Skip to main content

Cancer associated fibroblasts in cancer development and therapy

Abstract

Cancer-associated fibroblasts (CAFs) are key players in cancer development and therapy, and they exhibit multifaceted roles in the tumor microenvironment (TME). From their diverse cellular origins, CAFs undergo phenotypic and functional transformation upon interacting with tumor cells and their presence can adversely influence treatment outcomes and the severity of the cancer. Emerging evidence from single-cell RNA sequencing (scRNA-seq) studies have highlighted the heterogeneity and plasticity of CAFs, with subtypes identifiable through distinct gene expression profiles and functional properties. CAFs influence cancer development through multiple mechanisms, including regulation of extracellular matrix (ECM) remodeling, direct promotion of tumor growth through provision of metabolic support, promoting epithelial-mesenchymal transition (EMT) to enhance cancer invasiveness and growth, as well as stimulating cancer stem cell properties within the tumor. Moreover, CAFs can induce an immunosuppressive TME and contribute to therapeutic resistance. In this review, we summarize the fundamental knowledge and recent advances regarding CAFs, focusing on their sophisticated roles in cancer development and potential as therapeutic targets. We discuss various strategies to target CAFs, including ECM modulation, direct elimination, interruption of CAF-TME crosstalk, and CAF normalization, as approaches to developing more effective treatments. An improved understanding of the complex interplay between CAFs and TME is crucial for developing new and effective targeted therapies for cancer.

Graphical abstract

Introduction

During the development of the mammalian embryo, normal fibroblasts (NFs) that are important for forming connective tissues of the body can originate from the primitive mesenchyme and share a common embryonic lineage with other mesenchymal cell types, such as adipocytes, chondrocytes, and osteoblasts [1]. In normal physiological conditions, fibroblasts serve as primary contributors to the extracellular matrix (ECM) of connective tissues and are central to the process of tissue repair during stress and in injury states where, in such situations, these cells are activated in response to signals and tissue damage [2,3,4]. Beyond their reparative roles, fibroblasts also facilitate angiogenesis by secreting vascular endothelial growth factor A (VEGFA) [5,6,7], as well as orchestrate immune system functions through the release of chemokines and cytokines [7,8,9,10,11].

In cancer states, fibroblasts are known to constitute a significant component of the tumor stroma. Moreover, fibroblasts within tumors have been found to undergo phenotypic as well as functional transformation, typically observed as a result of interactions with resident tumor cells and, as such, these “cancer-associated fibroblasts (CAFs)” have been found to influence the tumor microenvironment (TME) [13, 14]. Despite our current understanding of CAFs, their precis identification continues to pose a challenge, owing to a lack of definitive molecular markers that distinguish them from other cells, as well as from embryonic mesenchyme [15,16,17,18]. Indeed, molecular markers such as α-SMA, fibroblast activation protein (FAP), platelet-derived growth factor receptor (PDGFR) and podoplanin (PDPN), have all been employed to facilitate the identification of CAFs [19,20,21,22]. Typically, these markers are combined with lineage markers to enable negative selection, through the exclusion of epithelial cells, tumor cells, myeloid-derived cells, endothelial cells [23, 24]. A list of typical molecular markers for CAFs is provided in Table 1.

Table 1 Frequently used makers for identifying CAFs

The importance of CAFs in cancer development is underscored by their roles in multiple hallmarks of cancer, including ECM formation, tumor cell proliferation, invasion, metastasis, angiogenesis, and immune suppression [13]. Moreover, the heterogeneity of CAFs within the TME adds a further dimension of complexity to their functions [25]. Studies using single-cell RNA sequencing (scRNA-seq) have identified distinct subsets of CAFs that feature unique gene expression profiles and functional properties. These subsets, such as myofibroblastic CAFs (myoCAFs), inflammatory CAFs (iCAFs), and antigen-presenting CAFs (apCAFs), have been found to influence tumor progression and immune responses in different ways, through a variety of mechanisms including the secretion of specific cytokines, chemokines, and ECM components [26,27,28,29,30].

In this review, we comprehensively describe the multifaceted roles of CAFs in cancer development. Moreover, we focus on the latest findings on the complex heterogeneity of CAFs and plasticity within the TME. Also, we review the potential for CAFs as therapeutic targets, discuss the emerging strategies to target CAFs in cancer treatment. Taken togther, through our exploration of the complex interplay between CAFs and the TME, we bring attention to current efforts that could lead to the development of more effective and targeted therapies for cancer.

Origins of CAFs and their activation

CAFs are known to derive from resident fibroblasts within normal tissues, however they have also been described to arise from the cellular transformation of adipocytes, pericytes, stellate cells, pericryptal cells, mesothelial cells, mesenchymal stem cells (MSCs) [31,32,33,34]. Continuous observations of the transformation process, from normal epithelia to atypical hyperplasia to invasive carcinoma, have revealed a concomitant proliferation and dysfunction of fibroblasts. Notably, cultured NFs have been found to be transformed into CAFs under certain conditions, such as when exposed to conditioned media (CM) derived from tumor cells, or under hypoxia conditions, or upon exposure to TGFβ, or through the addition of cancer-derived exosome preprations [35,36,37,38]. For example, in breast cancer (BC), resident human mammary fibroblasts can transform into myoCAFs via autocrine TGF-β which binds to TGF-β I/II receptor and activates TGFβ/Smad signaling pathway. Moreover, such signalling in these cells also promotes CXCL12 secretion, which additionally forms a positive feedback loop through CXCR4 stimulation. Thus, TGF-β and CXCL12 concurrently induce myoCAFs through an autostimulatory mechanism as well as through their signalling cross-communication [39].

In addition to myoCAFs, studies have shed light on the origins of other CAFs subtypes. For example, in mouse models of colitis-associated colorectal cancer (CRC), it has been reported that cells expressing the pericryptal leptin receptor (Lepr) proliferate and transform to CAFs that express the melanoma cell adhesion molecule (MCAM), and such CAFs promote an immune suppressive TME [40, 41]. In separate scenario, pancreatic stellate cells (PSCs), when activated, transform into αSMA-expressing CAFs that secrete factors that promote tumor growth, cell survival, and metastasis [42]. Moreover, in pancreatic ductal carcinoma (PDAC), mesothelial cells have been found to transform into apCAFs under the regulation of IL-1 and TGFβ, and this was associated with downregulation of mesothelial features and upregulation of fibroblastic features [43]. Also, in prostate cancer, TGF-β1 secreted by tumor cells and tumor stroma recruits MSCs to the tumor tissue site and induce their differentiation into CAFs [44]. Furthermore, in breast cancer, Wnt3a derived from tumor cells induces adipocytes to transform into adipocyte-derived fibroblasts (ADFs) [32]. In ovarian cancer, Eckert et al. discovered through a proteomic analysis study that Nicotinamide N-methyltransferase (NNMT) expression is essential for the functional aspects of cancer-associated fibroblasts (CAFs), including their expression of CAF markers, cytokine secretion characteristics, and their production of carcinogenic extracellular matrix molecules. Mechanistically, the expression of NNMT in CAFs leads to S-adenosylmethionine (SAM) depletion, reduced histone methylation, and widespread gene expression changes in cells of the tumor stroma. Accordingly, knockdown of NNMT in CAFs resulted in morphological changes such that they more closely resembled normal stromal fibroblasts, and this visual feature was concomitant with a reduction in the expression of CAF markers [45]. Taken together, these examples showcase the diverse cellular origins for CAFs and how each distinct CAF subtype shapes their functional specialization within the TME (Fig. 1). This topic is further explored below.

Fig. 1
figure 1

Origins of CAFs. CAFs may originate from various cell types including pericytes, adipocytes, stellate cells, mesothelial cells, epithelial cells, endothelial cells, mesenchymal Stem Cells (MSCs) and normal fibroblast. This figure was created with BioRender.com

Approaches to studying CAFs

The approaches to studying CAFs can be considered to comprise four interconnected strategies, namely: (1) isolation via primary culture or cytokine-induced differentiation [42, 46, 47] (Fig. 2A); (2) animal models such as genetically engineered mouse models (GEMMs) (e.g., KPC model for PDAC) that model spontaneous carcinogenesis and enable conditional CAFs depletion as well as xenograft transplantation [48,49,50,51,52,53] (Fig. 2B); (3) in vitro systems (2D/3D co-culture, CM analysis, organoids) or in vivo co-implantation to dissect tumor-CAF interactions at the cellular and molecular levels [54,55,56,57,58,59,60,61] (Fig. 2C); (4) combinations of classical techniques (such as immunohistochemistry, flow cytometry) with emerging tools (such as scRNA-seq and spatial transcriptomics (ST)) to reveal complex molecular functions of CAFs within the tumor microenvironment [62] (Fig. 2D).

Fig. 2
figure 2

Methodology for studying CAFs. (A) CAFs may be obtained through primary culture, or transformation from other cells, such as normal fibroblasts or pancreatic stellate cells (PSCs), for further researches. (B) Animal models of CAFs include patient-derived xenografts (PDX) and transplanted tumor models established from cell lines. Genetically engineered mouse models (GEMMs) can spontaneously develop tumors, better simulating the interaction between CAFs and TME. These genetically engineered mice also allow for the specific elimination of CAF subsets using drugs such as diphtheria toxin, as well as lineage tracing of CAF subsets through the expression of specific markers. (C) In vitro approaches to study CAF-TME interactions include 2D/3D co-culture, conditioned media transfer, and tumor organoid models. In vivo methods involve co-inoculation of CAFs and tumor cells. (D) CAF research integrates techniques such as western blotting (WB), flow cytometry, and immunofluorescence. Advanced tools like single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics provide deeper functional insights. This figure was created with BioRender.com

Multifaceted roles of CAFs in cancer development

Modulation of the ECM

CAFs are the primary contributors to ECM formation through synthesis and deposition of collagen fibers [32]. A study analyzing collagen expression across distinct CAFs subtypes has elucidated that CAF subtypes possess unique collagen expression profiles. For example, myoCAFs are characterized by the expression of collagens such as COL10A1 and COL11A1, while iCAFs predominantly express COL14A1 [26]. In another example, Lambrechts et al. performed a scRNA-seq to define five CAFs clusters in non-small cell lung cancers (NSCLCs). Cluster 1 expressed COL10A1 and shows high expression of ECM proteins and TGF-β-related genes, while Cluster 2 was found to express COL4A1, along with high levels of ACTA2, MEF2C, MYH11 expression [62]. Through their role in ECM deposition, CAFs can create a physical barrier that hinders the entry of immune cells as well as therapeutic agents into the tumor tissue and, as such, protect tumor cells from immune system attacks and anti-tumor treatment [47] (Fig. 3A).

