. 2015, 17, 1193–1204. [Google Scholar]
  41. Iwano, M.; Plieth, D.; Danoff, T.M.; Xue, C.; Okada, H.; Neilson, E.G. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J. Clin. Investig. 2002, 110, 341–350. [Google Scholar] [PubMed]
  42. Saito, A.; Horie, M.; Nagase, T. TGF-β Signaling in Lung Health and Disease. Int. J. Mol. Sci. 2018, 19, 2460. [Google Scholar] [CrossRef] [PubMed]
  43. Yang, F.; Yan, Y.; Yang, Y.; Hong, X.; Wang, M.; Yang, Z.; Liu, B.; Ye, L. MiR-210 in exosomes derived from CAFs promotes non-small cell lung cancer migration and invasion through PTEN/PI3K/AKT pathway. Cell. Signal. 2020, 73, 109675. [Google Scholar] [CrossRef]
  44. You, J.; Li, M.; Cao, L.M.; Gu, Q.H.; Deng, P.B.; Tan, Y.; Hu, C.P. Snail1-dependent cancer-associated fibroblasts induce epithelial-mesenchymal transition in lung cancer cells via exosomes. QJM 2019, 112, 581–590. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, L.; Cao, L.; Wang, H.; Liu, B.; Zhang, Q.; Meng, Z.; Wu, X.; Zhou, Q.; Xu, K. Cancer-associated fibroblasts enhance metastatic potential of lung cancer cells through IL-6/STAT3 signaling pathway. Oncotarget 2017, 8, 76116–76128. [Google Scholar] [CrossRef]
  46. Wang, Y.; Lan, W.; Xu, M.; Song, J.; Mao, J.; Li, C.; Du, X.; Jiang, Y.; Li, E.; Zhang, R.; et al. Cancer-associated fibroblast-derived SDF-1 induces epithelial-mesenchymal transition of lung adenocarcinoma via CXCR4/β-catenin/PPARδ signalling. Cell Death Dis. 2021, 12, 214. [Google Scholar]
  47. Shintani, Y.; Abulaiti, A.; Kimura, T.; Funaki, S.; Nakagiri, T.; Inoue, M.; Sawabata, N.; Minami, M.; Morii, E.; Okumura, M. Pulmonary fibroblasts induce epithelial mesenchymal transition and some characteristics of stem cells in non-small cell lung cancer. Ann. Thorac. Surg. 2013, 96, 425–433. [Google Scholar] [CrossRef]
  48. Zeisberg, E.M.; Potenta, S.; Xie, L.; Zeisberg, M.; Kalluri, R. Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res. 2007, 67, 10123–10128. [Google Scholar] [CrossRef]
  49. Ciszewski, W.M.; Sobierajska, K.; Wawro, M.E.; Klopocka, W.; Chefczyńska, N.; Muzyczuk, A.; Siekacz, K.; Wujkowska, A.; Niewiarowska, J. The ILK-MMP9-MRTF axis is crucial for EndMT differentiation of endothelial cells in a tumor microenvironment. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 2283–2296. [Google Scholar] [CrossRef]
  50. Choi, S.H.; Nam, J.K.; Kim, B.Y.; Jang, J.; Jin, Y.B.; Lee, H.J.; Park, S.; Ji, Y.H.; Cho, J.; Lee, Y.J. HSPB1 Inhibits the Endothelial-to-Mesenchymal Transition to Suppress Pulmonary Fibrosis and Lung Tumorigenesis. Cancer Res. 2016, 76, 1019–1030. [Google Scholar] [CrossRef]
  51. Wen, S.; Niu, Y.; Yeh, S.; Chang, C. BM-MSCs promote prostate cancer progression via the conversion of normal fibroblasts to cancer-associated fibroblasts. Int. J. Oncol. 2015, 47, 719–727. [Google Scholar] [PubMed]
  52. Iwamoto, C.; Ohuchida, K.; Shinkawa, T.; Okuda, S.; Otsubo, Y.; Okumura, T.; Sagara, A.; Koikawa, K.; Ando, Y.; Shindo, K.; et al. Bone marrow-derived macrophages converted into cancer-associated fibroblast-like cells promote pancreatic cancer progression. Cancer Lett. 2021, 512, 15–27. [Google Scholar] [CrossRef]
  53. Moratin, H.; Böhm, S.; Hagen, R.; Scherzad, A.; Hackenberg, S. Influence of Wound Fluid on the Transdifferentiation of Human Mesenchymal Bone Marrow Stem Cells into Cancer-Associated Fibroblasts. Cells Tissues Organs 2023, 212, 304–316. [Google Scholar] [CrossRef] [PubMed]
  54. Quante, M.; Tu, S.P.; Tomita, H.; Gonda, T.; Wang, S.S.; Takashi, S.; Baik, G.H.; Shibata, W.; Diprete, B.; Betz, K.S.; et al. Bone marrow-derived myofibroblasts contribute to the mesenchymal stem cell niche and promote tumor growth. Cancer Cell 2011, 19, 257–272. [Google Scholar] [PubMed]
  55. Spaeth, E.L.; Labaff, A.M.; Toole, B.P.; Klopp, A.; Andreeff, M.; Marini, F.C. Mesenchymal CD44 expression contributes to the acquisition of an activated fibroblast phenotype via TWIST activation in the tumor microenvironment. Cancer Res. 2013, 73, 5347–5359. [Google Scholar] [PubMed]
  56. Peng, Y.; Li, Z.; Li, Z. GRP78 secreted by tumor cells stimulates differentiation of bone marrow mesenchymal stem cells to cancer-associated fibroblasts. Biochem. Biophys. Res. Commun. 2013, 440, 558–563. [Google Scholar] [CrossRef] [PubMed]
  57. Sha, M.; Jeong, S.; Qiu, B.J.; Tong, Y.; Xia, L.; Xu, N.; Zhang, J.J.; Xia, Q. Isolation of cancer-associated fibroblasts and its promotion to the progression of intrahepatic cholangiocarcinoma. Cancer Med. 2018, 7, 4665–4677. [Google Scholar] [CrossRef]
  58. Zawieracz, K.; Eckert, M.A. Isolation of Normal and Cancer-Associated Fibroblasts. Methods Mol. Biol. 2022, 2424, 155–165. [Google Scholar] [CrossRef]
  59. Sharon, Y.; Alon, L.; Glanz, S.; Servais, C.; Erez, N. Isolation of normal and cancer-associated fibroblasts from fresh tissues by Fluorescence Activated Cell Sorting (FACS). J. Vis. Exp. 2013, 71, e4425. [Google Scholar] [CrossRef]
  60. Jiang, R.; Agrawal, S.; Aghaamoo, M.; Parajuli, R.; Agrawal, A.; Lee, A.P. Rapid isolation of circulating cancer associated fibroblasts by acoustic microstreaming for assessing metastatic propensity of breast cancer patients. Lab Chip 2021, 21, 875–887. [Google Scholar] [CrossRef]
  61. Nurmik, M.; Ullmann, P.; Rodriguez, F.; Haan, S.; Letellier, E. In search of definitions: Cancer-associated fibroblasts and their markers. Int. J. Cancer 2020, 146, 895–905. [Google Scholar] [CrossRef] [PubMed]
  62. Hu, D.; Li, Z.; Zheng, B.; Lin, X.; Pan, Y.; Gong, P.; Zhuo, W.; Hu, Y.; Chen, C.; Chen, L.; et al. Cancer-associated fibroblasts in breast cancer: Challenges and opportunities. Cancer Commun. 2022, 42, 401–434. [Google Scholar] [CrossRef]
  63. Peltier, A.; Seban, R.D.; Buvat, I.; Bidard, F.C.; Mechta-Grigoriou, F. Fibroblast heterogeneity in solid tumors: From single cell analysis to whole-body imaging. Semin. Cancer Biol. 2022, 86 Pt 3, 262–272. [Google Scholar] [CrossRef] [PubMed]
