Home Functions of CAFs in microenvironment of non-small cell lung cancer: based on updated hallmarks of cancer
Article Open Access

Functions of CAFs in microenvironment of non-small cell lung cancer: based on updated hallmarks of cancer

  • Xiaoyan Feng , Binghan Zhu , Yali Peng , Kaiyuan Zhang , Yangchun Wang , Guichun Huang EMAIL logo and Yan Li EMAIL logo
Published/Copyright: September 2, 2024

Abstract

Non-small cell lung cancer (NSCLC) is the most common subtype of lung cancer, which ranks as the first malignant tumor in mortality. The occurrence and development of NSCLC are closely related to the tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs) in the tumor microenvironment are considered to be critical regulators of the occurrence and development of NSCLC, which have essential effects on multiple biological characteristics of NSCLC. The hallmarks of cancer biology have been updated recently, however, there are no reviews revisiting the function of CAFs in tumor microenvironment. This article reviews the origin, markers, and classification of CAFs, their impacts on the characteristics of NSCLC, and potential therapeutic targets of CAFs to help develop individualized treatment plans for NSCLC.

Introduction

Lung cancer currently ranks as the first malignant tumor in mortality rate in the world [1]. The most essential pathological type of lung cancer is non-small cell lung cancer (NSCLC), which is further divided into lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), and large cell carcinoma, among which adenocarcinoma and squamous cell carcinoma are the most common subtypes [2]. Clinically, only a small proportion of NSCLC patients are diagnosed with localized disease and can be treated with surgical resection. More than 50 % of patients are already locally advanced or have distant metastases at the time of diagnosis [3]. Chemotherapy, radiotherapy, targeted therapy, anti-angiogenic therapy, and immunotherapy are the main treatment methods for this part of the patients 4], [5], [6. Patients with metastatic NSCLC who received only a conventional cytotoxic chemotherapy regimen had a 5-year relative survival rate of about 6 %, while patients with metastatic NSCLC who received targeted therapy or immunotherapy survived longer, with 5-year survival rates ranging from 15 to 50 % [7]. The choice of NSCLC treatment depends on tumor biology, such as gene mutations (driver mutation status such as EGFR, ALK, ROS1, BRAF, MET, KRAS), tumor immune evasion, and angiogenesis status [6, 8, 9]. The biological characteristics of NSCLC play critical roles in the occurrence and development of NSCLC [10, 11]. With the detailed study of the tumor cells and tumor microenvironment (TME) in NSCLC, the hallmarks of NSCLC have significantly changed [12]. TME and tumor immune microenvironment (TIME) involve a series parade of blood cells to be identified further [13]. Cancer-associated fibroblasts (CAFs) are one of the main components of the NSCLC microenvironment [14], which can modulate the tumor’s biological characteristics by secreting various inflammatory cytokines and extracellular matrix. Cancer biology changes daily, and several cancer hallmarks have emerged recently. Although a series of reviews have discussed the functions of CAFs [15], the relationship between CAFs and new biological characteristics is unclear. As the updated hallmarks of cancer biology [12], it is necessary to revisit the functions of CAFs in the tumor microenvironment of NSCLC. This article aims to review the origin, markers, and classification of CAFs, their impacts on the characteristics of NSCLC, and potential therapeutic targets of CAFs to help develop individualized treatment plans for NSCLC.

Characteristics of CAFs in NSCLC

Origin of CAFs

CAFs in NSCLC can be transformed from various stromal cells [16, 17]. Lung resident fibroblasts are one of the essential sources of CAFs. Quiescent lung resident fibroblasts are activated in response to stimuli such as tissue trauma, inflammation, and fibrosis, which promote tissue repair and regeneration [18]. NSCLC tumor cells can activate fibroblasts for epigenetic reprogramming to obtain a tumor-promoting phenotype [19]. Another important source of CAFs is mesenchymal stem cells, which can differentiate into CAFs after being stimulated by tumor signaling [20]. The high expression of α-1 antitrypsin (A1AT) in NSCLC promotes the Epithelial-to-Mesenchymal Transition (EMT) in epithelial cells and forms a pro-tumor CAFs phenotype [21]. Adipose mesenchymal stem cells (MSCs) can be induced by tumor cells, secrete high levels of interleukin-6 (IL-6) family cytokines, adipokines, and adhesion molecules that activate the signal transducer and activator of transcription 3 (STAT3) signaling pathway, increase the expression of matrix metalloproteinases in various cancer types (like bladder cancer), and induce EMT, exhibiting a pro-tumor CAFs phenotype [22]. Bone marrow mesenchymal stem cells can be recruited to tissue injury or inflammation sites through endocrine signals to exert their tissue repair function. In contrast, the tissue regeneration function of MSCs may be faulty in the tumor microenvironment of NSCLC and differentiate into CAFs [23]. Recent studies have found that macrophages and endothelial cells are also a source of CAFs in the NSCLC microenvironment [24, 25]. Although distributed in different locations in the tumor microenvironment, smooth muscle cells and pericytes exhibit similar phenotypes and functions and also might be another origin of CAFs through transdifferentiation [14].

Biomarkers of CAFs

Common CAFs markers include fibroblast activation protein (FAP), alpha-smooth muscle actin (α-SMA), platelet‐derived growth factor receptor (PDGFR), PDPN (podoplanin), vimentin, etc. FAP is a type II membrane protein that has been implicated in tissue repair, fibrosis, and degradation of the extracellular matrix in fibroblasts, and the expression of FAP on fibroblasts in NSCLC has been linked to its prognosis and survival [26]. α-SMA, a member of the actin family, induces myofibroblast contraction and promotes granulation tissue contraction through microfilament bundle and stress fiber regulation. Increased expression of α-SMA is associated with poor prognosis in tumors of NSCLC patients [27]. PDGFR is a universal marker for fibroblasts, and multiple studies have observed that PDGFR expression in CAFs is closely related to the prognosis of NSCLC. PDPN is a glycoprotein, also known as a lymphangitic marker, and multiple studies have found that PDPN+ CAFs have tumor-promoting functions and are associated with a poorer prognosis in patients with LUSC [28]. Vimentin is an intermediate filament protein (type III) that plays a vital role in organelle structure, cell migration, and adhesion in mesenchymal cells. Fibroblasts are characterized by their mesenchymal phenotype, and vimentin is highly expressed in NSCLC interstitial CAFs and is involved in CAFs motility to promote tumor cell invasion and metastasis [29]. In addition, the membrane-bound extracellular enzyme γ-glutamyltransferase (GGT) hydrolyzes γ-glutamyl bonds, and high expression of GGT in CAFs predicts poor survival in patients with LUAD [30]. CAFs generally have a pro-tumor phenotype, but FAP-positive CAFs have also been shown to be associated with a better prognosis in NSCLC patients with high levels of CD3 and CD8 double positive T cells, suggesting a tumor-suppressive effect of CAFs. Still, in most studies, CAFs have played a tumor-promoting role [31].