Fig. 3
figure 3

CAFs modulate ECM to promote tumor growth. (A) CAFs promote the deposition of extracellular matrix, forming a physical barrier that hinders immune cell infiltration and drug penetration, thereby creating obstacles for anti-tumor treatment. (B) CAFs remodel ECM, facilitating tumor cell invasion and metastasis. (C) CAFs regulate the balance between dense ECM and loose ECM, thus promoting cancer progression. This figure was created with BioRender.com

The ECM remodeling mediated by CAFs is also known to influence tumor invasion. CAFs can secrete matrix metalloproteinases (MMPs), which are zinc-dependent endopeptidases that degrade ECM components such as collagens, fibronectin, elastin and laminin, so as to facilitate stromal degradation and tumor cell invasion [63, 64]. Studies of cocultures of carcinoma cells and stromal fibroblasts have revealed that fibroblasts predominantly serve as lead cells during invasion, while carcinoma cells trail behind, navigating along pathways in ECM generated by the lead fibroblasts. As such, fibroblasts are shown as pivotal for facilitating cancer cell invasion through a mechanims that involves protease-driven and force-mediated reorganization of the ECM [65]. Of the molecular players involved, caveolin-1 (Cav1) is a membrane-associated protein that plays a crucial role cell signaling in the process, as well as in ECM reconstruction, and tumor progression [66]. Indeed, expression of Cav1 in CAFs enhances cell elongation in 3D cultures via p190RhoGAP, promoting force-dependent contraction, matrix alignment. Cav1-deficient mice exhibit stromal disorganization, while Cav1-rich stroma in human carcinomas and melanoma metastases promotes tumor aggressiveness [60]. Moreover, GP130-IL6ST and JAK1 stimulate RhoA-dependent actomyosin contractility in both tumor cells and CAFs, leading to matrix reconstruction and enhanced tumor cell migration. Notably, inhibition of the signaling axis that comprises these key factors may be crucial for developing interventions that effectively block tumor cell invasion [67] (Fig. 3B).

The balance between ECM deposition and degradation is crucial for maintaining ECM homeostasis, but this balance can be dysregulated in tumor microenvironments [68] (Fig. 3C). Moreover, CAFs secrete enzymes that facilitate collagen cross-linking which, in turn, further enhances the rigidity of the ECM. The stiffness of tumor tissues is generally significantly higher than that of normal tissues [69], and this stiffness of the ECM has been shown to promote tumor progression by restricting drug penetration, limiting the infiltration of immune cells, and activating signaling pathways such as integrin-mediated focal adhesion kinase (FAK/SRC) signaling as well as mechanotransduction pathways (e.g. Rho-GTPase, ERK, YAP/TAZ) [70,71,72]. For instance, studies of the impact of stiffness on cultured intrahepatic cholangiocarcinoma (ICC) and hepatocellular carcinoma (HCC) cells showed that numbers were higher at 16 kPa stiffness rather than at 2 kPa, and that such cells appearred more robust and appeared to be accompanied by features such as a more regular actin arrangement and higher YAP1 content [69]. In a cohort of 107 colorectal cancer patients, the median overall survival (OS) of the low-stiffness group was significantly longer than in the high-stiffness group (54.38 ± 2.64 months vs 64.05 ± 1.00 months, P = 0.007) [73]. Recently, Zheng et al. revealed that in PDAC, matrix stiffness could drive tumor proliferation through glycolytic metabolism via upregulated CLIC1 expression mediated by the Wnt/β-catenin/TCF4 signaling pathway [74]. Through digital pathology analysis, Maiques et al. found that the morphology of extracellular matrix fibers increased from the tumor core to the invasive front, and that fibres appeared longer and denser. The changes in fibers at the invasive front induced tumor cell cytoskeletal reconfiguration through the Rho-ROCK-MLC2 pathway, and promoted cell contraction, reduced adhesion, and induced pseudopod-like behaviors. In another example, it was reported that a “mechanical-inflammation signature” (MIS) gene set was upregulated at the invasive front and associated with poor patient prognosis [75]. Zheng et al. found through scRNA-seq that PDGFRα + ITGA11 + CAFs are associated with vascular invasion and lymph node metastasis in early-stage bladder cancer by promoting collagen deposition as well as enhancing the migratory ability of tumor cell spheroids via the glycoprotein CHI3L1. Notably, treatment that combined CHI3L1-neutralizing antibodies and ITGA11-neutralizing antibodies could inhibit vascular invasion in early BC [76]. While the current evidence largely supports the notion that ECM stiffness exerts a tumor-promoting effect, a few studies have conversely found that a reduction in ECM components could facilitate tumor progression [77,78,79]. For example, in PDAC models, an anti–lysyl oxidase like-2 (anti-LOXL2) antibody decreased matrix content and this was associated with accelerated tumor growth and lower tissue stiffness [78]. One possible explanation is that ECM remodeling involves intricate signaling networks, such that a reduction in ECM stiffness caused by specific factors might paradoxically trigger compensatory pro-tumorigenic signaling pathways. For example, Hedgehog signaling blockage reduces the stroma in PDAC, but accelerates tumor progression through increased blood vessel formation [80]. Further research is needed to further clarify these issues.

Components of the ECM, such as CTGF and integrins, play complex roles in signal transduction. For example, blocking CTGF signaling can enhance the efficacy of chemotherapy without altering the effective concentration of gemcitabine, through an effect that is accompanied by a reduction in the expression of X-linked inhibitor of apoptosis protein (XIAP) [81]. Remarkably, a recent study shows that in PDAC organoids, hyaluronic acid (HA) interacts with the CD44 receptor which, in turn, promotes the expression of drug efflux transporters, leading to chemotherapy resistance. Moreover, this resistance can be reversed by transferring the organoids from high-stiffness to low-stiffness matrices, and such findings offer a novel strategy for improving chemotherapy resistance by targeting the ECM [82].

CAFs interact with multiple immune cell types within the TIME (Tumor immune microenvironment)

CAFs have been found to have interactions with multiple immune cell types in the TIME, including effector T cells, regulatory T cells (Tregs), tumor associated macrophages (TAMs), tumor associated neutrophils (TANs), and myeloid derived suppresor cells (MDSCs) [83, 84], as explained further below.

Effector T cells and Tregs

CAFs can inhibit the functions of effector T cells through the production of immunosuppressive factors such as IL-6, CXCL9, CXCL12 and TGFβ, and by direct cell–cell interaction through PD-1/PD-L1 receptor signaling [36, 85, 86] (Fig. 4A). In PDAC that arises in the KPC mouse model, CXCL12 secreted by FAPα + CAFs is the primary cause of immunosuppression, where CD8 + T cells do not respond to anti-CTLA-4 and anti-PD-L1 therapies [87]. In studies of human esophageal carcinoma, the numbers of CAFs are negatively associated with the presence of CD8 + tumor infiltreated lymphcytes (TILs). Co-implantation of CT26 cells and CAFs have been shown to result in accelerated tumor growth, accompanied with decreased CD8 + T cells and increased FoxP3 + TILs [88]. Recently, a study that utilised ST and scRNA-seq by Song et al. revealed that CAFs suppress the infiltration and function of effector T cells by expressing CXCL12 and PD-L1, a mechanism maintained via autocrine IGF2. This finding is consistent with clinical studies that show that high expression of IGF2 is associated with suboptimal outcomes of anti-PD-1 therapy. Indeed, targeting the IGF2/IGF1R signaling pathway with the inhibitor linsitinib can significantly enhance the efficacy of immune checkpoint inhibitors (ICIs) [89].

Fig. 4
figure 4

CAFs modulate the tumor immune microenvironment (TIME). (A) CAFs inhibit the function of effector T cells by secreting IL-6, CXCL12, TGF-β, and by expressing PD-L1; they also recruit regulatory T cells (Tregs). (B) CAFs recruit macrophages through cytokines such as IL-6, CXCL12, and IL-33, and promote M2 polarization and inducing immunosuppresive subses like lipid-associated macrophages (LAMs). (C) CAFs promote N2 polarization of TANs, upregulate PD-L1, and stimulate neutrophil extracellular trap (NET) formation. (D) CAFs enhance infiltration of monocytic (M-MDSCs) and polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) via tumor-derived CSF-1. This figure was created with BioRender.com

Although the immunosuppressive roles of CAFs are well appreciated, some studies also suggest certain subgroups of CAFs may even play an immune promotive role. In PDAC, depletion of αSMA + myofibroblasts led to invasive, undifferentiated tumors with increased CD4 + Foxp3 + Tregs, suggesting their role in promoting anti-tumor immunity [90]. A subgroup of PDPN + CAFs were reported to be able to induce the initiation and expansion of tertiary lymphoid structures (TLSs) that could enhance the efficiency of immune surveillance and locally potentiate immune responses against tumor antigens. Notably, these TLSs were found to often correlate with increased numbers of mature dendritic cells and robust T-cell infiltration [91, 92]. Through scRNA-seq and ST analysis of HCC samples, it was found that tumors from patients with a good response to immunotherapy exhibited more infiltration of CCL19 + fibroblasts and plasma cells. This co-localization was associated with T-cell infiltration and the formation of TLSs. In contrast, in immune-exclusion samples, DKK1 + tumor cells accumulated at the tumor boundary, inhibiting the infiltration of CCL19 + fibroblasts and plasma cells into the tumor area [93]. Thus, the interactions between CAFs and effector T cells and Tregs remain to be better understood.

Tumor-associated macrophages (TAMs)

CAFs can recruit and induce monocyte differentiation into M2-like TAMs through cytokines such as CXCL12, CCL5, IL-6, IL-33, GM-CSF and glycoprotein Chitinase-3-like-1 (Chi3L1) [41, 94,95,96,97,98,99] (Fig. 4B). Besides, CAFs increase macrophage adhesion through interactions between the class A scavenger receptor (SR-A/CD204) and cleaved type I collagen, which is selectively cleaved by FAP expressed on CAFs [100]. Furthermore, in triple negative BC (TNBC), CAFs recruit and reprogram blood monocytes into an immune suppressive lipid associated macrophage (LAM) subset via the CXCL12–CXCR4 axis, which is marked by STAB1 as well as high TREM2 expression. This LAM subset has been reported to be expanded in patients refractory to immune checkpoint blockade (ICB). In preclinical models, genetic depletion of these LAMs leads to TNBC tumor growth suppression [101].