  64. Mathieson, L.; Koppensteiner, L.; Dorward, D.A.; O’Connor, R.A.; Akram, A.R. Cancer-associated fibroblasts expressing fibroblast activation protein and podoplanin in non-small cell lung cancer predict poor clinical outcome. Br. J. Cancer 2024, 130, 1758–1769. [Google Scholar] [CrossRef]
  65. Sugimoto, H.; Mundel, T.M.; Kieran, M.W.; Kalluri, R. Identification of fibroblast heterogeneity in the tumor microenvironment. Cancer Biol. Ther. 2006, 5, 1640–1646. [Google Scholar] [CrossRef]
  66. Samson, S.C.; Rojas, A.; Zitnay, R.G.; Carney, K.R.; Hettinga, W.; Schaelling, M.C.; Sicard, D.; Zhang, W.; Gilbert-Ross, M.; Dy, G.K.; et al. Tenascin-C in the early lung cancer tumor microenvironment promotes progression through integrin αvβ1 and FAK. bioRxiv 2024. [Google Scholar] [CrossRef]
  67. Hu, W.W.; Chen, P.C.; Chen, J.M.; Wu, Y.M.; Liu, P.Y.; Lu, C.H.; Lin, Y.F.; Tang, C.H.; Chao, C.C. Periostin promotes epithelial-mesenchymal transition via the MAPK/miR-381 axis in lung cancer. Oncotarget 2017, 8, 62248–62260. [Google Scholar] [CrossRef]
  68. Li, K.; Wang, R.; Liu, G.W.; Peng, Z.Y.; Wang, J.C.; Xiao, G.D.; Tang, S.C.; Du, N.; Zhang, J.; Zhang, J.; et al. Refining the optimal CAF cluster marker for predicting TME-dependent survival expectancy and treatment benefits in NSCLC patients. Sci. Rep. 2024, 14, 16766. [Google Scholar] [CrossRef]
  69. Zhu, G.; Wang, Y.; Wang, Y.; Huang, H.; Li, B.; Chen, P.; Chen, C.; Zhang, H.; Li, Y.; Liu, H.; et al. Myofibroblasts derived type V collagen promoting tissue mechanical stress and facilitating metastasis and therapy resistance of lung adenocarcinoma cells. Cell Death Dis. 2024, 15, 493. [Google Scholar] [CrossRef]
  70. Zhang, Q.; Yang, J.; Bai, J.; Ren, J. Reverse of non-small cell lung cancer drug resistance induced by cancer-associated fibroblasts via a paracrine pathway. Cancer Sci. 2018, 109, 944–955. [Google Scholar] [CrossRef]
  71. Sugai, M.; Yanagawa, N.; Shikanai, S.; Hashimoto, M.; Saikawa, H.; Osakabe, M.; Saito, H.; Maemondo, M.; Sugai, T. Correlation of tumor microenvironment-related markers with clinical outcomes in patients with squamous cell carcinoma of the lung. Transl. Lung Cancer Res. 2022, 11, 975–990. [Google Scholar] [CrossRef] [PubMed]
  72. Puré, E.; Blomberg, R. Pro-tumorigenic roles of fibroblast activation protein in cancer: Back to the basics. Oncogene 2018, 37, 4343–4357. [Google Scholar]
  73. Kilvaer, T.K.; Rakaee, M.; Hellevik, T.; Østman, A.; Strell, C.; Bremnes, R.M.; Busund, L.T.; Dønnem, T.; Martinez-Zubiaurre, I. Tissue analyses reveal a potential immune-adjuvant function of FAP-1 positive fibroblasts in non-small cell lung cancer. PLoS ONE 2018, 13, e0192157. [Google Scholar]
  74. Chen, L.; Chen, M.; Han, Z.; Jiang, F.; Xu, C.; Qin, Y.; Ding, N.; Liu, Y.; Zhang, T.; An, Z.; et al. Clinical significance of FAP-α on microvessel and lymphatic vessel density in lung squamous cell carcinoma. J. Clin. Pathol. 2018, 71, 721–728. [Google Scholar] [CrossRef]
  75. Fang, Y.; Wang, Y.J.; Zhao, H.L.; Huang, X.; Fang, Y.N.; Chen, W.Y.; Han, R.Z.; Zhao, A.; Gao, J.M. Development of FAP-Targeted Chimeric Antigen Receptor NK-92 Cells for Non-Small Cell Lung Cancer. Discov. Med. 2023, 35, 405–417. [Google Scholar] [CrossRef] [PubMed]
  76. Li, H.; Qiu, L.; Liu, Q.; Ma, Z.; Xie, X.; Luo, Y.; Wu, X. Senescent Fibroblasts Generate a CAF Phenotype through the Stat3 Pathway. Genes 2022, 13, 1579. [Google Scholar] [CrossRef]
  77. Lu, Y.; Li, H.; Zhao, P.; Tian, L.; Liu, Y.; Sun, X.; Cheng, Y. Dynamic phenotypic reprogramming and chemoresistance induced by lung fibroblasts in small cell lung cancer. Sci. Rep. 2024, 14, 2884. [Google Scholar] [CrossRef]
  78. Paulsson, J.; Ehnman, M.; Östman, A. PDGF receptors in tumor biology: Prognostic and predictive potential. Future Oncol. 2014, 10, 1695–1708. [Google Scholar] [CrossRef]
  79. Akiyama, T.; Yasuda, T.; Uchihara, T.; Yasuda-Yoshihara, N.; Tan, B.J.Y.; Yonemura, A.; Semba, T.; Yamasaki, J.; Komohara, Y.; Ohnishi, K.; et al. Stromal Reprogramming through Dual PDGFRα/β Blockade Boosts the Efficacy of Anti-PD-1 Immunotherapy in Fibrotic Tumors. Cancer Res. 2023, 83, 753–770. [Google Scholar] [CrossRef]
  80. Sun, C.; Zhang, K.; Ni, C.; Wan, J.; Duan, X.; Lou, X.; Yao, X.; Li, X.; Wang, M.; Gu, Z.; et al. Transgelin promotes lung cancer progression via activation of cancer-associated fibroblasts with enhanced IL-6 release. Oncogenesis 2023, 12, 18. [Google Scholar]
  81. Zeltz, C.; Alam, J.; Liu, H.; Erusappan, P.M.; Hoschuetzky, H.; Molven, A.; Parajuli, H.; Cukierman, E.; Costea, D.E.; Lu, N.; et al. α11β1 Integrin is Induced in a Subset of Cancer-Associated Fibroblasts in Desmoplastic Tumor Stroma and Mediates In Vitro Cell Migration. Cancers 2019, 11, 765. [Google Scholar] [CrossRef]
  82. Vasiukov, G.; Zou, Y.; Senosain, M.F.; Rahman, J.S.M.; Antic, S.; Young, K.M.; Grogan, E.L.; Kammer, M.N.; Maldonado, F.; Reinhart-King, C.A.; et al. Cancer-associated fibroblasts in early-stage lung adenocarcinoma correlate with tumor aggressiveness. Sci. Rep. 2023, 13, 17604. [Google Scholar]
  83. Nabeshima, K.; Inoue, T.; Shimao, Y.; Sameshima, T. Matrix metalloproteinases in tumor invasion: Role for cell migration. Pathol. Int. 2002, 52, 255–264. [Google Scholar] [CrossRef] [PubMed]
  84. Chen, B.; Chan, W.N.; Xie, F.; Mui, C.W.; Liu, X.; Cheung, A.H.K.; Lung, R.W.M.; Chow, C.; Zhang, Z.; Fang, C.; et al. The molecular classification of cancer-associated fibroblasts on a pan-cancer single-cell transcriptional atlas. Clin. Transl. Med. 2023, 13, e1516. [Google Scholar] [CrossRef] [PubMed]
  85. Pan, X.; Zhou, J.; Xiao, Q.; Fujiwara, K.; Zhang, M.; Mo, G.; Gong, W.; Zheng, L. Cancer-associated fibroblast heterogeneity is associated with organ-specific metastasis in pancreatic ductal adenocarcinoma. J. Hematol. Oncol. 2021, 14, 184. [Google Scholar] [CrossRef] [PubMed]
  86. Xu, K.; Wang, H.; Zou, Y.X.; Zhang, H.H.; Wang, Y.N.; Ren, X.R.; Wang, H.Q.; Xu, Y.H.; Li, J.J.; Tang, H.; et al. Distinct fibroblast subpopulations associated with bone, brain or intrapulmonary metastasis in advanced non-small-cell lung cancer. Clin. Transl. Med. 2024, 14, e1605. [Google Scholar] [CrossRef]