Subtypes of CAFs in NSCLC

The expression of CAFs-related markers has been reported to help analyze CAFs subsets [32]. However, their low specificity and heterogeneity pose significant challenges to typing CAFs. CAF-related markers are expressed in a variety of tumors. FAP is highly expressed in CAFs of malignant tumors such as NSCLC, pancreatic cancer, and breast cancer. It is also closely related to the progression of chronic inflammatory diseases [31], especially playing a pivotal role in the occurrence and development of interstitial lung diseases [33]. The myofibroblast phenotype with high expression of α-SMA can secrete ECM and cause protein deposition, which plays an important role in the occurrence and development of lung cancer and idiopathic pulmonary fibrosis [27]. Hu et al. established a gene pool of CAFs, including NSCLC-related CAFs. They determined the heterogeneity of CAFs by analyzing the differences in the expression levels of CAFs genes and then identified three functional subtypes: (1) hepatocyte growth factor (HGF) and fibroblast growth factor 7 (FGF-7) with a strong protective effect on tumors, (2) moderately protective tumors and highly expressed FGF-7 isoforms, and (3) functional subtypes that provide minimal protection [34].

Other new techniques, such as single-cell imaging mass cytometry (IMC), could also be applied for categorying CAFs. This functional classification of CAFs is related to the patient’s clinical response to targeted therapy and the tumor immune microenvironment and thus can provide a pathway for guiding personalized treatment [35].

Effects of CAFs on hallmarks of NSCLC

Promote tumor cell growth and maintain cell stemness

Tumor cells can activate CAFs through abnormal signaling and interact with CAFs to secrete growth factors and cytokines that have pro-tumor effects, silence tumor growth inhibitory signals, and maintain stem cell properties. It has been demonstrated that the hypoxic microenvironment of NSCLC can enhance the expression of hypoxia-inducible factor-1α (HIF-1α) in fibroblasts and induce the conversion of normal fibroblasts to CAFs. At the same time, the expression of HIF-1α in fibroblasts can activate the NF-κB signaling pathway and enhance C-C chemokine ligand 5 (CCL5) secretion, thereby promoting tumor growth [36]. Another study has shown that IL-22 secreted by CAFs reduces apoptosis and significantly promotes the proliferation of NSCLC cells by activating the phosphatidylinositol 3-kinase (PI3K)-Akt-mTOR signaling pathway [37]. The down-regulation of MiR-101-3p in CAFs can activate CAFs and promote tumor growth [38]. On the other hand, CAFs could silence inhibition signals of tumor cell growth. Disruption of the retinoblastoma protein (pRb) pathway releases the E2F transcription factor, allowing cells to enter the proliferative cycle. Bouchard et al. compared the phenotypic differences of CAFs (invasive margins and tumor centers) at different sites of LUAD and found that different CAFs with abnormal glucose metabolism can have different activation signaling pathways, including cyclin-dependent kinase 4 (CDK4) and phosphorylated retinoblastoma protein axis (CDK4-pRb axis), which indirectly regulate the invasion characteristics of cancer cells [39]. CAFs also could maintain tumor cell stemness. Several studies have shown that CAFs in NSCLC can regulate the expression of Nanog in tumor cells through paracrine signaling, which in turn regulates the plasticity of cancer stem cells (CSCs) [31]. The mechanism by which CAFs contribute to the occurrence and progression of NSCLC is very complex, and although many studies have been conducted, there are still many gaps in the understanding of this field.

Promote angiogenesis

Tumor angiogenesis is one of the important features of most tumors, and CAFs promote the formation of NSCLC angiogenesis through a variety of mechanisms, thereby promoting tumor progression. CAFs can promote the growth and infiltration of NSCLC through their pro-angiogenic effects. It has been found that CAFs are one of the essential sources of vascular endothelial growth factor A (VEGFA) in the NSCLC microenvironment, and the low expression of miR-101-3p in NSCLC-related CAFs can promote VEGFA secretion and thus angiogenesis [38]. NSCLC cell-derived exosomal mi-210 increases the expression of pro-angiogenic factors matrix metalloproteinase-9 (MMP9), fibroblast growth factor 2 (FGF2), and VEGFA by regulating the JAK2/STAT3 signaling pathway and promoting angiogenesis [40]. In LUAD models, CAFs overexpress transforming growth factor-β (TGFβ), MMP7, FGF9, and FGF2, synthesize more collagen, and promote tumor angiogenesis [41]. In some NSCLCs, the interaction between tumor cells and CAFs in the tumor microenvironment activates the paracrine Hedgehog/GLI signaling pathway, which promotes VEGF-dependent endothelial cell activation and tumor angiogenesis, thereby promoting tumor progression [42]. CAFs can also provide nutrients to tumor cells by promoting vascular mimicry (VM). Through in vitro experiments, Tsai et al. showed that CAFs can induce lung cancer cells to form capillary-like structures, provide a channel for tumor cells and tumor-promoting neutrophils, and alter the ability of tumors to form VM networks through Notch2-Jagged1-mediated direct contact [43].

Enhance invasion and metastasis

CAFs can promote tumor metastasis to distant organs by promoting increased tumor aggressiveness, which is also one of the crucial mechanisms of NSCLC progression. Studies have shown that CAFs can promote metastasis by secreting cytokines, and the stromal cell-derived factor-1 (SDF-1) secreted by CAFs enhances the expression of C-X-C chemokine receptor type 4 (CXCR4), β-catenin, and peroxisome proliferator-activated receptor delta (PPARδ) in LUAD cells, enhancing their aggressiveness [44]. RAS-related C3 botulinum toxin substrate 1 (RAC1), a small molecule G protein, is a signaling molecule that regulates various cellular activities and gene expression. T-cell lymphoma invasion and metastasis 2 (TIAM2), which is highly expressed in CAFs, can promote the invasion and migration of NSCLC cells by activating RAC1 [45]. CAFs-specific miR-196a promotes increased tumor aggressiveness by activating annexin A1 (ANXA1) and secreting CCL2 [46].

CAFs could also promote tumor metastasis through autocrine. CAFs promote NSCLC invasion by activating HMGB1/NF-κB while maintaining tumor cell viability through the autocrine high-mobility protein B1 (HMGB1) circuit [47]. In addition, CAFs can also promote lymphatic metastasis. LUAD cells co-cultured with PDPN+ CAFs have been found to have a higher risk of lymph node metastasis [28].