HIF2 expression in CAFs is known to induce the immunosuppressive environment of PDAC. Studies using scRNA-seq have showed that recruitment of MDSC in CAF-HIF2 knockout tumors was inhibited in this context. Accordingly, the proportion of M2-polarized TAMs were decreased in CAF-HIF2 knockout tumors [52]. In HCC, POSTN + CAFs attract and induce macrophages to SPP1 + TAM via the IL-6/STAT3 signaling pathway, which in turn hinders the effective infiltration of T-cells and diminishes the efficacy of immunotherapy. Patients with increased expression of both POSTN + CAFs and SPP1 + macrophages showed poorer therapeutic outcomes in an immunotherapy cohort [102]. Similarly, in the case of gastric cancer (GC), it has been shown that thrombospondin 2 (THBS2)-expressing matrix CAFs (mCAFs) promote the recruitment of peritoneum-specific macrophages, which then differentiate into SPP1 + TAMs through the C3-C3AR1 axis which, in turn, disrupts this signaling axis and enhances the efficacy of ICIs [103].

The interplay between CAFs and TAMs is reciprocal, such that TAMs also affect the functions of CAFs. In PDAC, IL-33-stimulated macrophages secret CXCL3, which induce the transition of myoCAFs and upregulates α-SMA expression thorugh activation of the CXCL3–CXCR2 signaling pathway [104]. M2 polarization of TAMs increases their capability to modulate the EMT in CAFs, resulting in their enhanced reactivity [105].

Tumor-associated neutrophils (TANs)

In HCC, CAFs secrete CXCL12, which recruits neutrophils and also upregulates their PD-L1 expression via a IL6-JAK-STAT3 signaling pathway. These activated PD-L1 + neutrophils exert a tumor-promoting effect by suppressing T-cell immunity [106]. Similar findings were reported in GC, in that myeloid stem cells differentiate into CAFs upon stimulation from neutrophils. The former reciprocally activate neutrophils through IL-6–STAT3–ERK1/2 signaling cascade [31]. Moreover, CAFs produced cardiotrophin-like cytokine factor 1 (CLCF1), which stimulated the secretion of CXCL6 and TGF-β from tumor cells. These secreted factors subsequently facilitated TAN infiltration and N2 polarization through a paracrine mechanism [107].

In addition to secretion of CXCL12, CAFs are known to secrete amyloid β to stimulate the formation of neutrophil extracellular traps (NETs) by a reactive oxygen species (ROS)-mediated pathway, which facilitates tumor progression [108]. Also, pancreatic cancer cells can trigger the formation of NETs, which subsequently induce hepatic stellate cells into CAFs. In liver metastasis mouse models, DNase I, an inhibitor of NETs, effectively suppressed the occurrence of liver metastasis in vivo (Fig. 4C) [109].

Myeloid-derived suppressor cells (MDSCs)

In liver cancer, CCL2 secreted by FAP + CAFs could promote tumor growth by enhancing the recruitment of MDSCs via a STAT3 signaling pathway, which is abolished in Ccr2-deficient mice or by exposure to the CCR2 inhibitor, sc-202525 [46, 110]. By secreting CXCL16, CAFs attract monocytes that then differentiate into monocytic MDSCs, activating the tumor stroma and exacerbating TNBC aggressiveness [111]. The expression of granulocytic chemokines in CAFs is downregulated by HDAC2, induced by tumor cell derived CSF1. This downregulation restricts the migration of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) towards tumors. Interventions with CSF1R inhibitors disrupt this crosstalk, leading to a marked enhancement in the recruitment of PMN-MDSCs to tumor sites which, in turn, contributes to immunosuppression [112,113,114] (Fig. 4C).

Angiogenesis

CAFs are capable of secreting a variety of angiogenic regulators such as VEGFA, platelet-derived growth factor-C (PDGF-C), fibroblast growth factor 2 (FGF2), CXCL12, and WNT2 [115,116,117,118,119,120]. A study by Orimo et al. found that CAFs co-planted with human BC MCF-7 cells resulted in significantly increased tumor vascular density. Further analysis revealed that CAFs could recruit endothelial progenitor cells (EPCs), a bone-marrow derived stem cell population, into the tumor by releasing CXCL12 which, in turn, resulted in an increase in the number of EPCs within the tumor. Experiments using in vivo approaches have found that, this process of EPC recruitment into tumors could be inhibited by neutralizing antibodies against CXCL12, thereby inhibiting tumor growth [121]. In a separate study utilising a CRC xenograft mouse model, overexpression of WNT2 resulted in increased tumor vessel density and tumor volume. Mass spectrometry and cytokine array analyses collectively revealed that WNT2 overexpression resulted in upregulation of the expression proteins associated with pro-angiogenic functions, including extracellular matrix molecules, ANG-2, IL-6, G-CSF, and placental growth factor (PGF) [120]. A cohort study of 535 prostate cancer patients who underwent surgery showed that PDGFRβ is expressed in the stromal tissues of prostate cancers, and that high expression was associated with poor prognosis [122]. Moreover, in a cervical cancer model, PDGF expressed by cancer cells activate PDGFR on CAFs, and this promoted their release of FGF which, in turn, enhanced angiogenesis. Studies have shown that inhibition of PDGFR signaling with a small molecular inhibitor Imatinib could be effective to suppress the growth of invasive carcinomas [115, 123, 124].

Collectively, by secreting a variety of angiogenic factors, CAFs can recruit and activate cells related to angiogenesis, such as endothelial cells and monocytes. These cells then gather and proliferate at the tumor site and form new vascular networks that provide essential nutrients and oxygen that further support tumor growth and metastasis. These studies shown that, by facilitating the formation of new blood vessels in tumors, CAFs enhance the metastatic capability of tumors. Thus, targeting CAFs or their capacity for facilitating angiogenesis may be a therapeutic avenue for intervention in some cancers [125].

The interplay between CAFs and tumor cells

Regulation of metabolism

Recent studies indicate that CAFs are key regulators in modulating tumor cell metabolism, particularly through the regulation of pathways for glucose, amino acid, and lipid metabolism [126, 127] (Fig. 5A). A lipid-rich CAFs subset marked by Abca8a expression has been identified in SETD2-deficient pancreatic tumors through scRNA-seq experiments. Notably, loss of SETD2 activates the BMP2 signaling pathway through ectopic gene expression, thereby promoting the differentiation of lipid-rich CAFs. These ABCA8a-expressing lipid-rich CAFs can promote tumor growth by transfer of lipids and can support oxidative phosphorylation by tumor cells [128]. In another example, FSP + CAFs have been shown to increase glycolysis in malignant cells through FAK signaling. Indeed, FAK depletion in CAFs leads to increases in levels of CCL6, CCL11, CCL12 and pentraxin-3, which altogether enhance glycolysis by malignant cells through activating protein kinase A via CCR1/CCR2 [129]. Moreover, secretion of acetate, a metabolic product, by CAFs serves as a critical driver for the accelerated growth of PDAC cells under low pH conditions. Acetate, mediated by acetyl-CoA synthetase 2 (ACSS2), induces acetylation of histones and the oncogenic SAT1 gene, upregulating its expression through epigenetic regulation. It has been shown that knockdown of SAT1 expression levels significantly inhibits PDAC cell growth rates and impairs their survival under low pH conditions. The ACSS2–SP1–SAT1 regulatory axis is a pivotal mechanism by which CAF-derived acetate fuels PDAC cell survival and proliferation, underscoring its potential as a therapeutic target for intervention [130].

Fig. 5
figure 5

Interplays between CAFs and tumor cells. (A) CAFs fuel tumor cell glycolysis via CCL6 and CCL12 secretion; In addition, Abca8a + CAFs facilitate tumor cell growth by secreting lipid droplets. (B) CAFs enhance tumor proliferation and therapy resistance through IL-6, HGF, and FGF2 secretion, and via exosomal long non-coding RNAs (lncRNAs). (C) CAFs induce epithelial-mesenchymal transition (EMT) via Hedgehog ligands, TGF-β, and HGF. (D) CAF-derived exosomes containing miR-522 suppress lipid peroxidation to inhibit ferroptosis. (E) Complement component 5a (C5a) activates GPR77 + CAFs, inducing tumor stemness via IL-6, IL-8, and TGF-β. This figure was created with BioRender.com

CAFs directly promoting tumor growth and treatment resistance

CAFs have been suggeseted to directly promote tumor growth by enhancing cancer cell proliferation and treatment resistance through IL-6 (Fig. 5B). In studies of co-cultures with ductal carcinoma in situ (DCIS) cells, CAFs have been reported to facilitate the formation of multicellular structures via paracrine IL-6 signaling [57]. In the context of esophageal cancer, IL-6 is aberrantly expressed in fibroblasts and epithelial cells, and its levels have been found to increase in co-cultures of cancer cells and CAFs, resulting in tumor progression [88, 131]. In BC, CAFs promote cancer cell proliferation and radioresistance via IL-6 secretion through the STAT3 pathway, and support cancer progression by secreting hepatocyte growth factor (HGF) and FGF2 via paracrine signaling [132,133,134]. Additionally, in the context of high dose cancer chemotherapy, CAFs have been found to secrete chemokines that mediate treatment resistance and tumor growth, while low-dose metronomic therapy is suppressive for such pro-cancer signaling [135]. Furthermore, exosomes derived from CAFs that contain LncRNAs such as miR-500a-5p and miR-181d-5p have been found to promote tumor growth through epigenetic mechanisms [136, 137]. Co-culture of PDPN + CAFs with cancer cells induced the resistance of such cells to EGFR-TKIs, and this effect was reversed by knockdown of PDPN expression. Accordingly, patients with PDPN + CAFs showed significantly lower response rates to EGFR-TKIs (53% vs. 83%; P < 0.01), suggesting that a better understanding of this subtype of CAFs may be key to enhancing the efficacy of EGFR-TKIs in cancer [138]. In squamous cell carcinoma, activation of nuclear receptors (NRs) in CAFs are associated with resistance to chemotherapy. While cisplatin alone showed limited efficacy, combination therapies with NRs antagonists such as LE135 and bicalutamide significantly delayed tumor growth and enhanced treatment efficacy by modulating CAF-driven processes like invasiveness, proliferation, and energy metabolism [139]. Taken together, while these examples show the capacity for CAFs to directly influence tumor growth and treatment resistance, the extent through which CAFs exert such direct effects across cancers remains unclear.