  87. Cords, L.; Engler, S.; Haberecker, M.; Rüschoff, J.H.; Moch, H.; de Souza, N.; Bodenmiller, B. Cancer-associated fibroblast phenotypes are associated with patient outcome in non-small cell lung cancer. Cancer Cell 2024, 42, 396–412.e5. [Google Scholar] [CrossRef]
  88. Zhao, Y.; Jia, Y.; Wang, J.; Chen, X.; Han, J.; Zhen, S.; Yin, S.; Lv, W.; Yu, F.; Wang, J.; et al. circNOX4 activates an inflammatory fibroblast niche to promote tumor growth and metastasis in NSCLC via FAP/IL-6 axis. Mol. Cancer 2024, 23, 47. [Google Scholar]
  89. Lu, Y.; Li, H.; Zhao, P.; Wang, X.; Shao, W.; Liu, Y.; Tian, L.; Zhong, R.; Liu, H.; Cheng, Y. Crosstalk between cancer-associated fibroblasts and non-neuroendocrine tumor cells in small cell lung cancer involves in glycolysis and antigen-presenting features. Mol. Med. 2024, 30, 274. [Google Scholar] [CrossRef]
  90. Ma, C.; Yang, C.; Peng, A.; Sun, T.; Ji, X.; Mi, J.; Wei, L.; Shen, S.; Feng, Q. Pan-cancer spatially resolved single-cell analysis reveals the crosstalk between cancer-associated fibroblasts and tumor microenvironment. Mol. Cancer 2023, 22, 170. [Google Scholar] [CrossRef]
  91. Wu, F.; Fan, J.; He, Y.; Xiong, A.; Yu, J.; Li, Y.; Zhang, Y.; Zhao, W.; Zhou, F.; Li, W.; et al. Single-cell profiling of tumor heterogeneity and the microenvironment in advanced non-small cell lung cancer. Nat. Commun. 2021, 12, 2540. [Google Scholar] [CrossRef]
  92. Hu, H.; Piotrowska, Z.; Hare, P.J.; Chen, H.; Mulvey, H.E.; Mayfield, A.; Noeen, S.; Kattermann, K.; Greenberg, M.; Williams, A.; et al. Three subtypes of lung cancer fibroblasts define distinct therapeutic paradigms. Cancer Cell 2021, 39, 1531–1547.e1510. [Google Scholar] [CrossRef] [PubMed]
  93. Hanley, C.J.; Waise, S.; Ellis, M.J.; Lopez, M.A.; Pun, W.Y.; Taylor, J.; Parker, R.; Kimbley, L.M.; Chee, S.J.; Shaw, E.C.; et al. Single-cell analysis reveals prognostic fibroblast subpopulations linked to molecular and immunological subtypes of lung cancer. Nat. Commun. 2023, 14, 387. [Google Scholar] [CrossRef]
  94. Si, Y.; Zhao, Z.; Meng, X.; Zhao, K. RNA-seq and bulk RNA-seq data analysis of cancer-related fibroblasts (CAF) in LUAD to construct a CAF-based risk signature. Sci. Rep. 2024, 14, 23243. [Google Scholar] [CrossRef] [PubMed]
  95. Bota-Rabassedas, N.; Banerjee, P.; Niu, Y.; Cao, W.; Luo, J.; Xi, Y.; Tan, X.; Sheng, K.; Ahn, Y.H.; Lee, S.; et al. Contextual cues from cancer cells govern cancer-associated fibroblast heterogeneity. Cell Rep. 2021, 35, 109009. [Google Scholar] [CrossRef]
  96. Zhang, X.; Xiao, Q.; Zhang, C.; Zhou, Q.; Xu, T. Construction of a prognostic model with CAFs for predicting the prognosis and immunotherapeutic response of lung squamous cell carcinoma. J. Cell. Mol. Med. 2024, 28, e18262. [Google Scholar] [CrossRef]
  97. Fang, X.; Li, D.; Wan, S.; Hu, J.; Zhang, P.; Jie, D.; Chen, L.; Jiang, G.; Song, N. Insights into the heterogeneity of the tumor microenvironment in lung adenocarcinoma and squamous carcinoma through single-cell transcriptomic analysis: Implications for distinct immunotherapy outcomes. J. Gene Med. 2024, 26, e3694. [Google Scholar] [CrossRef]
  98. Yang, Y.; Shao, X.; Li, Z.; Zhang, L.; Yang, B.; Jin, B.; Hu, X.; Qu, X.; Che, X.; Liu, Y. Prognostic heterogeneity of Ki67 in non-small cell lung cancer: A comprehensive reappraisal on immunohistochemistry and transcriptional data. J. Cell. Mol. Med. 2024, 28, e18521. [Google Scholar] [CrossRef]
  99. Liu, T.; Han, C.; Wang, S.; Fang, P.; Ma, Z.; Xu, L.; Yin, R. Cancer-associated fibroblasts: An emerging target of anti-cancer immunotherapy. J. Hematol. Oncol. 2019, 12, 86. [Google Scholar] [CrossRef]
  100. Harper, J.; Sainson, R.C. Regulation of the anti-tumour immune response by cancer-associated fibroblasts. Semin. Cancer Biol. 2014, 25, 69–77. [Google Scholar] [CrossRef]
  101. Ziani, L.; Chouaib, S.; Thiery, J. Alteration of the Antitumor Immune Response by Cancer-Associated Fibroblasts. Front. Immunol. 2018, 9, 414. [Google Scholar] [CrossRef] [PubMed]
  102. Kim, R.; Emi, M.; Tanabe, K. Cancer immunosuppression and autoimmune disease: Beyond immunosuppressive networks for tumour immunity. Immunology 2006, 119, 254–264. [Google Scholar] [CrossRef]
  103. Wang, S.; Fan, G.; Li, L.; He, Y.; Lou, N.; Xie, T.; Dai, L.; Gao, R.; Yang, M.; Shi, Y.; et al. Integrative analyses of bulk and single-cell RNA-seq identified cancer-associated fibroblasts-related signature as a prognostic factor for immunotherapy in NSCLC. Cancer Immunol. Immunother. 2023, 72, 2423–2442. [Google Scholar] [CrossRef]
  104. Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25, 677–686. [Google Scholar] [CrossRef]
  105. Shapouri-Moghaddam, A.; Mohammadian, S.; Vazini, H.; Taghadosi, M.; Esmaeili, S.A.; Mardani, F.; Seifi, B.; Mohammadi, A.; Afshari, J.T.; Sahebkar, A. Macrophage plasticity, polarization, and function in health and disease. J. Cell. Physiol. 2018, 233, 6425–6440. [Google Scholar] [CrossRef]
  106. Allavena, P.; Sica, A.; Garlanda, C.; Mantovani, A. The Yin-Yang of tumor-associated macrophages in neoplastic progression and immune surveillance. Immunol. Rev. 2008, 222, 155–161. [Google Scholar] [CrossRef] [PubMed]
  107. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef] [PubMed]
  108. Herrera, M.; Herrera, A.; Domínguez, G.; Silva, J.; García, V.; García, J.M.; Gómez, I.; Soldevilla, B.; Muñoz, C.; Provencio, M.; et al. Cancer-associated fibroblast and M2 macrophage markers together predict outcome in colorectal cancer patients. Cancer Sci. 2013, 104, 437–444. [Google Scholar] [CrossRef]
  109. Tan, B.; Shi, X.; Zhang, J.; Qin, J.; Zhang, N.; Ren, H.; Qian, M.; Siwko, S.; Carmon, K.; Liu, Q.; et al. Inhibition of Rspo-Lgr4 Facilitates Checkpoint Blockade Therapy by Switching Macrophage Polarization. Cancer Res. 2018, 78, 4929–4942. [Google Scholar] [CrossRef]