Promote chronic inflammation

The human immune system can recognize and respond to tumor cells, and inappropriate and sustained activation of the immune response can lead to a chronic inflammatory state, thereby increasing the risk of tumorigenesis and promoting tumor progression, in which CAFs play an important role. CAFs can promote tumor inflammatory states by secreting pro-inflammatory cytokines. CAFs produce large amounts of CXCL12, which enhances colony-forming capacity and promotes the production of pro-inflammatory chemokine CCL20 in NSCLC [48]. The expression of genes related to the inflammatory response pathway in CAFs was up-regulated, and the cardiotrophin-like cytokine factor 1/ciliary neurotrophic factor receptor (CLCF1/CNTFR) and IL-6/IL-6R signaling pathways played an important role in promoting the growth of NSCLC [49]. Conventional chemotherapy mainly targets actively-cycling cancer cells, while cancer progression in NSCLC patients may result from a small number of slow-cycling cancer cells with a quiescent-like phenotype. It has been found that slow-cycling cancer cells can form a pro-inflammatory microenvironment through activating transcription factor 6 (ATF6)-mediated transcriptional upregulation of various pro-inflammatory cytokines and synergistically recruit fibroblasts in a paracrine manner. Activated fibroblasts are up-regulated by COX2 and Col-I transcriptionally, triggering the transition of PGE2 and integrin/Src signal-mediated slow-cycling cancer cells (SCCs) to actively cycling cancer cells (ACCs) [50].

Promote immune escape

The human immune surveillance system is the line of defense against tumorigenesis, and CAFs can promote cancer progression by forming an immunosuppressive microenvironment. CAFs can promote the formation of immune microenvironment by secreting cytokines and chemokines to recruit immunosuppressive cells. CAFs recruit CCR2-positive monocytes by secreting CCL2 and induce their differentiation into a myeloid-derived suppressor cell (MDSC) phenotype in LUSC [51]. NSCLC-derived CAFs form an immunosuppressive microenvironment by decreasing NK cell proliferation rate and cytotoxic capacity [52]. Tumor-associated macrophages (TAMs) can change from an anti-tumor phenotype (M1 type) to a pro-tumor phenotype (M2 type) under the influence of TME [53]. Monocytes from NSCLC tumors were isolated and co-cultured with CAFs, and it was found that CAFs promoted the differentiation of macrophages to M2 type and inhibited the pro-inflammatory properties of M1 type macrophages [54]. Several studies have found that the expression of TGF-β1 in PDPN-positive CAFs in LUSC is up-regulated. TGFβ could inhibit the activation and proliferation of effector T cell subsets, including Th1, Th2 cells, and cytotoxic T lymphocytes, and promote the differentiation of naïve CD4 positive T cells into Treg cells, suggesting that PDPN positive CAFs are associated with immunosuppressive TME [31]. CAFs could also inhibit the infiltration and migration of immune cells by remodeling the extracellular matrix (ECM) and preventing the binding of immune checkpoint antibodies to their targets, thereby forming immune escape. T cells typically accumulate in fibrous loose areas of the tumor interstitium, and the dense ECM acts as a barrier to hinder the contact between T cells and tumor cells while inhibiting T cells’ binding to PD-1 inhibitors, thereby promoting resistance to immune checkpoint inhibitors [55].

Regulate tumor cell metabolism

Tumor cells adjust their metabolic patterns to maintain high proliferation rates in a hypoxic and low nutrient-concentration environment. CAFs are thought to be the primary regulators of tumor metabolism. The Warburg effect is currently the most widely accepted mode of tumor metabolism, the metabolic mode in which tumor cells are inclined to aerobic glycolysis. Pyruvate kinase M2 (PKM2) expression is up-regulated in NSCLC cells under hypoxia, and exosomes of tumor cells can deliver PKM2 to CAFs. Conversely, the PKM2 high CAFs release pyruvate, lactate, ketone bodies, and glutamine through autophagy, creating a favorable microenvironment that supports tumor growth [56]. As knowledge of tumor metabolomics continues to grow, it has been discovered that tumor cells can also be powered by the anti-Warburg effect. It has been found that CAFs can provide energy support to NSCLC cells by downregulating the expression of glycolytic enzymes in tumor cells and increasing the ability of CAFs to release lactate [57]. In vitro co-culture studies suggest that CAFs increase the glycolytic capacity of tumor cells, while tumor cells increase the mitochondrial function of CAFs, and metabolic reprogramming is associated with reactive oxygen species (ROS) and TGFβ signaling pathways [58]. Metabolic crosstalk between CAFs and tumor cells can lead to the progression of tumors.

Promote resistance to anti-tumor therapies

Despite the continuous development of therapeutic approaches, tumor resistance is inevitable, with CAFs mediating fundamental mechanisms of NSCLC resistance to chemotherapy, targeted therapy, immunotherapy, and radiation therapy [59]. CAFs could promote chemotherapy resistance in NSCLC by mediating EMT, remodeling the ECM, maintaining stemness of CSCs, and modulating metabolic reprogramming, making NSCLC cells resistant to tyrosine kinase inhibitors by promoting tumor EMT and forming a hypoxic microenvironment, and promote immune escape mainly by remodeling the ECM and preventing immune checkpoint inhibitors from binding to their targets [23]. CAFs can mediate resistance to NSCLC through a variety of paracrine signals. The study found that CAFs can promote the chemoresistance of NSCLC to cisplatin through the IL-11/IL-11R/STAT3 signaling pathway, mediate drug resistance through the IGF2/AKT/Sox2/ABCB1 signaling pathway, and secrete chondroitin sulfate proteoglycan serglycin (SRGN) and tumor cell receptor CD44 interactions promote a malignant phenotype [31]. CAFs after radiotherapy can promote radiotherapy resistance of NSCLC cells through the JAK/STAT pathway, and the apoptosis of CAFs can be specifically induced by the Foxo4-p53 interfering peptide FOXO4-DRI, which has a significant radiosensitizing effect on NSCLC cells and has a significant therapeutic impact on alleviating radiation-induced pulmonary fibrosis [59]. The resistance mechanism of CAFs to treatment also requires the involvement of other components of the TME. Studies have shown that CAFs and the ECMs they produce can act as physical barriers, thereby preventing efficient drug delivery and inhibiting apoptosis of tumor cells. TME changes caused by anti-tumor therapy will promote NSCLC treatment resistance through CAFs. Tumor necrosis factor superfamily member 4 (TNFSF4) was significantly up-regulated in CAFs under the stressful environment of chemotherapy, radiotherapy, and anti-angiogenic therapy, and TNFSF4 inhibited apoptosis of LUAD cells by enhancing the activity of the NF-κB/Bcl-XL pathway in LUAD cells, thereby promoting chemoresistance in LUAD cells [60]. In addition, the balance between apoptosis and autophagy of cancer cells could be modulated by CAFs through exosome-transferred non-coding RNAs and paracrine signaling (such as IL-6), which might be engaged in resistance to cancer treatment [15].