CAFs promote EMT

EMT is an important biological process in which tumor cells transform from epithelial-like cells into mesenchymal-like cells that are associated with specific features including enhanced migration and tissue invasion (Fig. 5C) [58, 140]. Studies of co-cultures of PDAC cells with CAFs have found that such mixtures of these cell types results in a remarkable transcriptional heterogeneity at the single-cell level, yet appears to foster gene expression signatures reminiscent of EMT and progenitor (PRO) phenotypes within PDAC cell lines [141]. CAFs have been reported to promote EMT through cytokines such as Hedgehog ligand, TGFβ, HGF, IL6 and IGF-1 [34, 37, 142, 143]. When co-cultured with CAFs, Panc01 cells exhibit EMT features characterized by their increased invasive ability, expression of Vimentin and decreased expression of E-cadherin, features of which could be blocked by inhibiting Hedgehog signaling. Co-culture with CAFs or addition of CM from CAFs can also promote EMT in BC and bladder cancer [37, 143, 144]. Mechanistically, CAFs induce EMT through the phosphorylation of annexin A2 (ANXA2) mediated by HGF and IGF-1 [145]. Moreover, TGFβ secreted by CAFs have been found to induce EMT through an epigenetic mechanism involving the lncRNA ZEB2NAT and HOTAIR [146, 147]. A recent pan-cancer transcriptomic analysis showed that most THBS family genes are upregulated in cancer tissue samples at advanced stages of disease. Immunohistochemistry and multiplex immunofluorescence experiments have revealed that THBS2 is primarily localized in the regions where CAFs reside, and its expression is higher in patients who are unresponsive to oxaliplatin treatment. Molecular docking and Co-IP experiments confirm that COL8A1 that is secreted by THBS2 + CAFs, interacts with ITGB1 and this, in turn, activates the PI3K-AKT signaling pathway to promote EMT, and leading to oxaliplatin resistance in colorectal cancer. Thus a THBS2 + CAFs-COL8A1-ITGB1 axis might be an important consideration in the development of therapeutic interventions for reversing chemotherapy resistance in this form of cancer [148].

Regulation of programmed cell death by CAFs

Programmed cell death (PCD) is an important cellular mechanism in tumor cell biology, and can significantly influence tumor initiation and progression, as well as enhance or curtical the efficacies of tumor treatments and immunotherapies [149,150,151]. Studies have demonstrated that CAFs can regulate the levels of ferroptosis by tumor cells through secretion of exosomes containing miR-522. When these CAF-derived exosomes are internalized by tumor cells to deliver miR-522, this results in the post-transcriptional inhibition of expression of ALOX15, a protein involved in lipid peroxidation regulation. Indeed, this reduction of ALOX15 reduces the accumulation of lipid ROS and inhibits the occurrence of ferroptosis [152] (Fig. 5D).

CAFs maintaining cancer stem cell property

CAFs have been reported to modulate the properties of cancer stem cells (CSCs) in scirrhous GC, through the TGFβ signaling pathway (Fig. 5E). In BC, NF-κB activation through GPR77-mediated complement signaling induces a subgroup of CD10 + GPR77 + CAFs, which promote tumor growth and chemoresistance by nurturing CSCs through secretion of IL-6 and IL-8 [153]. Moreover, CM derived from CAFs enhanced the formation of spheroid colonies and upregulated the expression of CSC markers in OCUM-12/SP and OCUM-2MD3/SP cells. The level of phospho-Smad2 was elevated upon co-culturing with CAFs, and the stimulatory effects observed with CAF-derived CM were attenuated by the application of TGFβ inhibitors [154]. In murine models of TNBC, neoplastic cells produce Hedgehog ligands that reprogram CAFs to promote a chemo-resistant, CSC phenotype. This reprogramming involves FGF5 expression and increased production of fibrillar collagen [155]. Together, these lines of evidence indicate that CAFs could directly modulate the features of CSCs so as to influence tumor growth and progression.

Heterogeneity and plasticity of CAFs

In recent years, the emerging data provided by scRNA-seq has facilitated the discovery of an increasing number of subpopulations of CAFs that are deinfed by their diverse features and unique functions. Yet, due to the lack of definitive molecular markers for CAFs, our current understanding of the spectrum of reported subpopulations across different studies remains to be clarified [25]. Table 2 summarizes the features of currently reported CAF subpopulations, as shown.

Table 2 The heterogeneity of CAFs

When considering the expression levels of αSMA and the spatial relationships with tumor cells, two CAFs subsets, myoCAFs and iCAFs, are identified in PDAC. MyoCAFs are located immediately adjacent to neoplastic cells and promote disease progression by producing pro-tumoral cytokines and participate in the formation of desmoplastic stroma. On the other hand, iCAFs are typified by their low expression of αSMA and are located further away from tumor cells and affect the tumor microenvironment by secreting IL-6 and other inflammatory mediators [26]. Moreover, myoCAFs and iCAFs show pleiotropic effects when exposed to certain factors. For example, IL-1 and TGF-β that is secreted from cancer cells have been found to antagonistically regulate the transformation of CAFs subsets in opposing ways. Notably IL-1 activates the JAK/STAT signaling pathway by inducing the expression of leukemia inhibitory factor (LIF), leading to the generation of iCAFs; while TGF-β inhibits the IL-1-induced JAK/STAT signaling pathway by downregulating the expression of IL-1 receptor 1 (IL-1R1), promoting the differentiation of CAFs towards myoCAF subtype [156].

Schwann cells are another source of IL-1α that promote the transformation of myoCAFs into iCAFs [27]. Upon inhibition of the Hedgehog pathway or NF-κb signaling pathway with the IRAK4 inhibitor CA-4948, the composition of CAFs transitions from myoCAFs to iCAFs [28, 29]. In addition, a recent study found that inhibition of TAK1 leads to decreased IL-6 expression in CAFs and increased α-SMA expression, and this promoted the conversion of iCAFs to myoCAFs which was concomitant with enhanced anti-tumor immune responses. This mechanistic switch thus positions TAK1 as a strategic therapeutic target within CAFs that could be explored further to determine the effectiveness of combining TAK1 inhibitors with ICIs in the treatment of PDAC [157]. Furthermore, along with these two subsets, studies using scRNA-seq have led to the identification of another CAF subset that is marked by its expression of MHC class II molecules and costimulatory molecules, which activate T-cells and promote anti-tumor immune responses, and thus named antigen-presenting CAFs (apCAFs) [29, 30, 158, 159].

FAP expression combined with other markers, can identify multiple CAF subtypes that typically exhibit immunosuppressive effects [160,161,162]. In BC, four CAFs subsets described which were identifiable through their expression of a series markers including FAP, CD29, aSMA, S100-A4/ FSP1, PDGFRb, and CAV1. The subset one (CAF-S1) was characterized by high FAP expression, promoted immunosuppression by secreting CXCL12, attracting CD4 + CD25 + T lymphocytes and retaining them through expression of OX40L, PD-L2, and JAM. Morevoer, CAF-S1 also prolonged T lymphocyte survival and facilitated their differentiating into Tregs, through expression of B7H3, CD73, and DPP4 [163]. Also, a CAFs population marked by co-expression of FAP + PDPN + is predominantly located at the tumor periphery near T cells and inhibits their proliferation via nitric oxide [164]. Correspondingly, a high ratio of S100A4 + to PDPN + CAFs is predictive of good prognosis across subtypes [165]. In studies of 4T1 and 4T07 orthotopic TNBC models, the heterogeneity of CAFs subsets was found to gradually diminish with toumor progression, with CD26 + CAFs and FAP + CAFs becoming the dominant subsets observed in the latter stages of tumor growth [23]. In PDAC, FAP + CAFs promote the growth of pancreatic cancer tumors through CXCL12 and CCL2 signaling, while αSMA + CAFs mainly inhibit tumor growth and contribute to the polarization of tumor-infiltrating T-cells, as revealed by studies using scRNA-seq [166].

In recent years, the advent of scRNA-seq has led to the identification of an increasing number of CAF subpopulations in cancer. Bartoschek et. al. grouped CAFs from BC mouse models into four subgroups, including vascular CAFs (vCAFs), matrix CAFs (mCAFs), cycling CAFs (cCAFs) and developmental CAFs (dCAFs). In cohorts derived from the TCGA database, the signatures of vCAF and mCAF were both independent prognostic factors for BC patients [24]. Studies of the Panc02 tumor model have found that, gemcitabine chemotherapy upregulates placental growth factor (PlGF), and induces CD141 + CD74- CAFs [86]. In loose-type PDAC, a subgroup of CAFs with a highly activated metabolic state (meCAFs) marked by PLA2G2A and CRABP2 expression was discovered, which increased activity of immune cells [79]. Moreover, a subset of myoCAFs marked by RGS5 + has been found to be crucial to the formation of metastatic lesions, predominantly in hepatic metastasis tissues [167]. Also, a lipid-rich CAFs subpopulation marked by ABCA8a in pancreatic tumors with SETD2 deficiency metabolically supports the growth and survival of tumor cells [128]. In addition, LRRC15 + CAFs, which rely on TGFβ signaling, play a pivotal role in establishing the tumor-fibroblast equilibrium that fosters tumor expansion. Notably, these cells exert a direct inhibitory effect on CD8 + T-cell function, thereby constraining the efficacy of ICB therapies [36]. Neutralization of TGFβ reshaped CAFs subpopulations, with decreased frequency and activity of myoCAFs, while promoting a CD73 + CAF population that exhibited strong response to interferon and enhanced immunomodulatory properties, which improved efficacy of PD-1 immunotherapy [168]. Using multi-omics techniques, two CAF subtypes were identified in prostate cancer regulated by: immunosuppressive ECM-associated CAFs (ECM-CAFs), and lymphocyte-associated CAF (Lym-CAFs) that promoted anti-tumor immunity. YAP1 is a key factor that regulates the transition from Lym-CAF to ECM-CAF phenotype by directly interacting with IKKα to inhibit the activation of NF-κB p65 (also known as RelA). Indeed, selective depletion of YAP1 in ECM-CAFs may be a potential strategy through which to significantly enhance the therapeutic effects of anti-PD-1 antibodies in some cancers [169].

Therapeutic implications of CAFs

Therapies that target CAFs can be categorized into four primary approaches: targeting the ECM generated by CAFs, directly eliminating CAFs marked by their specific gene expression signatures, interrupting the cross-talk between CAFs and the TME, and normalizing CAFs [170,171,172]. Studies using preclinical models have demonstrated promising anti-tumor effects of CAF-targeted treatments and this has led to clinical studies, however effective treatments remain to be determined through large-scale Phase III clinical trials [98, 168, 173,174,175,176]. Strategies to targeting CAFs for cancer therapy are summarized in Tables 3 and 4.