  110. Comito, G.; Giannoni, E.; Segura, C.P.; Barcellos-de-Souza, P.; Raspollini, M.R.; Baroni, G.; Lanciotti, M.; Serni, S.; Chiarugi, P. Cancer-associated fibroblasts and M2-polarized macrophages synergize during prostate carcinoma progression. Oncogene 2014, 33, 2423–2431. [Google Scholar] [CrossRef]
  111. Cohen, N.; Shani, O.; Raz, Y.; Sharon, Y.; Hoffman, D.; Abramovitz, L.; Erez, N. Fibroblasts drive an immunosuppressive and growth-promoting microenvironment in breast cancer via secretion of Chitinase 3-like 1. Oncogene 2017, 36, 4457–4468. [Google Scholar] [CrossRef] [PubMed]
  112. Hegab, A.E.; Ozaki, M.; Kameyama, N.; Gao, J.; Kagawa, S.; Yasuda, H.; Soejima, K.; Yin, Y.; Guzy, R.D.; Nakamura, Y.; et al. Effect of FGF/FGFR pathway blocking on lung adenocarcinoma and its cancer-associated fibroblasts. J. Pathol. 2019, 249, 193–205. [Google Scholar] [CrossRef]
  113. Suzuki, J.; Aokage, K.; Neri, S.; Sakai, T.; Hashimoto, H.; Su, Y.; Yamazaki, S.; Nakamura, H.; Tane, K.; Miyoshi, T.; et al. Relationship between podoplanin-expressing cancer-associated fibroblasts and the immune microenvironment of early lung squamous cell carcinoma. Lung Cancer 2021, 153, 1–10. [Google Scholar] [CrossRef] [PubMed]
  114. Chen, C.; Guo, Q.; Liu, Y.; Hou, Q.; Liao, M.; Guo, Y.; Zang, Y.; Wang, F.; Liu, H.; Luan, X.; et al. Single-cell and spatial transcriptomics reveal POSTN(+) cancer-associated fibroblasts correlated with immune suppression and tumour progression in non-small cell lung cancer. Clin. Transl. Med. 2023, 13, e1515. [Google Scholar] [CrossRef]
  115. Tang, P.C.; Chung, J.Y.; Xue, V.W.; Xiao, J.; Meng, X.M.; Huang, X.R.; Zhou, S.; Chan, A.S.; Tsang, A.C.; Cheng, A.S.; et al. Smad3 Promotes Cancer-Associated Fibroblasts Generation via Macrophage-Myofibroblast Transition. Adv. Sci. 2022, 9, e2101235. [Google Scholar] [CrossRef] [PubMed]
  116. Tang, P.C.; Chan, M.K.; Chung, J.Y.; Chan, A.S.; Zhang, D.; Li, C.; Leung, K.T.; Ng, C.S.; Wu, Y.; To, K.F.; et al. Hematopoietic Transcription Factor RUNX1 is Essential for Promoting Macrophage-Myofibroblast Transition in Non-Small-Cell Lung Carcinoma. Adv. Sci. 2024, 11, e2302203. [Google Scholar] [CrossRef]
  117. Zhou, J.; Liu, H.; Jiang, S.; Wang, W. Role of tumor-associated neutrophils in lung cancer (Review). Oncol. Lett. 2023, 25, 2. [Google Scholar] [CrossRef]
  118. Piccard, H.; Muschel, R.J.; Opdenakker, G. On the dual roles and polarized phenotypes of neutrophils in tumor development and progression. Crit. Rev. Oncol. Hematol. 2012, 82, 296–309. [Google Scholar] [CrossRef]
  119. Sionov, R.V.; Fridlender, Z.G.; Granot, Z. The Multifaceted Roles Neutrophils Play in the Tumor Microenvironment. Cancer Microenviron. 2015, 8, 125–158. [Google Scholar] [CrossRef]
  120. Coffelt, S.B.; Wellenstein, M.D.; de Visser, K.E. Neutrophils in cancer: Neutral no more. Nat. Rev. Cancer 2016, 16, 431–446. [Google Scholar] [CrossRef]
  121. Fridlender, Z.G.; Albelda, S.M. Tumor-associated neutrophils: Friend or foe? Carcinogenesis 2012, 33, 949–955. [Google Scholar] [CrossRef] [PubMed]
  122. Tsai, Y.M.; Wu, K.L.; Liu, Y.W.; Chang, W.A.; Huang, Y.C.; Chang, C.Y.; Tsai, P.H.; Liao, S.H.; Hung, J.Y.; Hsu, Y.L. Cooperation Between Cancer and Fibroblasts in Vascular Mimicry and N2-Type Neutrophil Recruitment via Notch2-Jagged1 Interaction in Lung Cancer. Front. Oncol. 2021, 11, 696931. [Google Scholar] [CrossRef]
  123. Raman, D.; Baugher, P.J.; Thu, Y.M.; Richmond, A. Role of chemokines in tumor growth. Cancer Lett. 2007, 256, 137–165. [Google Scholar] [CrossRef]
  124. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef]
  125. Mucciolo, G.; Araos Henríquez, J.; Jihad, M.; Pinto Teles, S.; Manansala, J.S.; Li, W.; Ashworth, S.; Lloyd, E.G.; Cheng, P.S.W.; Luo, W.; et al. EGFR-activated myofibroblasts promote metastasis of pancreatic cancer. Cancer Cell 2024, 42, 101–118.e111. [Google Scholar] [CrossRef]
  126. Park, Y.; Chung, C. Immune Evasion of G-CSF and GM-CSF in Lung Cancer. Tuberc. Respir. Dis. 2024, 87, 22–30. [Google Scholar] [CrossRef]
  127. Guillerey, C.; Huntington, N.D.; Smyth, M.J. Targeting natural killer cells in cancer immunotherapy. Nat. Immunol. 2016, 17, 1025–1036. [Google Scholar] [CrossRef]
  128. Souza-Fonseca-Guimaraes, F.; Cursons, J.; Huntington, N.D. The Emergence of Natural Killer Cells as a Major Target in Cancer Immunotherapy. Trends Immunol. 2019, 40, 142–158. [Google Scholar] [CrossRef]
  129. Myers, J.A.; Miller, J.S. Exploring the NK cell platform for cancer immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18, 85–100. [Google Scholar] [CrossRef]
  130. Chiossone, L.; Dumas, P.Y.; Vienne, M.; Vivier, E. Natural killer cells and other innate lymphoid cells in cancer. Nat. Rev. Immunol. 2018, 18, 671–688. [Google Scholar] [CrossRef]
  131. Sivori, S.; Vacca, P.; Del Zotto, G.; Munari, E.; Mingari, M.C.; Moretta, L. Human NK cells: Surface receptors, inhibitory checkpoints, and translational applications. Cell Mol. Immunol. 2019, 16, 430–441. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, X.; Song, E. Turning foes to friends: Targeting cancer-associated fibroblasts. Nat. Rev. Drug Discov. 2019, 18, 99–115. [Google Scholar] [CrossRef] [PubMed]
  133. Turley, S.J.; Cremasco, V.; Astarita, J.L. Immunological hallmarks of stromal cells in the tumour microenvironment. Nat. Rev. Immunol. 2015, 15, 669–682. [Google Scholar] [CrossRef] [PubMed]
  134. Yang, N.; Lode, K.; Berzaghi, R.; Islam, A.; Martinez-Zubiaurre, I.; Hellevik, T. Irradiated Tumor Fibroblasts Avoid Immune Recognition and Retain Immunosuppressive Functions Over Natural Killer Cells. Front. Immunol. 2020, 11, 602530. [Google Scholar] [CrossRef]
  135. Flavell, R.A.; Sanjabi, S.; Wrzesinski, S.H.; Licona-Limón, P. The polarization of immune cells in the tumour environment by TGFbeta. Nat. Rev. Immunol. 2010, 10, 554–567. [Google Scholar] [CrossRef]
  136. Batlle, E.; Massagué, J. Transforming Growth Factor-β Signaling in Immunity and Cancer. Immunity 2019, 50, 924–940. [Google Scholar] [CrossRef]