Unlock the phenotypic plasticity of NSCLC cells

Cell differentiation results in cell inhibition of proliferative activity in most cases, and unlocking tumor cell phenotypic plasticity is an important feature of tumor progression. CAFs can unlock phenotypic plasticity in tumor cells by promoting differentiation and metastasis. The expression of SOX2/SOX9 and NKX2-1 (TTF1) in the lung epithelium is critical in determining lung progenitor cells’ lineage properties and cell fate. In the TME co-culture system composed of epithelial cells in LUSC, CAFs, and extracellular mesenchymal cells, Chen et al. found that under the influence of the microenvironment, epithelial cells changed from proliferation and dysplasia to invasion and phenotypic transition of stem cells, during which SOX2 overexpression mediated the transition from hyperplasia to dysplasia, while CAFs mediated invasive changes [61]. Netrin-1 is up-regulated in most human tumors, and interference with Netrin-1 is associated with tumor growth and metastasis inhibition. It has been found that the secretion of Netrin-1 by CAFs during co-culture with cancer cells is associated with an increase in CSCs, and the Netrin-1 inhibitor Net-1-mab inhibits the plasticity of NSCLC cells by regulating the expression of CAFs-mediated cytokines such as IL-6 and inhibiting cell-to-cell signaling between CAFs and cancer cells [62]. In the NSCLC model, CAFs can also activate the IGF-1R signaling pathway through the secreted insulin-like growth factor (IGF), induce the expression of stem cell-related gene Nanog, and thus convert tumor cells to CSCs.

Target therapy against CAFs in NSCLC

CAFs play an essential role in the occurrence and progression of NSCLC, and more and more studies have demonstrated that targeting CAFs is a promising therapeutic strategy. Currently, there are two main strategies: to inhibit the production of pro-tumor CAFs and to block the downstream signaling pathway of CAFs (Figure 1).

Figure 1: 
Therapeutic targets to CAFs in NSCLC. Various stromal cells in the tumor microenvironment could be transformed into CAFs, including resident fibroblast, mesenchymal stem cells, pericytes, smooth muscle cells, adipocytes, endothelial cells, and epithelial cells. There are two pathways to target CAFs: (Strategy 1) by inhibiting the production of tumor-promoting CAFs, such as inhibiting the differentiation of other cells into CAFs by blocking TGFβ and inhibiting Smad3, eliminating tumor-promoting subtypes of CAFs by targeting FAP, and differentiating CAFs into resting or anti-tumor phenotypes by modulating metabolism of CAFs; (Strategy 2) through targeting downstream signaling pathways of CAFs, such as inhibiting generation of growth signals and metabolic reprogramming by targeting CDK4-pRb axis, inhibiting the stemness of CSCs by targeting IGF-2/IGF1R/Nanog pathway, inhibiting angiogenesis by targeting the mir-210/JAK2/STAT3 pathway, inhibiting migratory and invasive capability by targeting SDF-1/CXCR4/β-catenin/PPARδ pathway, inhibiting the creation of pro-inflammatory niche by targeting CLCF1–CNTFR axis, inhibiting immune escape by targeting CCR2, and inhibiting therapeutic resistance by targeting TNFSF4.
Figure 1:

Therapeutic targets to CAFs in NSCLC. Various stromal cells in the tumor microenvironment could be transformed into CAFs, including resident fibroblast, mesenchymal stem cells, pericytes, smooth muscle cells, adipocytes, endothelial cells, and epithelial cells. There are two pathways to target CAFs: (Strategy 1) by inhibiting the production of tumor-promoting CAFs, such as inhibiting the differentiation of other cells into CAFs by blocking TGFβ and inhibiting Smad3, eliminating tumor-promoting subtypes of CAFs by targeting FAP, and differentiating CAFs into resting or anti-tumor phenotypes by modulating metabolism of CAFs; (Strategy 2) through targeting downstream signaling pathways of CAFs, such as inhibiting generation of growth signals and metabolic reprogramming by targeting CDK4-pRb axis, inhibiting the stemness of CSCs by targeting IGF-2/IGF1R/Nanog pathway, inhibiting angiogenesis by targeting the mir-210/JAK2/STAT3 pathway, inhibiting migratory and invasive capability by targeting SDF-1/CXCR4/β-catenin/PPARδ pathway, inhibiting the creation of pro-inflammatory niche by targeting CLCF1–CNTFR axis, inhibiting immune escape by targeting CCR2, and inhibiting therapeutic resistance by targeting TNFSF4.

CAFs can form pro-tumor phenotypic CAFs through interactions with tumor cells, profibrotic cytokines such as TGFβ, the surrounding extracellular matrix, and other environmental factors, and epigenetic reprogramming through DNA methylation, histone modification, and mRNA expression changes, and targeted CAFs reprogramming is also a potential therapeutic target for NSCLC [19]. TGFβ signals can be transported into CAFs through the transporter Smad3, which regulates target genes. Studies have shown that M2 phenotypic TAMs can generate CAFs through macrophage-myofibroblast transition (MMT), and Smad3 expression is up-regulated in this process. At the same time, the use of Smad3 inhibitors can inhibit the differentiation of CAFs and tumor progression in vivo, which will be a potential therapeutic target for NSCLC [24]. A KRAS G12D-driven LUAD model has found that FAP+ CAFs depletion can indirectly inhibit tumor cell proliferation, increase collagen accumulation, reduce myofibroblast content, and reduce vascular density in tumors, suggesting that targeting specific subtypes of CAFs may be an effective treatment for NSCLC [26]. In addition, there are subsets of CAFs with tumor-suppressive effects in NSCLC. CD200-positive CAFs can enhance the sensitivity of NSCLC to gefitinib, and CD99 overexpression in CAFs may inhibit tumor progression, suggesting that restoring the activated state of pro-tumor CAFs to a quiescent state or even inducing them to acquire a tumor suppressive phenotype is also a possible and practical option [29].