Table 3 Strategies to targete CAFs for cancer therapy
Table 4 Clinical trials evaluating CAFs directed therapies

Targeting CAF-derived ECM to treat cancer

HA is a major glycosaminoglycan in PDAC stroma, elevates interstitial fluid pressure and compromises vascular perfusion, limiting chemotherapeutic delivery. PEGPH20 is a pegylated recombinant hyaluronidase enzyme that targets HA. Studies shows PEGPH20 reduces tumor-associated hyaluronan, which can enhance drug delivery, decrease tumor interstitial pressure, and improve vascular permeability [177, 178]. Preclinical and phase I/Ib studies have demonstrated that PEGPH20 in combination with gemcitabine was well-tolerated and may have therapeutic benefits [179]. Although early studies have shown effectiveness, a phase III study (HALO 109–301) found that while the addition of PEGPH20 to nab-paclitaxel/gemcitabine increased the objective response rate (ORR), it did not improve OS or progression free survival (PFS) in patients with hyaluronan-high metastatic PDAC [180,181,182]. Losartan is a commonly used angiotensin II receptor blocker, originally used to treat high blood pressure by blocking the angiotensin II receptors to dilate blood vessels [183]. More recently, it has been demonstrated to exhibit anti-tumor effects by targeting the tumor stroma, reducing collagen and hyaluronan production, and consequently decreasing solid stress in tumors, which leads to improved vascular perfusion and drug delivery which, in turn, potentiates chemotherapy in BC and PDAC models [184, 185]. Moreover, losartan improves the penetration and effectiveness of nanotherapeutics in desmoplastic tumors by inhibiting collagen production [186]. Additionally, in a single-arm phase 2 clinical trial, it was associated with a high R0 resection rate when incorporated into a total neoadjuvant therapy regimen for locally advanced pancreatic cancer [187]. Tranilast is a TGF-β inhibitor, originally approved as an anti-allergic drug used to treat allergic diseases [188]. Tranilast exhibits antitumor effects by normalizing the TME, improving tumor perfusion and oxygenation, and enhancing the efficacy of both nanomedicine and immunotherapy [189, 190]. As a further example, Metformin, a widely used medication primarily prescribed for the treatment of type 2 diabetes, and the anti-fibrotic drug Pirfenidone, have both been found to reduce ECM production by inhibiting the activation of CAFs [191,192,193,194,195].

Hedgehog signal pathway plays a vital role in ECM deposition. In a hypo-perfused gemcitabine-refractory mouse model of PDAC, coadministration of IPI-926, a hedgehog inhibitor, increased tumor vasculature and gemcitabine concentration, leading to temporary disease stabilization [196]. In phase I trials, treatment with IPI-926 showed preliminary efficacy, but this approach failed in a phase II trial [197,198,199,200]. Similarly, in phase Ib and phase II trials, another antagonist, vismodegib, also failed to enhance the ORR, PFS or OS in patients with metastatic PDAC in combination with gemcitabine [201, 202]. Lee et al. showed that modulation of Hedgehog pathway disrupts the balance between epithelial and stromal elements, leading to accelerated growth of epithelial cancer cells, which may explain its failure in clinical trials [203].

ROCK, a protein kinase involved in cell migration and ECM synthesis, is overexpressed in pancreatic cancer cells and CAFs, suggesting it may be a therapeutic target [204]. Fasudil is a ROCK inhibitor that is primarily used to treat cerebral vasospasm and has shown potential in various other conditions, including cancer, due to its effects on cell motility and smooth muscle contraction. Inhibition of ROCK with fasudil reduced tumor collagen deposition, enhanced survival, and increased chemotherapy uptake, highlighting the potential of targeting tumor stroma to enhance treatment efficacy in pancreatic cancer [205, 206]. Therefore, targeting ECM produced by CAFs may be a viable approach to cancer, but futher research is necessary to develop their clinical application as an effective treatment option.

Targeting signaling pathways relevant to cross-talk between CAFs and TME

Many growth factors, interleukins, chemokines and other cytokines such as TGFβ, IL1, IL6, CXCL12, CCL6 are involved in signaling cross-talk between CAFs and TME [47, 166, 207,208,209] (Fig. 6D). Such factors are known to activate various signal pathways including Smad, JAK-STAT, NF-κB among others to induce the expression of downstream genes. Since CAFs and the TME interact through various signaling molecules, specific blockade of their signaling cross-talks likely exerts targeted anti-tumor effects. Through this line of reasoning, many targeted drugs for these signaling pathways have been evaluated in preclinical or clinical studies for such effects [87, 210,211,212,213]. Although the initial purpose of developing these drugs may not have been specifically for use to affect CAFs, they can still potentially be repurposed for application due to their ability to interfere CAF signaling [214].

Fig. 6
figure 6

Therapeutic strategies targeting CAFs. (A) CAFs normalization. Treatments such as calcipotriol, ATRA, and minnelide are designed to convert activated CAFs into a more quiescent, normal fibroblast-like state. Another approach includes CAF subpopulations shift from tumor-promoting to tumor-restrain CAFs (TR-CAFs). (B) Targeting ECM generated by CAFs. Drugs such as pirfenidone, PEGPH20 and vismodegib modify ECM remodeling process to enhance vascular perfusion and oxygenation within the tumor, thereby facilitating drug delivery. (C) Direct depletion of CAFs., strategies utilizing CAR-T cells, radiopharmaceuticals, and monoclonal antibodies (ADCs) have been employed to deplete specific CAFs subgroups through markers such as FAP and GPR77. (D) Targeting cross-talk between CAFs and TME. Specific blockade of signal pathway involved in cross-talks between CAFs and TME such as TGF-β, LIF and mTOR, can exert targeted anti-tumor effects. This figure was created with BioRender.com

It is known that expression of IL-6 is significantly increased by αSMA + CAFs during gemcitabine treatment. When the IL-6 signaling pathway was blocked, the combined use of gemcitabine and ICB therapies significantly improved the survival of mice with pancreatic cancer [166]. Similarly, IL-6 was found to protect GC cells from 5-fluorouracil-induced apoptosis via the JAK/STAT3 signaling pathway, which is reversed by treatment with the anti-IL6 mAb, tocilizumab [215]. Both pharmacological inhibition of LIF and genetic ablation of Lifr significantly impedes tumor progression and enhances the effectiveness of chemotherapy, thereby prolonging survival in mouse models of PDAC [216]. In another scenario, the Hedgehog signaling pathway is critical to promote CSCs, and therefore blocking this pathway can inhibit tumor growth. Stromal treatment of patient-derived xenografts with smoothened inhibitors (SMOi) effectively downregulates CSC marker expression by inhibiting the Hedgehog signaling pathway, thus sensitizing the tumors to docetaxel treatment. In the phase I clinical trial EDALINE, 3 out of 12 patients with TNBC derived clinical benefit from the combination therapy of the SMOi Sonidegib and docetaxel chemotherapy, with one patient achieving a complete response [155].

CAFs contribute to the development of resistance against HER2-targeted therapies by secretion of FGF5, which triggers the activation of fibroblast growth factor receptor 2 (FGFR2) in adjacent BC cells. The subsequent transactivation of HER2 by FGFR2, mediated through c-Src, ultimately leads to the emergence of resistance to HER2-directed therapeutic interventions [217, 218]. The PI3K-AKT-mTOR pathway mediates the resistance of HER2 + BC to HER2 inhibitors through bypass activation [219]. In PDAC, inhibition of the mTOR pathway with Pasireotide also enhances chemotherapy efficacy by reducing CAFs protein synthesis and tumor growth [220]. Inhibition of CSF1R leads to increased PMN-MDSC. To overcome this unfavorable effect, combination therapy of CSF1R inhibitor and CXCR2 antagonist is reasonable. This strategy not only halts the infiltration of granulocytes into tumors, but also demonstrates potent anti-tumor effects [112,113,114]. TGF-β1 secreted by CAFs activates Laminin 2 (Ln-2) through the JNK/AP1 signaling pathway thus contributing to immunosuppression. These effects could be blocked by the TGF-β receptor inhibitor galunisertib and a neutralizing antibody against TGF-β1 [47].

Recently, multi-omics analyses have revealed that AKAP12 + CAFs in TNBC secrete IL-34, which binds to CSF1R on macrophages, activating the PI3K/AKT/IL-34 signaling pathway to promote M2 polarization and induce an immunosuppressive tumor microenvironment. This is associated with resistance to PD-1 inhibitors. Blocking the IL-34/CSF1R pathway with an anti-IL-34 antibody is a promising approach to improve the effectiveness of anti-PD-1 therapy in TNBC [221]. Metabolic cross-talk of CAFs and TME is associated with ICIs resistance. Intriguingly, an elevated NAM (nicotinamide)/MNAM (methylnicotinamide) ratio positively correlates with enhanced effector T cell function, while CAFs and macrophages regulate CD8 + T cell function via nicotinamide metabolism, revealing a “face-off” mechanism. CAFs suppress CD8 + T cell cytotoxicity by expressing nicotinamide N-methyltransferase (NNMT) and producing MNAM, which is inhibited by extracellular vesicles (EVs) containing nicotinamide phosphoribosyltransferase (NAMPT) derived from macrophages via NOCTH signaling pathway. Co-administration of NAMPT-containing EVs significantly enhances the therapeutic efficacy of anti-PD-1 therapy in GC. This strategy of modulating NAM metabolism to restore sensitivity to anti-PD-1 treatment demonstrates broad therapeutic potential [222]. In addition, a Phase I/II study showed that adding the anti-CTGF antibody Pamrevlumab to gemcitabine and albumin-bound paclitaxel chemotherapy increased the ORR in unresectable PDAC (65% vs 42%). However, subsequent Phase III clinical trials failed to demonstrate a benefit in OS [223, 224]. Taken together, these examples show potential efficiacy for the use of disruptors of cross-talk signaling between CAFs and TME cells to ameliorate cancer.