  137. Trotta, R.; Dal Col, J.; Yu, J.; Ciarlariello, D.; Thomas, B.; Zhang, X.; Allard, J., 2nd; Wei, M.; Mao, H.; Byrd, J.C.; et al. TGF-beta utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and antibody-dependent cellular cytotoxicity in human NK cells. J. Immunol. 2008, 181, 3784–3792. [Google Scholar] [CrossRef]
  138. Lee, Y.S.; Radford, K.J. The role of dendritic cells in cancer. Int. Rev. Cell Mol. Biol. 2019, 348, 123–178. [Google Scholar] [CrossRef]
  139. Gardner, A.; Ruffell, B. Dendritic Cells and Cancer Immunity. Trends Immunol. 2016, 37, 855–865. [Google Scholar] [CrossRef]
  140. Berzaghi, R.; Tornaas, S.; Lode, K.; Hellevik, T.; Martinez-Zubiaurre, I. Ionizing Radiation Curtails Immunosuppressive Effects From Cancer-Associated Fibroblasts on Dendritic Cells. Front. Immunol. 2021, 12, 662594. [Google Scholar] [CrossRef]
  141. Kuo, P.L.; Hung, J.Y.; Huang, S.K.; Chou, S.H.; Cheng, D.E.; Jong, Y.J.; Hung, C.H.; Yang, C.J.; Tsai, Y.M.; Hsu, Y.L.; et al. Lung cancer-derived galectin-1 mediates dendritic cell anergy through inhibitor of DNA binding 3/IL-10 signaling pathway. J. Immunol. 2011, 186, 1521–1530. [Google Scholar] [CrossRef] [PubMed]
  142. Hsu, Y.L.; Hung, J.Y.; Chiang, S.Y.; Jian, S.F.; Wu, C.Y.; Lin, Y.S.; Tsai, Y.M.; Chou, S.H.; Tsai, M.J.; Kuo, P.L. Lung cancer-derived galectin-1 contributes to cancer associated fibroblast-mediated cancer progression and immune suppression through TDO2/kynurenine axis. Oncotarget 2016, 7, 27584–27598. [Google Scholar] [CrossRef] [PubMed]
  143. Ohshio, Y.; Teramoto, K.; Hanaoka, J.; Tezuka, N.; Itoh, Y.; Asai, T.; Daigo, Y.; Ogasawara, K. Cancer-associated fibroblast-targeted strategy enhances antitumor immune responses in dendritic cell-based vaccine. Cancer Sci. 2015, 106, 134–142. [Google Scholar] [CrossRef] [PubMed]
  144. Gabrilovich, D.I. Myeloid-Derived Suppressor Cells. Cancer Immunol. Res. 2017, 5, 3–8. [Google Scholar] [CrossRef]
  145. Adah, D.; Hussain, M.; Qin, L.; Qin, L.; Zhang, J.; Chen, X. Implications of MDSCs-targeting in lung cancer chemo-immunotherapeutics. Pharmacol. Res. 2016, 110, 25–34. [Google Scholar] [CrossRef]
  146. Xiang, H.; Ramil, C.P.; Hai, J.; Zhang, C.; Wang, H.; Watkins, A.A.; Afshar, R.; Georgiev, P.; Sze, M.A.; Song, X.S.; et al. Cancer-Associated Fibroblasts Promote Immunosuppression by Inducing ROS-Generating Monocytic MDSCs in Lung Squamous Cell Carcinoma. Cancer Immunol. Res. 2020, 8, 436–450. [Google Scholar] [CrossRef]
  147. Zhu, Y.; Zhang, L.; Zha, H.; Yang, F.; Hu, C.; Chen, L.; Guo, B.; Zhu, B. Stroma-derived Fibrinogen-like Protein 2 Activates Cancer-associated Fibroblasts to Promote Tumor Growth in Lung Cancer. Int. J. Biol. Sci. 2017, 13, 804–814. [Google Scholar] [CrossRef]
  148. Kumar, V.; Donthireddy, L.; Marvel, D.; Condamine, T.; Wang, F.; Lavilla-Alonso, S.; Hashimoto, A.; Vonteddu, P.; Behera, R.; Goins, M.A.; et al. Cancer-Associated Fibroblasts Neutralize the Anti-tumor Effect of CSF1 Receptor Blockade by Inducing PMN-MDSC Infiltration of Tumors. Cancer Cell 2017, 32, 654–668.e655. [Google Scholar] [CrossRef]
  149. Kumar, B.V.; Connors, T.J.; Farber, D.L. Human T Cell Development, Localization, and Function throughout Life. Immunity 2018, 48, 202–213. [Google Scholar] [CrossRef]
  150. O’Connor, R.A.; Chauhan, V.; Mathieson, L.; Titmarsh, H.; Koppensteiner, L.; Young, I.; Tagliavini, G.; Dorward, D.A.; Prost, S.; Dhaliwal, K.; et al. T cells drive negative feedback mechanisms in cancer associated fibroblasts, promoting expression of co-inhibitory ligands, CD73 and IL-27 in non-small cell lung cancer. Oncoimmunology 2021, 10, 1940675. [Google Scholar] [CrossRef]
  151. Tanaka, A.; Sakaguchi, S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017, 27, 109–118. [Google Scholar] [CrossRef] [PubMed]
  152. Kinoshita, T.; Ishii, G.; Hiraoka, N.; Hirayama, S.; Yamauchi, C.; Aokage, K.; Hishida, T.; Yoshida, J.; Nagai, K.; Ochiai, A. Forkhead box P3 regulatory T cells coexisting with cancer associated fibroblasts are correlated with a poor outcome in lung adenocarcinoma. Cancer Sci. 2013, 104, 409–415. [Google Scholar] [CrossRef]
  153. O’Connor, R.A.; Martinez, B.R.; Koppensteiner, L.; Mathieson, L.; Akram, A.R. Cancer-associated fibroblasts drive CXCL13 production in activated T cells via TGF-beta. Front. Immunol. 2023, 14, 1221532. [Google Scholar] [CrossRef]
  154. Zhu, J. T Helper Cell Differentiation, Heterogeneity, and Plasticity. Cold Spring Harb. Perspect. Biol. 2018, 10, a030338. [Google Scholar] [CrossRef]
  155. Zhang, Y.; Zhang, Y.; Gu, W.; He, L.; Sun, B. Th1/Th2 cell’s function in immune system. Adv. Exp. Med. Biol. 2014, 841, 45–65. [Google Scholar] [CrossRef] [PubMed]
  156. Koeck, S.; Amann, A.; Kern, J.; Zwierzina, M.; Lorenz, E.; Sopper, S.; Zwierzina, H.; Mildner, F.; Sykora, M.; Sprung, S.; et al. Whole stromal fibroblast signature is linked to specific chemokine and immune infiltration patterns and to improved survival in NSCLC. Oncoimmunology 2023, 12, 2274130. [Google Scholar] [CrossRef] [PubMed]
  157. Miyai, Y.; Sugiyama, D.; Hase, T.; Asai, N.; Taki, T.; Nishida, K.; Fukui, T.; Chen-Yoshikawa, T.F.; Kobayashi, H.; Mii, S.; et al. Meflin-positive cancer-associated fibroblasts enhance tumor response to immune checkpoint blockade. Life Sci. Alliance 2022, 5, e202101230. [Google Scholar] [CrossRef]
  158. Freeman, P.; Mielgo, A. Cancer-Associated Fibroblast Mediated Inhibition of CD8+ Cytotoxic T Cell Accumulation in Tumours: Mechanisms and Therapeutic Opportunities. Cancers 2020, 12, 2687. [Google Scholar] [CrossRef]
  159. Yang, H.; Sun, B.; Fan, L.; Ma, W.; Xu, K.; Hall, S.R.R.; Wang, Z.; Schmid, R.A.; Peng, R.W.; Marti, T.M.; et al. Multi-scale integrative analyses identify THBS2(+) cancer-associated fibroblasts as a key orchestrator promoting aggressiveness in early-stage lung adenocarcinoma. Theranostics 2022, 12, 3104–3130. [Google Scholar] [CrossRef]