Tumor inhibition by inhibiting the downstream pathway of CAFs is another well-established option. High expression of GGT5 in CAFs enhances LUAD resistance by increasing intracellular glutathione and decreasing reactive oxygen species within cancer cells. In contrast, cancer cell proliferation is attenuated in a conditioned medium for GGT5-silencing CAFs and targeting GGT5 with the small molecule inhibitor GGsTop can inhibit tumor growth and increase the chemosensitivity of cancer cells [30]. Glycosylation of the CDK4-pRb axis in LUAD CAFs can silence tumor cell growth inhibition signals, and targeting the CDK4-pRb axis glycosylation pathway is a potential pathway to enhance the anticancer effect of CKD4 inhibitors [40]. CAFs can maintain tumor cell stemness through IGF-2/IGF1R/Nanog paracrine signaling, while inhibition of Nanog expression by IGF-2/IGF1R signaling blockade can attenuate the stem cell characteristics of CSCs, providing a new strategy for the treatment of NSCLC [31]. CAFs can promote the angiogenesis of NSCLC, and targeting the miR-210/JAK2/STAT3 signal transduction pathway can inhibit the expression of CAFs’ pro-angiogenic factors MMP9, FGF2, and VEGFA, inhibit the production of blood vessels, and thus inhibit the nutrient acquisition of NSCLC [40]. CAFs regulate EMT in LUAD tissues through the SDF-1/CXCR4/β-catenin/PPARδ pathway, while SDF-1 antibody blockade SDF-1 signaling reduces tumor tissue invasiveness, suggesting that SDF-1/CXCR4/β-catenin/PPARδ is a potential therapeutic target for NSCLC [44]. CAFs can up-regulate the expression of genes related to inflammatory response pathways in cells, promote the chronic inflammatory environment of tumors through the CLCF1–CNTFR axis, and promote tumor growth. In contrast, knockdown of the expression of the CLCF1–CNTFR axis will inhibit tumor growth [49]. In lung squamous cell carcinoma cells, CAFs recruit CCR2+ monocytes and induce their differentiation into an MDSC phenotype, and CCR2 inhibition or ROS clearance inhibits the CAFs-MDSC axis, reversing the CAFs-mediated immunosuppressive microenvironment [51]. CAFs interact with tumor cells to interact with each other and metabolically reprogram through ROS and TGFβ signaling pathways, thereby promoting tumor growth, suggesting that targeting the crosstalk pathway between the two is a potential therapeutic strategy [58]. Inhibition of TNFSF4 can reduce the activity of the NF-κB/Bcl-XL pathway in LUAD cells and inhibit the formation of chemoresistance [60]. CAFs regulate the plasticity of tumor cells by secreting Netrin-1 and IGF while inhibiting the secretion of Netrin-1 and IGF can inhibit the increase of CSCs, thereby inhibiting tumor growth [62].

Conclusions

As an essential component of TME, CAFs are related to many characteristics of NSCLC, including promoting tumor growth and maintaining stemness, promoting angiogenesis and metastatic invasion, promoting tumor chronic inflammatory state, promoting immunosuppressive microenvironment, changing metabolic regulation, affecting tumor drug resistance, and unlocking tumor cell phenotypic plasticity. Due to their essential role in tumors, CAFs have become a potential target for cancer therapy.

However, the heterogeneity of the source, function, and phenotype of CAFs provides the possibility and challenges for the individualized treatment of NSCLC. Several typical CAFs-related markers have been identified to help distinguish them from normal fibroblasts, but due to the low specificity and heterogeneity of the markers, a single marker is not feasible, and the combination of different markers is of great significance for the classification of CAFs. Therefore, more in-depth research on CAFs will further deepen the understanding of TME and facilitate more individualized treatment of NSCLC.


Corresponding author: Yan Li, Department of Respiratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China; and Department of Respiratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China, E-mail: ; and Guichun Huang, Medical School, Nanjing University, Nanjing, Jiangsu, China, E-mail:

Award Identifier / Grant number: BK20221168

Funding source: Nanjing Municipal Health and Family Planning Commission

Award Identifier / Grant number: YKK22084

Acknowledgments

We thank Dr. Chunyan Zhu for revising the figure.

  1. Research ethics: Not applicable.

  2. Informed consent: Not applicable.

  3. Author contributions: The authors confirm contribution to the paper as follows: data collection: Xiaoyan Feng and Binghan Zhu; draft manuscript preparation: Xiaoyan Feng, Binghan Zhu, Yali Peng, Kaiyuan Zhang, Yangchun Wang; revision: Guichun Huang and Yan Li. All authors reviewed the results and approved the final version of the manuscript.

  4. Competing interests: The authors declare that they have no conflicts of interest to report regarding the present work.

  5. Research funding: The work was supported by grants from the Natural Science Foundation of Jiangsu Province (No. BK20221168) and Nanjing Municipal Health and Family Planning Commission (YKK22084).

  6. Data availability: The readers can access the data used in the study by contacting Yan Li.

References

1. Siegel, RL, Giaquinto, AN, Jemal, A. Cancer statistics, 2024. CA Cancer J Clin 2024;74:12–49. https://doi.org/10.3322/caac.21820.Search in Google Scholar PubMed

2. Zhang, Y, Vaccarella, S, Morgan, E, Li, M, Etxeberria, J, Chokunonga, E, et al.. Global variations in lung cancer incidence by histological subtype in 2020: a population-based study. Lancet Oncol 2023;24:1206–18. https://doi.org/10.1016/s1470-2045(23)00444-8.Search in Google Scholar

3. Kratzer, TB, Bandi, P, Freedman, ND, Smith, RA, Travis, WD, Jemal, A, et al.. Lung cancer statistics, 2023. Cancer 2024;130:1330–48. https://doi.org/10.1002/cncr.35128.Search in Google Scholar PubMed

4. Li, S, Meng, C, Hao, Q, Zhou, R, Dai, L, Guo, Y, et al.. “On/off” – switchable crosslinked PTX-nanoformulation with improved precise delivery for NSCLC brain metastases and restrained adverse reaction over nab-PTX. Biomaterials 2024;307:122537. https://doi.org/10.1016/j.biomaterials.2024.122537.Search in Google Scholar PubMed

5. Nakamichi, S, Kubota, K, Misumi, T, Kondo, T, Murakami, S, Shiraishi, Y, et al.. Phase II study of durvalumab immediately after completion of chemoradiotherapy in unresectable stage III non-small cell lung cancer: TORG1937 (DATE study). Clin Cancer Res 2024;30:1104–10. https://doi.org/10.1158/1078-0432.ccr-23-2568.Search in Google Scholar PubMed PubMed Central

6. Park, S, Kim, TM, Han, JY, Lee, GW, Shim, BY, Lee, YG, et al.. Phase III, randomized study of atezolizumab plus bevacizumab and chemotherapy in patients with EGFR- or ALK-mutated non-small-cell lung cancer (ATTLAS, KCSG-LU19-04). J Clin Oncol 2024;42:1241–51. https://doi.org/10.1200/jco.23.01891.Search in Google Scholar PubMed PubMed Central

7. Herbst, RS, Garon, EB, Kim, DW, Cho, BC, Gervais, R, Perez-Gracia, JL, et al.. Five year survival update from KEYNOTE-010: pembrolizumab versus docetaxel for previously treated, programmed death-ligand 1-positive advanced NSCLC. J Thorac Oncol 2021;16:1718–32. https://doi.org/10.1016/j.jtho.2021.05.001.Search in Google Scholar PubMed

8. Jaiyesimi, IA, Leighl, NB, Ismaila, N, Alluri, K, Florez, N, Gadgeel, S, et al.. Therapy for stage IV non–small cell lung cancer with driver alterations: ASCO Living Guideline, Version 2023.3. J Clin Oncol 2024;42:e1–e22. https://doi.org/10.1200/jco.23.02744.Search in Google Scholar