Elimination of specific CAF subtypes to treat cancer

A variety of CAF subpopulations marked by candidate gene expression have been shown to promote tumor growth. Thus, the elimination of CAF subtypes via the targeting of their specific markers could be an avenue to treat cancer [225] (Fig. 6C). For example, FAP + CAFs are associated with an immunosuppressive microenvironment, and various therapies targeting FAP have been explored, including monoclonal antibodies, tumor vaccines, radioimmunoconjugates and immunotoxin [226,227,228,229,230,231]. Tumor cell vaccines or DNA vaccines targeting FAP have been found to enhance the infiltration of CD8 + T lymphocytes and inhibit the accumulation of immunosuppressive cells in the tumor microenvironment. Concurrently, the number of FAP + CAFs within the tumor was significantly reduced [174, 175]. In a C57BL/6 mouse model bearing MCA205 tumors expressing FAP, the radiolabeled drug 177Lu-FAP-2287 rapidly accumulated in MCA205-mFAP tumors, resulting in significant tumor growth inhibition and extended survival time. This treatment also augmented the therapeutic efficacy of anti-PD-1 monoclonal antibodies. 177Lu-FAP-2287 increased the infiltration of CD8 + T cells in the tumor, accompanied by the induction of STING-mediated type I interferon responses and elevated levels of costimulatory molecules such as CD86[173]. FAP-targeted CAR-T cells, followed by CLDN18.2-targeted CAR-T cells, showed enhanced antitumor activity against PDAC by reducing CAFs and MDSCs recruitment, and promoting T-cell survival in the TME. This sequential approach also improve clinical outcomes for PDAC [232]. OMTX705, an antibody–drug conjugate targeting FAP-expressing CAFs, induces tumor shrinkage and regression in NSCLC cancer models in combination with Pembrolizumab [233]. Photodynamic therapy suppressed tumor growth and improved therapeutic outcomes in preclinical models, which selectively eliminates FAP + CAFs by FAP-specific single chain variable fragments (scFv) and ferritin nanoparticles conjugated with photosensitizers [234,235,236,237]. Besides, a subgroup of CD10 + GPR77 + CAFs foster tumor growth and chemoresistance, while anti-GPR77 antibodies hinder tumor formation and restore chemosensitivity. Blocking GPR77 with an antibody disrupts the phosphorylation and acetylation of p65, a key component of NF-κB, thereby inhibiting its nuclear accumulation and activity. This leads to reduced secretion of inflammatory cytokines IL-6 and IL-8, chemoresistance, and CSC enrichment [153]. Recently, a subset of senescent CAFs in PDAC has been shown to exhibit increased expression of immune-regulatory genes, contributing to the inhibition of anti-tumor immune responses. Eliminating these senescent CAFs by ABT-199 (venetoclax),a BCL-2 inhibitor, increases activated CD8 + T cells in mouse tumors, while inducing CAF senescence had the opposite effect. Combining ABT-199 with immune checkpoint therapy significantly reduced tumor burden in mice, suggesting that elimination of senescent CAFs through senolytic treatment may be a promising approach to enhance immunotherapy [238, 239].

Normalization of CAFs to treat cancer

An alternative therapeutic strategy targeitng CAFs that is currently explored is to induce the transformation of tumor-promoting CAFs into quiescent fibroblasts, or into CAFs subpopulations that exert tumor-suppressing effects [240,241,242] (Fig. 6B). All-trans retinoic acid (ATRA), has been shown to reprogram activated PSCs into a quiescent state [243,244,245]. Furthermore, a nanosystem has been developed to co-deliver ATRA and siRNA targeting heat shock protein 47 (HSP47) to PSCs, simultaneously inducing quiescence and inhibiting extracellular matrix hyperplasia, which significantly enhances drug delivery and anti-tumour efficacy [246]. Minnelide, a derivative of triptolide reverse the activate CAFs to a quiescent, nonproliferative state [247]. This transformation disrupts the cross-talk between CAFs and tumor epithelial cells, leading to a downregulation of oncogenic signaling. Moreover, Minnelide has been shown to deplete reactive stromal fibroblasts and enhance drug delivery to pancreatic tumors, as evidenced in studies using both patient-derived xenografts and spontaneous pancreatic cancer mouse models [248]. Calcipotriol, a vitamin D receptor (VDR) ligand, effectively reprograms activated CAFs back to a quiescent state in PDAC, improving survival when combined with chemotherapy [249]. Scriptaid, a selective HDACs 1/3/8 inhibitor, reverses CAF differentiation, reducing ECM secretion, cell invasion, and tumor growth in vitro and in vivo, suggesting its potential as an anti-cancer strategy [250].

Treatment with IRAK4 inhibitor CA-494 increases the number of iCAFs with enhanced TNF signaling, inflammatory response, which turns ‘cold tumor’ refractory to immunotherapy to ‘hot tumor’ with increased immune cells infiltration [29]. Similarly, neutralizing TGFβ in vivo alters CAFs dynamics, reducing myoCAFs and promoting iCAFs, which correlates with enhanced anti-tumor immunity and PD-1 immunotherapy efficacy. This mechanism provides a rationale for TGFβ and PD-1 co-blockade in cancer treatment [168]. Fgl2 from tumor stroma accelerates lung cancer progression by activating CAFs and modulating MDSCs in a mouse study, suggesting its role as a therapeutic target [251]. In PKT mice, concurrent inhibition of MEK and STAT3 attenuated the iCAFs phenotype expressing Il6/Cxcl1 and myoCAFs phenotype expressing Lrrc15. While Ly6a/Cd34 + CAFs exhibited mesenchymal stem cell-like characteristics were enriched. The addition of MEKi + STAT3i to PD-1 blockade significantly improves antitumor responses and survival rates in PKT mice. Importantly, this combination achieved clinical benefits in a patient with chemotherapy-refractory metastatic PDAC [98]. The PDGFR inhibitor, Dasatinib, partially reversed the gene expression profile of CAFs, making it more similar to the phenotype of NFs. Specifically, 64 genes that were highly expressed in CAFs were downregulated, 26 genes that were lowly expressed in CAFs were upregulated, and it blocked the tumor-promoting effects of CAF-conditioned medium [252]. Recently, a study suggests that autophagy can promote CAFs into a quiescent state, with reduced secretion of IL-6 and upregulated expression of PD-L1 on tumor cells, thus enhancing the efficacy of ICIs. The combination of anti-PD-L1 and CQ-MSC-Lipo (a biomimetic drug delivery system carrying autophagy inhibitor chloroquine diphosphate) which specifically targets to CAFs effectively suppressed tumor growth [253]. Besides, Mazzeo et al. through ChIP-seq found that androgen receptor negatively regulates ANKRD1, which plays a negative regulatory role in the early stages of CAF activation. Treatment of CAFs with ANKRD1-targeting antisense oligonucleotides (ASOs) led to the simultaneous downregulation of ANKRD1 and key CAFs effector genes such as ACTA2, COL1A1, INHBA, and HAS2. This significantly reduced the proliferation of co-cultured SCC cancer cells, and corresponding in vivo validation was also obtained in mouse models. Therefore, targeting ANKRD1 with ASOs could be a feasible approach to restoring CAF activation and inhibiting tumor progression [254].

Future Perspectives

With the rapid development of cutting-edge technologies such as scRNA-seq, ST, spatial pathology analysis, and organoid models, the complex interactions of CAFs within the TME are gradually being unveiled [82, 89, 93, 157, 221, 255, 256]. These cutting-edge technologies allow for a comprehensive understanding of the diversity of CAFs as well as their functions, both at the single-cell level as well as the spatial levels. The advent of scRNA-seq has revolutionized our understanding of the remarkable heterogeneity, plasticity, and context-dependent roles for CAFs across tumor types. However, while single-cell technologies excel at cataloging cellular diversity, they inherently lack spatial context, and this limitation affects our ability to map CAF interactions within the architectural and functional niches of the TME [257]. One prediction is that emerging technologies that can clarify the spatial pathology of tumors will be an important new frontier for CAF research, and this challenge could be addressed using spatial multi-omics technologies integrated with artificial intelligence-driven tools [258, 259]. As an interesting example, Onder et al. recently revealed the physical connections between multiple TLSs and intratumoral T-cell tracks derived from CCL19 + CAFs in NSCLC. These connections were supported by CCL19⁺ CAFs that formed a three-dimensional (3D) network spanning tumor tissues which guided T-cell migration via CCL19 chemokine gradients, promoting the distribution and activation of intratumoral T cells. In a murine model, a coronavirus-vectored vaccine (mCOV-Flt3l-gp33) was used to induce CCL19⁺ CAFs, which increased TLS formation and amplified anti-tumor immunity through interactions with CD8⁺ T cells. The discovery of this 3D network has provided novel insights into the tumor immune microenvironment, and these findings lay the groundwork for developing fibroblast-based immunotherapeutic strategies in the future [260].

The clinical translation of CAF-related discoveries remains a critical unmet need in oncology. Despite robust preclinical evidence implicating CAF subsets in immune evasion, metabolic symbiosis, and therapeutic resistance, few CAF-targeted therapies have advanced to late-stage clinical trials. Key challenges include the functional plasticity of CAFs (e.g., tumor-restrictive vs. tumor-promoting subsets), biomarker paucity to stratify patients for stromal modulation, and off-target effects of broad stromal depletion. To bridge this gap, interdisciplinary efforts integrating spatial multi-omics, AI-driven digital pathology, and functional organoid models could be mobilized to enhance efforts in this area of exploration. Early successes, such as the development of TAK1 inhibitors to reprogram IL-6-driven CAFs, underscore the potential of mechanistically informed strategies [157]. Concurrently, clinical trials must adopt composite endpoints that evaluate both stromal reprogramming (e.g., ECM normalization, immune infiltration) and traditional oncologic outcomes. By prioritizing translational pipelines that reconcile CAFs heterogeneity with patient-specific TME dynamics, we can unlock the potential for CAFs-targeted therapies to be developed as next-generation combination regimens that are effective for cancer.

Conclusions

CAFs have emerged as pivotal players in the TME, exerting multifaceted influences on tumor progression, immune modulation, angiogenesis, and therapeutic response. In this review, we have showcased the most recent advances regarding the roles of CAFs in cancer development, as well as therapies that target CAFs. Indeed, therapeutic targeting of CAFs encompasses multiple approaches, including targeting the ECM generated by CAFs, directly eliminating CAF subtypes identified by their expression of key markers, interrupting the cross-talk between CAFs and the TME, and through the normalization of CAFs. While preclinical models have demonstrated promising anti-tumor effects of CAF-targeted treatments, large-scale Phase III clinical trials are still needed to validate these findings.