  160. Sakai, T.; Aokage, K.; Neri, S.; Nakamura, H.; Nomura, S.; Tane, K.; Miyoshi, T.; Sugano, M.; Kojima, M.; Fujii, S.; et al. Link between tumor-promoting fibrous microenvironment and an immunosuppressive microenvironment in stage I lung adenocarcinoma. Lung Cancer 2018, 126, 64–71. [Google Scholar] [CrossRef]
  161. Grout, J.A.; Sirven, P.; Leader, A.M.; Maskey, S.; Hector, E.; Puisieux, I.; Steffan, F.; Cheng, E.; Tung, N.; Maurin, M.; et al. Spatial Positioning and Matrix Programs of Cancer-Associated Fibroblasts Promote T-cell Exclusion in Human Lung Tumors. Cancer Discov. 2022, 12, 2606–2625. [Google Scholar] [CrossRef] [PubMed]
  162. Lee, H.; Hwang, M.; Jang, S.; Um, S.W. Immune Regulatory Function of Cancer- Associated Fibroblasts in Non-small Cell Lung Cancer. Tuberc. Respir. Dis. 2023, 86, 304–318. [Google Scholar] [CrossRef]
  163. Mao, G.; Liu, J. Research on the mechanism of exosomes from different sources influencing the progression of lung cancer. Environ. Toxicol. 2024, 39, 4231–4248. [Google Scholar] [CrossRef] [PubMed]
  164. Zheng, X.; Jiang, K.; Xiao, W.; Zeng, D.; Peng, W.; Bai, J.; Chen, X.; Li, P.; Zhang, L.; Zheng, X.; et al. CD8(+) T cell/cancer-associated fibroblast ratio stratifies prognostic and predictive responses to immunotherapy across multiple cancer types. Front. Immunol. 2022, 13, 974265. [Google Scholar] [CrossRef]
  165. Koppensteiner, L.; Mathieson, L.; Neilson, L.; O’Connor, R.A.; Akram, A.R. IFNγ and TNFα drive an inflammatory secretion profile in cancer-associated fibroblasts from human non-small cell lung cancer. FEBS Lett. 2025, 599, 713–723. [Google Scholar] [CrossRef]
  166. Thommen, D.S.; Schumacher, T.N. T Cell Dysfunction in Cancer. Cancer Cell 2018, 33, 547–562. [Google Scholar] [CrossRef]
  167. Thommen, D.S.; Schreiner, J.; Müller, P.; Herzig, P.; Roller, A.; Belousov, A.; Umana, P.; Pisa, P.; Klein, C.; Bacac, M.; et al. Progression of Lung Cancer Is Associated with Increased Dysfunction of T Cells Defined by Coexpression of Multiple Inhibitory Receptors. Cancer Immunol. Res. 2015, 3, 1344–1355. [Google Scholar] [CrossRef]
  168. Sun, C.; Mezzadra, R.; Schumacher, T.N. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018, 48, 434–452. [Google Scholar] [CrossRef]
  169. Lakins, M.A.; Ghorani, E.; Munir, H.; Martins, C.P.; Shields, J.D. Cancer-associated fibroblasts induce antigen-specific deletion of CD8 (+) T Cells to protect tumour cells. Nat. Commun. 2018, 9, 948. [Google Scholar] [CrossRef]
  170. Nazareth, M.R.; Broderick, L.; Simpson-Abelson, M.R.; Kelleher, R.J., Jr.; Yokota, S.J.; Bankert, R.B. Characterization of human lung tumor-associated fibroblasts and their ability to modulate the activation of tumor-associated T cells. J. Immunol. 2007, 178, 5552–5562. [Google Scholar] [CrossRef]
  171. Zhao, Y.; Liu, Y.; Jia, Y.; Wang, X.; He, J.; Zhen, S.; Wang, J.; Liu, L. Fibroblast activation protein in the tumor microenvironment predicts outcomes of PD-1 blockade therapy in advanced non-small cell lung cancer. J. Cancer Res. Clin. Oncol. 2023, 149, 3469–3483. [Google Scholar] [CrossRef] [PubMed]
  172. Bu, Y.; Zhang, L.; Ma, X.; Wang, R.; Zhang, X.; Li, J. Systematic Analysis of the Oncogenic Role of WDR62 in Human Tumors. Dis. Markers 2021, 2021, 9940274. [Google Scholar] [CrossRef] [PubMed]
  173. Inoue, C.; Miki, Y.; Saito, R.; Hata, S.; Abe, J.; Sato, I.; Okada, Y.; Sasano, H. PD-L1 Induction by Cancer-Associated Fibroblast-Derived Factors in Lung Adenocarcinoma Cells. Cancers 2019, 11, 1257. [Google Scholar] [CrossRef]
  174. Eble, J.A.; Niland, S. The extracellular matrix in tumor progression and metastasis. Clin. Exp. Metastasis 2019, 36, 171–198. [Google Scholar] [CrossRef]
  175. Ozbek, S.; Balasubramanian, P.G.; Chiquet-Ehrismann, R.; Tucker, R.P.; Adams, J.C. The evolution of extracellular matrix. Mol. Biol. Cell 2010, 21, 4300–4305. [Google Scholar] [CrossRef]
  176. Miles, F.L.; Sikes, R.A. Insidious changes in stromal matrix fuel cancer progression. Mol. Cancer Res. 2014, 12, 297–312. [Google Scholar] [CrossRef] [PubMed]
  177. Chakravarthy, A.; Khan, L.; Bensler, N.P.; Bose, P.; De Carvalho, D.D. TGF-β-associated extracellular matrix genes link cancer-associated fibroblasts to immune evasion and immunotherapy failure. Nat. Commun. 2018, 9, 4692. [Google Scholar] [CrossRef]
  178. Sorokin, L. The impact of the extracellular matrix on inflammation. Nat. Rev. Immunol. 2010, 10, 712–723. [Google Scholar] [CrossRef]
  179. Joyce, J.A.; Fearon, D.T. T cell exclusion, immune privilege, and the tumor microenvironment. Science 2015, 348, 74–80. [Google Scholar] [CrossRef]
  180. Salmon, H.; Franciszkiewicz, K.; Damotte, D.; Dieu-Nosjean, M.C.; Validire, P.; Trautmann, A.; Mami-Chouaib, F.; Donnadieu, E. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J. Clin. Investig. 2012, 122, 899–910. [Google Scholar] [CrossRef]
  181. Stahl, M.; Schupp, J.; Jäger, B.; Schmid, M.; Zissel, G.; Müller-Quernheim, J.; Prasse, A. Lung collagens perpetuate pulmonary fibrosis via CD204 and M2 macrophage activation. PLoS ONE 2013, 8, e81382. [Google Scholar] [CrossRef]
  182. Zhang, J.; Lu, S.; Lu, T.; Han, D.; Zhang, K.; Gan, L.; Wu, X.; Li, Y.; Zhao, X.; Li, Z.; et al. Single-cell analysis reveals the COL11A1(+) fibroblasts are cancer-specific fibroblasts that promote tumor progression. Front. Pharmacol. 2023, 14, 1121586. [Google Scholar] [CrossRef]
  183. Berzaghi, R.; Gundersen, K.; Dille Pedersen, B.; Utne, A.; Yang, N.; Hellevik, T.; Martinez-Zubiaurre, I. Immunological signatures from irradiated cancer-associated fibroblasts. Front. Immunol. 2024, 15, 1433237. [Google Scholar] [CrossRef]
  184. Weichselbaum, R.R.; Liang, H.; Deng, L.; Fu, Y.X. Radiotherapy and immunotherapy: A beneficial liaison? Nat. Rev. Clin. Oncol. 2017, 14, 365–379. [Google Scholar] [CrossRef] [PubMed]
  185. Gameiro, S.R.; Ardiani, A.; Kwilas, A.; Hodge, J.W. Radiation-induced survival responses promote immunogenic modulation to enhance immunotherapy in combinatorial regimens. Oncoimmunology 2014, 3, e28643. [Google Scholar] [CrossRef]