9. Ricciuti, B, Lamberti, G, Puchala, SR, Mahadevan, NR, Lin, JR, Alessi, JV, et al.. Genomic and immunophenotypic landscape of acquired resistance to PD-(L)1 blockade in non-small-cell lung cancer. J Clin Oncol 2024;42:1311–21. https://doi.org/10.1200/jco.23.00580.Search in Google Scholar PubMed PubMed Central

10. Saigí, M, Carcereny, E, Morán, T, Cucurull, M, Domènech, M, Hernandez, A, et al.. Biological and clinical perspectives of the actionable gene fusions and amplifications involving tyrosine kinase receptors in lung cancer. Cancer Treat Rev 2022;109:102430. https://doi.org/10.1016/j.ctrv.2022.102430.Search in Google Scholar PubMed

11. John, N, Schlintl, V, Sassmann, T, Lindenmann, J, Fediuk, M, Wurm, R, et al.. Longitudinal analysis of PD-L1 expression in patients with relapsed NSCLC. J Immunother Cancer 2024;12:e008592. https://doi.org/10.1136/jitc-2023-008592.Search in Google Scholar PubMed PubMed Central

12. Hanahan, D. Hallmarks of cancer: new dimensions. Cancer Discov 2022;12:31–46. https://doi.org/10.1158/2159-8290.cd-21-1059.Search in Google Scholar PubMed

13. Elanany, MM, Mostafa, D, Hamdy, NM. Remodeled tumor immune microenvironment (TIME) parade via natural killer cells reprogramming in breast cancer. Life Sci 2023;330:121997. https://doi.org/10.1016/j.lfs.2023.121997.Search in Google Scholar PubMed

14. Xiang, H, Pan, Y, Sze, MA, Wlodarska, M, Li, L, van de Mark, KA, et al.. Single-cell analysis identifies NOTCH3-mediated interactions between stromal cells that promote microenvironment remodeling and invasion in lung adenocarcinoma. Cancer Res 2024;84:1410–25. https://doi.org/10.1158/0008-5472.can-23-1183.Search in Google Scholar

15. Chen, C, Liu, J, Lin, X, Xiang, A, Ye, Q, Guo, J, et al.. Crosstalk between cancer-associated fibroblasts and regulated cell death in tumors: insights into apoptosis, autophagy, ferroptosis, and pyroptosis. Cell Death Discov 2024;10:189. https://doi.org/10.1038/s41420-024-01958-9.Search in Google Scholar PubMed PubMed Central

16. Kalluri, R. The biology and function of fibroblasts in cancer. Nat Rev Cancer 2016;16:582–98. https://doi.org/10.1038/nrc.2016.73.Search in Google Scholar PubMed

17. Lambrechts, D, Wauters, E, Boeckx, B, Aibar, S, Nittner, D, Burton, O, et al.. Phenotype molding of stromal cells in the lung tumor microenvironment. Nat Med 2018;24:1277–89. https://doi.org/10.1038/s41591-018-0096-5.Search in Google Scholar PubMed

18. Para, R, Romero, F, George, G, Summer, R. Metabolic reprogramming as a driver of fibroblast activation in pulmonary fibrosis. Am J Med Sci 2019;357:394–8. https://doi.org/10.1016/j.amjms.2019.02.003.Search in Google Scholar PubMed PubMed Central

19. Alcaraz, J, Ikemori, R, Llorente, A, Díaz-Valdivia, N, Reguart, N, Vizoso, M. Epigenetic reprogramming of tumor-associated fibroblasts in lung cancer: therapeutic opportunities. Cancers 2021;13:3782. https://doi.org/10.3390/cancers13153782.Search in Google Scholar PubMed PubMed Central

20. Sentek, H, Klein, D. Lung-resident mesenchymal stem cell fates within lung cancer. Cancers 2021;13:4637. https://doi.org/10.3390/cancers13184637.Search in Google Scholar PubMed PubMed Central

21. Wu, DM, Liu, T, Deng, SH, Han, R, Zhang, T, Li, J, et al.. Alpha-1 antitrypsin induces epithelial-to-mesenchymal transition, endothelial-to-mesenchymal transition, and drug resistance in lung cancer cells. Onco Targets Ther 2020;13:3751–63. https://doi.org/10.2147/ott.s242579.Search in Google Scholar PubMed PubMed Central

22. Wang, Y, Chu, Y, Ren, X, Xiang, H, Xi, Y, Ma, X, et al.. Epidural adipose tissue-derived mesenchymal stem cell activation induced by lung cancer cells promotes malignancy and EMT of lung cancer. Stem Cell Res Ther 2019;10:168. https://doi.org/10.1186/s13287-019-1280-3.Search in Google Scholar PubMed PubMed Central

23. Saw, PE, Chen, J, Song, E. Targeting CAFs to overcome anticancer therapeutic resistance. Trends Cancer 2022;8:527–55. https://doi.org/10.1016/j.trecan.2022.03.001.Search in Google Scholar PubMed

24. Tang, PC, Chung, JY, Xue, VW, Xiao, J, Meng, XM, Huang, XR, et al.. Smad3 promotes cancer-associated fibroblasts generation via macrophage-myofibroblast transition. Adv Sci 2022;9:e2101235. https://doi.org/10.1002/advs.202101235.Search in Google Scholar PubMed PubMed Central

25. Kou, Z, Liu, C, Zhang, W, Sun, C, Liu, L, Zhang, Q. Heterogeneity of primary and metastatic CAFs: from differential treatment outcomes to treatment opportunities (review). Int J Oncol 2024;64:54. https://doi.org/10.3892/ijo.2024.5642.Search in Google Scholar PubMed PubMed Central

26. Chen, X, Song, E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discov 2019;18:99–115. https://doi.org/10.1038/s41573-018-0004-1.Search in Google Scholar PubMed

27. Holm, NS, Willumsen, N, Leeming, DJ, Daniels, SJ, Brix, S, Karsdal, MA, et al.. Serological assessment of activated fibroblasts by alpha-smooth muscle actin (α-SMA): a noninvasive biomarker of activated fibroblasts in lung disorders. Transl Oncol 2019;12:368–74. https://doi.org/10.1016/j.tranon.2018.11.004.Search in Google Scholar PubMed PubMed Central

28. Suzuki, H, Kaneko, MK, Kato, Y. Roles of podoplanin in malignant progression of tumor. Cells 2022;11:575. https://doi.org/10.3390/cells11030575.Search in Google Scholar PubMed PubMed Central

29. Chen, C, Hou, J, Yu, S, Li, W, Wang, X, Sun, H, et al.. Role of cancer-associated fibroblasts in the resistance to antitumor therapy, and their potential therapeutic mechanisms in non-small cell lung cancer. Oncol Lett 2021;21:413. https://doi.org/10.3892/ol.2021.12674.Search in Google Scholar PubMed PubMed Central