While this review comprehensively synthesizes the current understanding of CAFs in cancer biology, several limitations warrant acknowledgment by the field, such as the current lack of reliable, universal markers to define CAF subsets, discrepancies in findings and tumor progression between animal models and human tumors, technological gaps, as well as interpretation of preclinical observations [25, 89, 257]. The ability to effectively address such limitations requires interdisciplinary collaboration, leveraging advanced spatial technologies, functional genomics, and biomarker-driven clinical trials. Nevertheless, this review demonstrates that CAFs are an important therapeutic target in cancer treatment. A deeper understanding of their roles and their signaling mechanisms in cancer will facilitate the development of more effective and targeted therapies that exploit CAFs, and move this field of research closer towards the promise of novel cancer treatments.

Availability of data and materials

No datasets were generated or analysed during the current study.

Abbreviations

Abca8a:

ATP-binding cassette subfamily A member 8a

ACTA2:

Actin alpha 2, smooth muscle

ADFs:

Adipose-derived fibroblasts

AKAP12:

A-kinase anchor protein 12

AKT:

Protein kinase B

ALOX15:

Arachidonate 15-lipoxygenase

ANG-2:

Angiopoietin-2

ANKRD1:

Ankyrin repeat domain 1

ANXA2:

Annexin A2

apCAFs:

Antigen-presenting cancer-associated fibroblasts

ASOs:

Antisense oligonucleotides

ATRA:

All-trans retinoic acid

B7H3:

B7 homolog 3

BC:

Breast cancer

BMP2:

Bone morphogenetic protein 2

CAFs:

Cancer-associated fibroblasts

cCAFs:

Cycling cancer-associated fibroblasts

CCL19:

C–C motif chemokine ligand 19

CHI3L1:

Chitinase-3-like protein 1

CLCF1:

Cardiotrophin-like cytokine factor 1

CLIC1:

Chloride intracellular channel 1

CM:

Conditioned medium

COL10A1:

Collagen type X alpha 1 chain

COL11A1:

Collagen type XI alpha 1 chain

COL14A1:

Collagen type XIV alpha 1 chain

COL4A1:

Collagen type IV alpha 1 chain

CRABP2:

Cellular retinoic acid-binding protein 2

CRC:

Colorectal cancer

CSCs:

Cancer stem cells

CSF1:

Colony-stimulating factor 1

CXCL12:

C-X-C motif chemokine ligand 12

CXCL3:

C-X-C motif chemokine ligand 3

CXCL9:

C-X-C motif chemokine ligand 9

CXCR4:

C-X-C chemokine receptor type 4

dCAFs:

Developmental cancer-associated fibroblasts

DCIS:

Ductal carcinoma in situ

DKK1:

Dickkopf-related protein 1

DPP4:

Dipeptidyl peptidase-4

ECM:

Extracellular matrix

ECM-CAFs:

Extracellular matrix-associated cancer-associated fibroblasts

EPC:

Endothelial progenitor cell

ERK:

Extracellular signal-regulated kinase

EVs:

Extracellular vesicles

FAK:

Focal adhesion kinase

FAP:

Fibroblast activation protein

FGF2:

Fibroblast growth factor 2

Foxp3:

Forkhead box protein P3

FSP1:

Fibroblast-specific protein 1

GC:

Gastric cancer

G-CSF:

Granulocyte colony-stimulating factor

GEMMs:

Genetically engineered mouse models

GM-CSF:

Granulocyte–macrophage colony-stimulating factor

GP130-IL6ST:

Glycoprotein 130-Interleukin 6 signal transducer

GPR77:

G protein-coupled receptor 77

GTPase:

Guanosine triphosphatase

HCC:

Hepatocellular carcinoma

HDAC2:

Histone deacetylase 2

HER2:

Human epidermal growth factor receptor 2

HGF:

Hepatocyte growth factor

iCAFs:

Inflammatory cancer-associated fibroblasts

ICC:

Intrahepatic cholangiocarcinoma

IGF-1:

Insulin-like growth factor 1

IL-1:

Interleukin-1

IL-33:

Interleukin-33

IL-6:

Interleukin-6

IRAK4:

Interleukin-1 receptor-associated kinase 4

ITGA11:

Integrin subunit alpha 11

ITGB1:

Integrin subunit beta 1

JAK:

Janus kinase

JAM:

Junctional adhesion molecule

KPC:

Kras; Trp53; Pdx-Cre (pancreatic cancer model)

Lepr:

Leptin receptor

LIF:

Leukemia inhibitory factor

LncRNAs:

Long non-coding RNAs

LOXL2:

Lysyl oxidase-like 2

LRRC15:

Leucine-rich repeat-containing protein 15

Lym-CAFs:

Lymphoid cancer-associated fibroblasts

mCAFs:

Matrix cancer-associated fibroblasts

MCAM:

Melanoma cell adhesion molecule

meCAFs:

Metabolic cancer-associated fibroblasts

MEF2C:

Myocyte enhancer factor 2C

MMPs:

Matrix metalloproteinases

MNAM:

Methylnicotinamide

MYH11:

Myosin heavy chain 11

myoCAFs:

Myofibroblastic cancer-associated fibroblasts

NAM:

Nicotinamide

NAMPT:

Nicotinamide phosphoribosyltransferase

NETs:

Neutrophil extracellular traps

NFs:

Normal fibroblasts

NF-κB:

Nuclear factor kappa-light-chain-enhancer of activated B cells

NNMT:

Nicotinamide N-methyltransferase

NSCLC:

Non-small cell lung cancer

ORR:

Objective response rate

OS:

Overall survival

OX40L:

OX40 ligand

PD-1/PD-L1:

Programmed cell death protein 1/Programmed death-ligand 1

PDGF-C:

Platelet-derived growth factor C

PDGFR:

Platelet-derived growth factor receptor

PDPN:

Podoplanin

PFS:

Progression-free survival

PGF:

Placental growth factor

PI3K:

Phosphoinositide 3-kinase

PLA2G2A:

Phospholipase A2 group IIA

PlGF:

Placental growth factor

PMN-MDSCs:

Polymorphonuclear myeloid-derived suppressor cells

RhoGAP:

Rho GTPase-activating protein

RGS5:

Regulator of G-protein signaling 5

ROCK:

Rho-associated protein kinase

ROS:

Reactive oxygen species

SAM:

S-adenosylmethionine

scRNA-seq:

Single-cell RNA sequencing

SETD2:

SET domain containing 2, histone lysine methyltransferase

SPP1:

Secreted Phosphoprotein 1(also named Osteopontin)

SR-A/CD204:

Scavenger receptor A/CD204

ST:

Spatial transcriptomics

STAT3:

Signal transducer and activator of transcription 3

TAK1:

Transforming growth factor-beta-activated kinase 1

TAZ:

Transcriptional co-activator with PDZ-binding motif

TGF-β:

Transforming growth factor-beta

THBS2:

Thrombospondin 2

TLSs:

Tertiary lymphoid structures

TME:

Tumor microenvironment

vCAFs:

Vascular cancer-associated fibroblasts

VDR:

Vitamin D receptor

VEGFA:

Vascular endothelial growth factor A

WNT2:

Wnt family member 2

YAP1:

Yes-associated protein 1

α-SMA:

Alpha-smooth muscle actin

References

  1. Han L, Wu Y, Fang K, Sweeney S, Roesner U, Parrish M, et al. The splanchnic mesenchyme is the tissue of origin for pancreatic fibroblasts during homeostasis and tumorigenesis. Nat Commun. 2023;14:1.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Dobaczewski M, de Haan J, Frangogiannis N. The extracellular matrix modulates fibroblast phenotype and function in the infarcted myocardium. J Cardiovasc Transl Res. 2012;5:837–47.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Younesi FS, Miller AE, Barker TH, Rossi FMV, Hinz B. Fibroblast and myofibroblast activation in normal tissue repair and fibrosis. Nat Rev Mol Cell Biol. 2024;25:617–38.

    Article  CAS  PubMed  Google Scholar 

  4. McAndrews KM, Miyake T, Ehsanipour EA, Kelly PJ, Becker LM, McGrail DJ, et al. Dermal α SMA+ myofibroblasts orchestrate skin wound repair via β 1 integrin and independent of type i collagen production. EMBO J. 2022;41: e109470.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hughes CC. Endothelial-stromal interactions in angiogenesis. Curr Opin Hematol. 2008;15:204–9.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Odell AF, Mannion AJ. In vitro co-culture of fibroblast and endothelial cells to assess angiogenesis. Methods Mol Biol. 2022;2441:277–86.

    Article  CAS  PubMed  Google Scholar 

  7. Hetheridge C, Mavria G, Mellor H. Uses of the in vitro endothelial-fibroblast organotypic co-culture assay in angiogenesis research. Biochem Soc Trans. 2011;39:1597–600.

    Article  CAS  PubMed  Google Scholar 

  8. Jordana M, Sarnstrand B, Sime P, Ramis I. Immune-inflammatory functions of fibroblasts. Eur Respir J. 1994;7:2212–22.

    Article  CAS  PubMed  Google Scholar 

  9. Correa-Gallegos D, Jiang D, Rinkevich Y. Fibroblasts as confederates of the immune system. Immunol Rev. 2021;302:147–62.

    Article  CAS  PubMed  Google Scholar 

  10. Buechler MB, Turley SJ. A short field guide to fibroblast function in immunity. Semin Immunol. 2018;35:48–58.

    Article  CAS  PubMed  Google Scholar 

  11. Franco OE, Shaw AK, Strand DW, Hayward SW. Cancer associated fibroblasts in cancer pathogenesis. 2010;21:33–9

  12. Sano M, Ijichi H, Takahashi R, Miyabayashi K, Fujiwara H, Yamada T, et al. Blocking CXCLs–CXCR2 axis in tumor–stromal interactions contributes to survival in a mouse model of pancreatic ductal adenocarcinoma through reduced cell invasion/migration and a shift of immune-inflammatory microenvironment. Oncogenesis. 2019;8:8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 2020;20:174–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Shiga K, Hara M, Nagasaki T, Sato T, Takahashi H, Takeyama H. Cancer-associated fibroblasts: their characteristics and their roles in tumor growth. Cancers. 2015;7:2443–58.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Kahounová Z, Kurfürstová D, Bouchal J, Kharaishvili G, Navrátil J, Remšı́k J, et al. The fibroblast surface markers FAP, anti-fibroblast, and FSP are expressed by cells of epithelial origin and may be altered during epithelial-to-mesenchymal transition. Cytometry A. 2018;93:941–51.