  186. Meng, J.; Li, Y.; Wan, C.; Sun, Y.; Dai, X.; Huang, J.; Hu, Y.; Gao, Y.; Wu, B.; Zhang, Z.; et al. Targeting senescence-like fibroblasts radiosensitizes non-small cell lung cancer and reduces radiation-induced pulmonary fibrosis. JCI Insight 2021, 6, e146334. [Google Scholar] [CrossRef] [PubMed]
  187. Gorchs, L.; Hellevik, T.; Bruun, J.A.; Camilio, K.A.; Al-Saad, S.; Stuge, T.B.; Martinez-Zubiaurre, I. Cancer-associated fibroblasts from lung tumors maintain their immunosuppressive abilities after high-dose irradiation. Front. Oncol. 2015, 5, 87. [Google Scholar] [CrossRef]
  188. Akanda, M.R.; Ahn, E.J.; Kim, Y.J.; Salam, S.M.A.; Noh, M.G.; Kim, S.S.; Jung, T.Y.; Kim, I.Y.; Kim, C.H.; Lee, K.H.; et al. Different Expression and Clinical Implications of Cancer-Associated Fibroblast (CAF) Markers in Brain Metastases. J. Cancer 2023, 14, 464–479. [Google Scholar] [CrossRef]
  189. Hellevik, T.; Pettersen, I.; Berg, V.; Winberg, J.O.; Moe, B.T.; Bartnes, K.; Paulssen, R.H.; Busund, L.T.; Bremnes, R.; Chalmers, A.; et al. Cancer-associated fibroblasts from human NSCLC survive ablative doses of radiation but their invasive capacity is reduced. Radiat. Oncol. 2012, 7, 59. [Google Scholar] [CrossRef]
  190. Hodkinson, P.S.; Elliott, T.; Wong, W.S.; Rintoul, R.C.; Mackinnon, A.C.; Haslett, C.; Sethi, T. ECM overrides DNA damage-induced cell cycle arrest and apoptosis in small-cell lung cancer cells through beta1 integrin-dependent activation of PI3-kinase. Cell Death Differ. 2006, 13, 1776–1788. [Google Scholar] [CrossRef]
  191. Carracedo, S.; Lu, N.; Popova, S.N.; Jonsson, R.; Eckes, B.; Gullberg, D. The fibroblast integrin alpha11beta1 is induced in a mechanosensitive manner involving activin A and regulates myofibroblast differentiation. J. Biol. Chem. 2010, 285, 10434–10445. [Google Scholar] [CrossRef]
  192. Puthawala, K.; Hadjiangelis, N.; Jacoby, S.C.; Bayongan, E.; Zhao, Z.; Yang, Z.; Devitt, M.L.; Horan, G.S.; Weinreb, P.H.; Lukashev, M.E.; et al. Inhibition of integrin alpha(v)beta6, an activator of latent transforming growth factor-beta, prevents radiation-induced lung fibrosis. Am. J. Respir. Crit. Care Med. 2008, 177, 82–90. [Google Scholar] [CrossRef] [PubMed]
  193. Barcellos-Hoff, M.H.; Derynck, R.; Tsang, M.L.; Weatherbee, J.A. Transforming growth factor-beta activation in irradiated murine mammary gland. J. Clin. Investig. 1994, 93, 892–899. [Google Scholar] [CrossRef] [PubMed]
  194. Neuzillet, C.; Tijeras-Raballand, A.; Cohen, R.; Cros, J.; Faivre, S.; Raymond, E.; de Gramont, A. Targeting the TGFβ pathway for cancer therapy. Pharmacol. Ther. 2015, 147, 22–31. [Google Scholar] [CrossRef]
  195. Yao, F.; Shi, W.; Fang, F.; Lv, M.Y.; Xu, M.; Wu, S.Y.; Huang, C.L. Exosomal miR-196a-5p enhances radioresistance in lung cancer cells by downregulating NFKBIA. Kaohsiung J. Med. Sci. 2023, 39, 554–564. [Google Scholar] [CrossRef]
  196. Wang, Y.; Gan, G.; Wang, B.; Wu, J.; Cao, Y.; Zhu, D.; Xu, Y.; Wang, X.; Han, H.; Li, X.; et al. Cancer-associated Fibroblasts Promote Irradiated Cancer Cell Recovery Through Autophagy. EBioMedicine 2017, 17, 45–56. [Google Scholar] [CrossRef]
  197. Berzaghi, R.; Ahktar, M.A.; Islam, A.; Pedersen, B.D.; Hellevik, T.; Martinez-Zubiaurre, I. Fibroblast-Mediated Immunoregulation of Macrophage Function Is Maintained after Irradiation. Cancers 2019, 11, 689. [Google Scholar] [CrossRef] [PubMed]
  198. Domogauer, J.D.; de Toledo, S.M.; Howell, R.W.; Azzam, E.I. Acquired radioresistance in cancer associated fibroblasts is concomitant with enhanced antioxidant potential and DNA repair capacity. Cell Commun. Signal. 2021, 19, 30. [Google Scholar] [CrossRef]
  199. Li, H.; Zhang, Q.; Wu, Q.; Cui, Y.; Zhu, H.; Fang, M.; Zhou, X.; Sun, Z.; Yu, J. Interleukin-22 secreted by cancer-associated fibroblasts regulates the proliferation and metastasis of lung cancer cells via the PI3K-Akt-mTOR signaling pathway. Am. J. Transl. Res. 2019, 11, 4077–4088. [Google Scholar]
  200. Ceuppens, H.; Pombo Antunes, A.R.; Navarro, L.; Ertveldt, T.; Berdal, M.; Nagachinta, S.; De Ridder, K.; Lahoutte, T.; Keyaerts, M.; Devoogdt, N.; et al. Efficient α and β(-) radionuclide therapy targeting fibroblast activation protein-α in an aggressive preclinical mouse tumour model. Eur. J. Nucl. Med. Mol. Imaging 2025, 52, 444–457. [Google Scholar] [CrossRef]
  201. Xie, Y.; Ma, J.; Tang, W.; Zhang, Y.; Zhang, C.; Chen, Y. Efficacy and Safety Evaluation of 177Lu-FAP-2286 in the Treatment of Advanced Lung Cancer. Clin. Nucl. Med. 2024, 49, 830–837. [Google Scholar] [CrossRef] [PubMed]
  202. Wang, R.; Jakobsson, V.; Wang, J.; Zhao, T.; Peng, X.; Li, B.; Xue, J.; Liang, N.; Zhu, Z.; Chen, X.; et al. Dual targeting PET tracer [(68)Ga]Ga-FAPI-RGD in patients with lung neoplasms: A pilot exploratory study. Theranostics 2023, 13, 2979–2992. [Google Scholar] [CrossRef] [PubMed]
  203. Lee, I.K.; Noguera-Ortega, E.; Xiao, Z.; Todd, L.; Scholler, J.; Song, D.; Liousia, M.; Lohith, K.; Xu, K.; Edwards, K.J.; et al. Monitoring Therapeutic Response to Anti-FAP CAR T Cells Using [18F]AlF-FAPI-74. Clin. Cancer Res. 2022, 28, 5330–5342. [Google Scholar] [CrossRef]
  204. Huang, J.; Zhang, X.; Liu, Q.; Gong, F.; Huang, Y.; Huang, S.; Fu, L.; Tang, G. (68)Ga/(177)Lu-Labeled Theranostic Pair for Targeting Fibroblast Activation Protein with Improved Tumor Uptake and Retention. J. Med. Chem. 2024, 67, 17785–17795. [Google Scholar] [CrossRef] [PubMed]
  205. Giesel, F.L.; Kratochwil, C.; Schlittenhardt, J.; Dendl, K.; Eiber, M.; Staudinger, F.; Kessler, L.; Fendler, W.P.; Lindner, T.; Koerber, S.A.; et al. Head-to-head intra-individual comparison of biodistribution and tumor uptake of (68)Ga-FAPI and (18)F-FDG PET/CT in cancer patients. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 4377–4385. [Google Scholar] [CrossRef] [PubMed]
  206. Gong, X.; Li, J.; Xu, X.; Wu, Y.; Lei, Y.; Liu, H.; Qian, X.; Li, Y.; Zhang, Z. Microvesicle-inspired oxygen-delivering nanosystem potentiates radiotherapy-mediated modulation of tumor stroma and antitumor immunity. Biomaterials 2022, 290, 121855. [Google Scholar] [CrossRef]