30. Wei, JR, Dong, J, Li, L. Cancer-associated fibroblasts-derived gamma-glutamyltransferase 5 promotes tumor growth and drug resistance in lung adenocarcinoma. Aging 2020;12:13220–33. https://doi.org/10.18632/aging.103429.Search in Google Scholar PubMed PubMed Central

31. Zeltz, C, Primac, I, Erusappan, P, Alam, J, Noel, A, Gullberg, D. Cancer-associated fibroblasts in desmoplastic tumors: emerging role of integrins. Semin Cancer Biol 2020;62:166–81. https://doi.org/10.1016/j.semcancer.2019.08.004.Search in Google Scholar PubMed

32. Li, K, Wang, R, Liu, GW, Peng, ZY, Wang, JC, Xiao, GD, 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. https://doi.org/10.1038/s41598-024-55375-0.Search in Google Scholar PubMed PubMed Central

33. Yang, P, Luo, Q, Wang, X, Fang, Q, Fu, Z, Li, J, et al.. Comprehensive analysis of fibroblast activation protein expression in interstitial lung diseases. Am J Respir Crit Care Med 2023;207:160–72. https://doi.org/10.1164/rccm.202110-2414oc.Search in Google Scholar PubMed PubMed Central

34. Hu, H, Piotrowska, Z, Hare, PJ, Chen, H, Mulvey, HE, Mayfield, A, et al.. Three subtypes of lung cancer fibroblasts define distinct therapeutic paradigms. Cancer Cell 2021;39:1531–47.e10. https://doi.org/10.1016/j.ccell.2021.09.003.Search in Google Scholar PubMed PubMed Central

35. Cords, L, Engler, S, Haberecker, M, Rüschoff, JH, Moch, H, de Souza, N, et al.. Cancer-associated fibroblast phenotypes are associated with patient outcome in non-small cell lung cancer. Cancer Cell 2024;42:396–412.e5. https://doi.org/10.1016/j.ccell.2023.12.021.Search in Google Scholar PubMed PubMed Central

36. Zhang, Y, Bian, Y, Wang, Y, Wang, Y, Duan, X, Han, Y, et al.. HIF-1α is necessary for activation and tumour-promotion effect of cancer-associated fibroblasts in lung cancer. J Cell Mol Med 2021;25:5457–69. https://doi.org/10.1111/jcmm.16556.Search in Google Scholar PubMed PubMed Central

37. Li, H, Zhang, Q, Wu, Q, Cui, Y, Zhu, H, Fang, M, et al.. 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;11:4077–88.Search in Google Scholar

38. Guo, X, Chen, M, Cao, L, Hu, Y, Li, X, Zhang, Q, et al.. Cancer-associated fibroblasts promote migration and invasion of non-small cell lung cancer cells via miR-101-3p mediated VEGFA secretion and AKT/eNOS pathway. Front Cell Dev Biol 2021;9:764151. https://doi.org/10.3389/fcell.2021.764151.Search in Google Scholar PubMed PubMed Central

39. Bouchard, G, Garcia-Marques, FJ, Karacosta, LG, Zhang, W, Bermudez, A, Riley, NM, et al.. Multiomics analysis of spatially distinct stromal cells reveals tumor-induced O-glycosylation of the CDK4-pRB axis in fibroblasts at the invasive tumor edge. Cancer Res 2022;82:648–64. https://doi.org/10.1158/0008-5472.can-21-1705.Search in Google Scholar PubMed PubMed Central

40. Fan, J, Xu, G, Chang, Z, Zhu, L, Yao, J. miR-210 transferred by lung cancer cell-derived exosomes may act as proangiogenic factor in cancer-associated fibroblasts by modulating JAK2/STAT3 pathway. Clin Sci 2020;134:807–25. https://doi.org/10.1042/cs20200039.Search in Google Scholar

41. Hegab, AE, Ozaki, M, Kameyama, N, Gao, J, Kagawa, S, Yasuda, H, et al.. Effect of FGF/FGFR pathway blocking on lung adenocarcinoma and its cancer-associated fibroblasts. J Pathol 2019;249:193–205. https://doi.org/10.1002/path.5290.Search in Google Scholar PubMed

42. Katoh, M. Genomic testing, tumor microenvironment and targeted therapy of Hedgehog-related human cancers. Clin Sci (Lond) 2019;133:953–70. https://doi.org/10.1042/cs20180845.Search in Google Scholar PubMed

43. Tsai, YM, Wu, KL, Liu, YW, Chang, WA, Huang, YC, Chang, CY, et al.. 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. https://doi.org/10.3389/fonc.2021.696931.Search in Google Scholar PubMed PubMed Central

44. Wang, Y, Lan, W, Xu, M, Song, J, Mao, J, Li, C, 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. https://doi.org/10.1038/s41419-021-03509-x.Search in Google Scholar PubMed PubMed Central

45. Li, S, Ou, Y, Liu, S, Yin, J, Zhuo, W, Huang, M, et al.. The fibroblast TIAM2 promotes lung cancer cell invasion and metastasis. J Cancer 2019;10:1879–89. https://doi.org/10.7150/jca.30477.Search in Google Scholar PubMed PubMed Central

46. Lee, S, Hong, JH, Kim, JS, Yoon, JS, Chun, SH, Hong, SA, et al.. Cancer-associated fibroblasts activated by miR-196a promote the migration and invasion of lung cancer cells. Cancer Lett 2021;508:92–103. https://doi.org/10.1016/j.canlet.2021.03.021.Search in Google Scholar PubMed

47. Ren, Y, Cao, L, Wang, L, Zheng, S, Zhang, Q, Guo, X, et al.. Autophagic secretion of HMGB1 from cancer-associated fibroblasts promotes metastatic potential of non-small cell lung cancer cells via NFκB signaling. Cell Death Dis 2021;12:858. https://doi.org/10.1038/s41419-021-04150-4.Search in Google Scholar PubMed PubMed Central

48. Wald, O, Izhar, U, Amir, G, Kirshberg, S, Shlomai, Z, Zamir, G, et al.. Interaction between neoplastic cells and cancer-associated fibroblasts through the CXCL12/CXCR4 axis: role in non-small cell lung cancer tumor proliferation. J Thorac Cardiovasc Surg 2011;141:1503–12. https://doi.org/10.1016/j.jtcvs.2010.11.056.Search in Google Scholar PubMed

49. Vicent, S, Sayles, LC, Vaka, D, Khatri, P, Gevaert, O, Chen, R, et al.. Cross-species functional analysis of cancer-associated fibroblasts identifies a critical role for CLCF1 and IL-6 in non-small cell lung cancer in vivo. Cancer Res 2012;72:5744–56. https://doi.org/10.1158/0008-5472.can-12-1097.Search in Google Scholar