  207. Kolset, S.O.; Pejler, G. Serglycin: A structural and functional chameleon with wide impact on immune cells. J. Immunol. 2011, 187, 4927–4933. [Google Scholar] [CrossRef]
  208. Beer, T.W.; Ng, L.B.; Murray, K. Mast cells have prognostic value in Merkel cell carcinoma. Am. J. Dermatopathol. 2008, 30, 27–30. [Google Scholar] [CrossRef]
  209. Medina, V.; Cricco, G.; Nuñez, M.; Martín, G.; Mohamad, N.; Correa-Fiz, F.; Sanchez-Jimenez, F.; Bergoc, R.; Rivera, E.S. Histamine-mediated signaling processes in human malignant mammary cells. Cancer Biol. Ther. 2006, 5, 1462–1471. [Google Scholar] [CrossRef]
  210. Carmeliet, P.; Jain, R.K. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011, 473, 298–307. [Google Scholar] [CrossRef]
  211. Ribatti, D.; Crivellato, E. Mast cells, angiogenesis and cancer. Adv. Exp. Med. Biol. 2011, 716, 270–288. [Google Scholar] [CrossRef] [PubMed]
  212. Maltby, S.; Khazaie, K.; McNagny, K.M. Mast cells in tumor growth: Angiogenesis, tissue remodelling and immune-modulation. Biochim. Biophys. Acta 2009, 1796, 19–26. [Google Scholar] [CrossRef] [PubMed]
  213. Alashkar Alhamwe, B.; Ponath, V.; Alhamdan, F.; Dörsam, B.; Landwehr, C.; Linder, M.; Pauck, K.; Miethe, S.; Garn, H.; Finkernagel, F.; et al. BAG6 restricts pancreatic cancer progression by suppressing the release of IL33-presenting extracellular vesicles and the activation of mast cells. Cell Mol. Immunol. 2024, 21, 918–931. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The heterogeneity of CAF origin and subtypes. Different colors represent different subtypes of CAFs.
Figure 1. The heterogeneity of CAF origin and subtypes. Different colors represent different subtypes of CAFs.
Ijms 26 03234 g001
Figure 2. The relationships between CAF and tumor immune microenvironment in lung cancer.
Figure 2. The relationships between CAF and tumor immune microenvironment in lung cancer.
Ijms 26 03234 g002
Table 1. Markers of CAFs in lung cancer.
Table 1. Markers of CAFs in lung cancer.
MarkerFunctionSpecific Role in Lung CancerClinical SignificanceRefs
α-SMACell shrinkage, matrix remodelingA core marker of CAF activation that promotes tumor invasion and fibrosis.High expression is associated with poor prognosis[63]
FAPExtracellular matrix degradation, immunomodulationPromote tumor growth and metastasis and inhibit anti-tumor immune response.Associated with chemotherapy resistance and shortened survival[45]
PDGFRβCell proliferation, migrationHigh expression of CAF in lung cancer drives mesenchymal remodeling and angiogenesis.Potential markers for targeted therapies[64]
FSP1 (S100A4)Cell migration, metastasisSpecifically labeling lung cancer CAF subpopulations, metabolic plasticity can drive tumor invasion and metastasis in non-small cell lung cancer cells.Targeted metabolic reprogramming provides therapeutic window for enhanced glycolysis inhibition[65]
Tenascin-CCell migration, metastasisSignificantly expressed in the microenvironment of lung cancer, promoting cancer cell invasion and metastasis.High expression predicts advanced staging[66]
PeriostinEMTPeriostin expression was positively correlated with the EMT markers Snail and Twist and with lung cancer stage, in which mature epithelial cells undergo phenotypic morphological changes and become invasive, motile cells.Higher periostin levels correlate with poorer overall survival[67]
VimentinCell migration and EMTHigh expression in CAFs enhances lung cancer cell metastasis by inducing EMT signaling; correlates with immunosuppression in the tumor microenvironment.Associated with late staging and increased risk of metastasis[45]
COL1A1Malignant progression and metastasisHigher proportion of COL1A1-positive CAFs associated with shorter patient survival.Predicting TME-dependent survival expectancy and treatment benefits[68]
COL5A1Regulates collagen fiber assembly and matrix structureCOL5A1 promotes metastasis by regulating protease activity and migration-associated proteins, and inhibition of its expression reduces invasiveness and enhances chemosensitivity.Potential therapeutic targets[69]
P-glycoproteinReduces drug retention and increases drug outflowInduced by the AKT/Sox2 signaling pathway, P-GP expression induced chemoresistance in NSCLC cells.Associated with chemotherapy resistance[70]
Adipocyte enhancer-binding protein 1 (AEBP1)Promotes proliferation, migration, invasion and transferAEBP1 is not only oncogenic in epithelial tumor cells, but may also be oncogenic in stromal cells.Overexpression is associated with the prediction of poor prognosis in patients with pulmonary SCC[71]
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

Shi, Z.; Hu, C.; Li, Q.; Sun, C. Cancer-Associated Fibroblasts as the “Architect” of the Lung Cancer Immune Microenvironment: Multidimensional Roles and Synergistic Regulation with Radiotherapy. Int. J. Mol. Sci. 2025, 26, 3234. https://doi.org/10.3390/ijms26073234

AMA Style

Shi Z, Hu C, Li Q, Sun C. Cancer-Associated Fibroblasts as the “Architect” of the Lung Cancer Immune Microenvironment: Multidimensional Roles and Synergistic Regulation with Radiotherapy. International Journal of Molecular Sciences. 2025; 26(7):3234. https://doi.org/10.3390/ijms26073234

Chicago/Turabian Style

Shi, Zheng, Cuilan Hu, Qiang Li, and Chao Sun. 2025. "Cancer-Associated Fibroblasts as the “Architect” of the Lung Cancer Immune Microenvironment: Multidimensional Roles and Synergistic Regulation with Radiotherapy" International Journal of Molecular Sciences 26, no. 7: 3234. https://doi.org/10.3390/ijms26073234

APA Style

Shi, Z., Hu, C., Li, Q., & Sun, C. (2025). Cancer-Associated Fibroblasts as the “Architect” of the Lung Cancer Immune Microenvironment: Multidimensional Roles and Synergistic Regulation with Radiotherapy. International Journal of Molecular Sciences, 26(7), 3234. https://doi.org/10.3390/ijms26073234

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

Article Metrics

Back to TopTop