50. Cho, J, Lee, HJ, Hwang, SJ, Min, HY, Kang, HN, Park, AY, et al.. The interplay between slow-cycling, chemoresistant cancer cells and fibroblasts creates a proinflammatory niche for tumor progression. Cancer Res 2020;80:2257–72. https://doi.org/10.1158/0008-5472.can-19-0631.Search in Google Scholar

51. Xiang, H, Ramil, CP, Hai, J, Zhang, C, Wang, H, Watkins, AA, et al.. Cancer-associated fibroblasts promote immunosuppression by inducing ROS-generating monocytic MDSCs in lung squamous cell carcinoma. Cancer Immunol Res 2020;8:436–50. https://doi.org/10.1158/2326-6066.cir-19-0507.Search in Google Scholar PubMed

52. 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. https://doi.org/10.3389/fimmu.2020.602530.Search in Google Scholar PubMed PubMed Central

53. Singhal, S, Stadanlick, J, Annunziata, MJ, Rao, AS, Bhojnagarwala, PS, O’Brien, S, et al.. Human tumor-associated monocytes/macrophages and their regulation of T cell responses in early-stage lung cancer. Sci Transl Med 2019;11:eaat1500. https://doi.org/10.1126/scitranslmed.aat1500.Search in Google Scholar PubMed PubMed Central

54. Berzaghi, R, Ahktar, MA, Islam, A, Pedersen, BD, Hellevik, T, Martinez-Zubiaurre, I. Fibroblast-mediated immunoregulation of macrophage function is maintained after irradiation. Cancers 2019;11:689. https://doi.org/10.3390/cancers11050689.Search in Google Scholar PubMed PubMed Central

55. Pei, L, Liu, Y, Liu, L, Gao, S, Gao, X, Feng, Y, et al.. Roles of cancer-associated fibroblasts (CAFs) in anti-PD-1/PD-L1 immunotherapy for solid cancers. Mol Cancer 2023;22:29. https://doi.org/10.1186/s12943-023-01731-z.Search in Google Scholar PubMed PubMed Central

56. Gustafsson, J, Roshanzamir, F, Hagnestål, A, Patel, SM, Daudu, OI, Becker, DF, et al.. Metabolic collaboration between cells in the tumor microenvironment has a negligible effect on tumor growth. Innovation 2024;5:100583. https://doi.org/10.1016/j.xinn.2024.100583.Search in Google Scholar PubMed PubMed Central

57. Duda, P, Janczara, J, McCubrey, JA, Gizak, A, Rakus, D. The reverse Warburg effect is associated with Fbp2-dependent Hif1α regulation in cancer cells stimulated by fibroblasts. Cells 2020;9:205. https://doi.org/10.3390/cells9010205.Search in Google Scholar PubMed PubMed Central

58. Cruz-Bermúdez, A, Laza-Briviesca, R, Vicente-Blanco, RJ, García-Grande, A, Coronado, MJ, Laine-Menéndez, S, et al.. Cancer-associated fibroblasts modify lung cancer metabolism involving ROS and TGF-β signaling. Free Radic Biol Med 2019;130:163–73. https://doi.org/10.1016/j.freeradbiomed.2018.10.450.Search in Google Scholar PubMed

59. Meng, J, Li, Y, Wan, C, Sun, Y, Dai, X, Huang, J, et al.. Targeting senescence-like fibroblasts radiosensitizes non-small cell lung cancer and reduces radiation-induced pulmonary fibrosis. JCI Insight 2021;6:e146334. https://doi.org/10.1172/jci.insight.146334.Search in Google Scholar PubMed PubMed Central

60. Li, Y, Chen, Y, Miao, L, Wang, Y, Yu, M, Yan, X, et al.. Stress-induced upregulation of TNFSF4 in cancer-associated fibroblast facilitates chemoresistance of lung adenocarcinoma through inhibiting apoptosis of tumor cells. Cancer Lett 2021;497:212–20. https://doi.org/10.1016/j.canlet.2020.10.032.Search in Google Scholar PubMed

61. Chen, S, Giannakou, A, Wyman, S, Gruzas, J, Golas, J, Zhong, W, et al.. Cancer-associated fibroblasts suppress SOX2-induced dysplasia in a lung squamous cancer coculture. Proc Natl Acad Sci U S A 2018;115:E11671–e80. https://doi.org/10.1073/pnas.1803718115.Search in Google Scholar PubMed PubMed Central

62. Sung, PJ, Rama, N, Imbach, J, Fiore, S, Ducarouge, B, Neves, D, et al.. Cancer-associated fibroblasts produce Netrin-1 to control cancer cell plasticity. Cancer Res 2019;79:3651–61. https://doi.org/10.1158/0008-5472.can-18-2952.Search in Google Scholar

Received: 2024-05-06
Accepted: 2024-08-06
Published Online: 2024-09-02

© 2024 the author(s), published by De Gruyter on behalf of Tech Science Press (TSP)

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Frontmatter
  2. Review Articles
  3. Autophagy and radiotherapy in esophageal cancer: modulating treatment sensitivity and overcoming challenges
  4. Functions of CAFs in microenvironment of non-small cell lung cancer: based on updated hallmarks of cancer
  5. Cisplatin-induced pyroptosis: a double-edged sword in cancer treatment
  6. Radiotherapy directed to inferior vena cava tumor thrombus among patients with renal cell carcinoma: an illustrative case and review of the literature
  7. Research Articles
  8. A lipid metabolism-related gene model reveals the prognosis and immune microenvironment of cutaneous melanoma
  9. Boanmycin induces apoptosis and overcomes venetoclax resistance in acute myeloid leukemia
  10. Identification of GNB1 as a downstream effector of the circRNA-0133711/miR-145-5p axis involved in breast cancer proliferation and metastasis
  11. Clinical characteristics and prognosis of lung metastases from unknown primary cancer sites
  12. Integrating bulk-RNA and single-cell analysis reveals heterogeneous expression of cuproptosis-related sorafenib-resistant genes in hepatocellular carcinoma
  13. Exploring the mechanism of genistein in treating hepatocellular carcinoma through network pharmacology and molecular docking
  14. Investigating the potential mechanisms of Litsea cubeba essential oil for anti-melanoma through experimental validation, network pharmacology, and molecular docking analysis
  15. Exploring the anti-lung cancer mechanism of Ganoderma lucidum and its relationship with the level of immune cell infiltration based on network pharmacology and molecular docking
  16. Constructing a prognostic model for colon cancer patients on basis of coagulation genes enriched in cancer-associated fibroblasts to guide personalized immunotherapy
  17. Rapid Communication
  18. Evaluation of HER2 immunohistochemistry-positive and immunohistochemistry-negative FISH amplification breast cancers using next-generation sequencing
  19. Short Commentary
  20. Unveiling the unexplored: shedding light on a novel aspect of colorectal carcinoma
Downloaded on 17.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/oncologie-2024-0232/html
Scroll to top button