    Article  PubMed  Google Scholar 

  16. Sil S, Periyasamy P, Thangaraj A, Chivero ET, Buch S. PDGF/PDGFR axis in the neural systems. Mol Aspects Med. 2018;62:63–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Groot ACG, DeRuiter MC, Bergwerff M, Poelmann RE. Smooth muscle cell origin and its relation to heterogeneity in development and disease. Arterioscler Thromb Vasc Biol. 1999;19:1589–94.

    Article  Google Scholar 

  18. Smyth LC, Rustenhoven J, Scotter EL, Schweder P, Faull RL, Park TI, et al. Markers for human brain pericytes and smooth muscle cells. J Chem Neuroanat. 2018;92:48–60.

    Article  CAS  PubMed  Google Scholar 

  19. Astarita JL, Acton SE, Turley SJ. Podoplanin: Emerging functions in development, the immune system, and cancer. Front Immunol. 2012;3:283.

    Article  PubMed  PubMed Central  Google Scholar 

  20. 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.

    Article  CAS  PubMed  Google Scholar 

  21. Zhang X, Dong M, Pan Y, Chen J, Huang X, Jin Y, et al. Expression pattern of cancer-associated fibroblast and its clinical relevance in intrahepatic cholangiocarcinoma. Hum Pathol. 2017;65:92–100.

    Article  CAS  PubMed  Google Scholar 

  22. Ha SY, Yeo S-Y, Xuan Y, Kim S-H. The prognostic significance of cancer-associated fibroblasts in esophageal squamous cell carcinoma. PLoS ONE. 2014;9: e99955.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Venning FA, Zornhagen KW, Wullkopf L, Sjölund J, Rodriguez-Cupello C, Kjellman P, et al. Deciphering the temporal heterogeneity of cancer-associated fibroblast subpopulations in breast cancer. J Exp Clin Cancer Res. 2021;40:175.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bartoschek M, Oskolkov N, Bocci M, Lövrot J, Larsson C, Sommarin M, et al. Spatially and functionally distinct subclasses of breast cancer-associated fibroblasts revealed by single cell RNA sequencing. Nat Commun. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-018-07582-3.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Cords L, de Souza N, Bodenmiller B. Classifying cancer-associated fibroblasts—the good, the bad, and the target. Cancer Cell. 2024;42:1480–5.

    Article  CAS  PubMed  Google Scholar 

  26. Thorlacius-Ussing J, Jensen C, Nissen NI, Cox TR, Kalluri R, Karsdal M, et al. The collagen landscape in cancer profiling collagens in tumors and in circulation reveals novel markers of cancer-associated fibroblast subtypes. J Pathol. 2024;262:22–36.

    Article  CAS  PubMed  Google Scholar 

  27. Xue M, Zhu Y, Jiang Y, Han L, Shi M, Su R, et al. Schwann cells regulate tumor cells and cancer-associated fibroblasts in the pancreatic ductal adenocarcinoma microenvironment. Nat Commun. 2023;14:4600.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Steele NG, Biffi G, Kemp SB, Zhang Y, Drouillard D, Syu L, et al. Inhibition of hedgehog signaling alters fibroblast composition in pancreatic cancer. Clin Cancer Res. 2021;27:2023–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Somani V, Zhang D, Dodhiawala PB, Lander VE, Liu X, Kang L-I, et al. IRAK4 signaling drives resistance to checkpoint immunotherapy in pancreatic ductal adenocarcinoma. Gastroenterology. 2022;162:2047–62.

    Article  CAS  PubMed  Google Scholar 

  30. Elyada E, Bolisetty M, Laise P, Flynn WF, Courtois ET, Burkhart RA, et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 2019;9:1102–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhu Q, Zhang X, Zhang L, Li W, Wu H, Yuan X, et al. The IL-6–STAT3 axis mediates a reciprocal crosstalk between cancer-derived mesenchymal stem cells and neutrophils to synergistically prompt gastric cancer progression. Cell Death Dis. 2014;5:e1295–305.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bochet L, Lehuédé C, Dauvillier S, Wang YY, Dirat B, Laurent V, et al. Adipocyte-derived fibroblasts promote tumor progression and contribute to the desmoplastic reaction in breast cancer. Can Res. 2013;73:5657–68.

    Article  CAS  Google Scholar 

  33. Somerville TD, Biffi G, Daßler-Plenker J, Hur SK, He X-Y, Vance KE, et al. Squamous trans-differentiation of pancreatic cancer cells promotes stromal inflammation. Elife. 2020;9: e53381.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Giannoni E, Bianchini F, Masieri L, Serni S, Torre E, Calorini L, et al. Reciprocal activation of prostate cancer cells and cancer-associated fibroblasts stimulates epithelial-mesenchymal transition and cancer stemness. Can Res. 2010;70:6945–56.

    Article  CAS  Google Scholar 

  35. Becker LM, O’Connell JT, Vo AP, Cain MP, Tampe D, Bizarro L, et al. Epigenetic reprogramming of cancer-associated fibroblasts deregulates glucose metabolism and facilitates progression of breast cancer. Cell Rep. 2020;31:107701.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Krishnamurty AT, Shyer JA, Thai M, Gandham V, Buechler MB, Yang YA, et al. LRRC15+ myofibroblasts dictate the stromal setpoint to suppress tumour immunity. Nature. 2022;611:148–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Goulet CR, Champagne A, Bernard G, Vandal D, Chabaud S, Pouliot F, et al. Cancer-associated fibroblasts induce epithelial–mesenchymal transition of bladder cancer cells through paracrine IL-6 signalling. BMC Cancer. 2019;19:1–13.

    Article  Google Scholar 

  38. Ringuette Goulet C, Bernard G, Tremblay S, Chabaud S, Bolduc S, Pouliot F. Exosomes induce fibroblast differentiation into cancer-associated fibroblasts through TGF β signaling. Mol Cancer Res. 2018;16:1196–204.

    Article  CAS  PubMed  Google Scholar 

  39. Kojima Y, Acar A, Eaton EN, Mellody KT, Scheel C, Ben-Porath I, et al. Autocrine TGF-β and stromal cell-derived factor-1 (SDF-1) signaling drives the evolution of tumor-promoting mammary stromal myofibroblasts. Proc Natl Acad Sci. 2010;107:20009–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kobayashi H, Gieniec KA, Lannagan TR, Wang T, Asai N, Mizutani Y, et al. The origin and contribution of cancer-associated fibroblasts in colorectal carcinogenesis. Gastroenterology. 2022;162:890–906.

    Article  CAS  PubMed  Google Scholar 

  41. Erez N, Truitt M, Olson P. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kB-dependent manner. Cancer Cell. 2010;17:135–47.

    Article  CAS  PubMed  Google Scholar 

  42. Öhlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med. 2017;214:579–96.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Huang H, Wang Z, Zhang Y, Pradhan RN, Ganguly D, Chandra R, et al. Mesothelial cell-derived antigen-presenting cancer-associated fibroblasts induce expansion of regulatory t cells in pancreatic cancer. Cancer Cell. 2022;40:656–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Barcellos-de-Souza P, Comito G, Pons-Segura C, Taddei ML, Gori V, Becherucci V, et al. Mesenchymal stem cells are recruited and activated into carcinoma-associated fibroblasts by prostate cancer microenvironment-derived TGF-β 1. Stem cells. 2016;34:2536–47.

    Article  CAS  PubMed  Google Scholar 

  45. Eckert MA, Coscia F, Chryplewicz A, Chang JW, Hernandez KM, Pan S, et al. Proteomics reveals NNMT as a master metabolic regulator of cancer-associated fibroblasts. Nature. 2019;569:723–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Yang X, Lin Y, Shi Y, Li B, Liu W, Yin W, et al. FAP promotes immunosuppression by cancer-associated fibroblasts in the tumor microenvironment via STAT3-CCL2 signaling. Can Res. 2016;76:4124–35.

    Article  CAS  Google Scholar 

  47. Li L, Wei J-R, Dong J, Lin Q-G, Tang H, Jia Y-X, et al. Laminin γ 2–mediating t cell exclusion attenuates response to anti–PD-1 therapy. Sci Adv. 2021;7:eabc8346.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Kühn R, M. Torres R. Cre/loxP recombination system and gene targeting. transgenesis techniques: principles and protocols. 2002;175–204.

  49. Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, Jacobetz MA, et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell. 2003;4:437–50.

    Article  CAS  PubMed  Google Scholar 

  50. Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005;7:469–83.

    Article  CAS  PubMed  Google Scholar 

  51. Lee JW, Komar CA, Bengsch F, Graham K, Beatty GL. Genetically engineered mouse models of pancreatic cancer: The KPC model (LSL-KrasG12D/+; LSL-Trp53R172H/+; pdx-1-cre), its variants, and their application in immuno-oncology drug discovery. Curr Protoc Pharmacol. 2016;73:14–39.

    Article  PubMed Central  Google Scholar 

  52. Garcia Garcia CJ, Huang Y, Fuentes NR, Turner MC, Monberg ME, Lin D, et al. Stromal HIF2 regulates immune suppression in the pancreatic cancer microenvironment. Gastroenterology. 2022;162:2018–31.

    Article  CAS  PubMed  Google Scholar 

  53. Saito M, Iwawaki T, Taya C, Yonekawa H, Noda M, Inui Y, et al. Diphtheria toxin receptor–mediated conditional and targeted cell ablation in transgenic mice. Nat Biotechnol. 2001;19:746–50.

    Article  CAS  PubMed  Google Scholar 

  54. Ravi M, Paramesh V, Kaviya S, Anuradha E, Solomon FP. 3D cell culture systems: advantages and applications. J Cell Physiol. 2015;230:16–26.

    Article  CAS  PubMed  Google Scholar 

  55. Drost J, Clevers H. Organoids in cancer research. Nat Rev Cancer. 2018;18:407–18.

    Article  CAS  PubMed  Google Scholar 

  56. Veninga V, Voest EE. Tumor organoids: opportunities and challenges to guide precision medicine. Cancer Cell. 2021;39:1190–201.

    Article  CAS  PubMed  Google Scholar 

  57. Osuala KO, Sameni M, Shah S, Aggarwal N, Simonait ML, Franco OE, et al. Il-6 signaling between ductal carcinoma in situ cells and carcinoma-associated fibroblasts mediates tumor cell growth and migration. BMC Cancer. 2015;15:1–15.

    Article  CAS  Google Scholar 

  58. Schuth S, Le Blanc S, Krieger TG, Jabs J, Schenk M, Giese NA, et al. Patient-specific modeling of stroma-mediated chemoresistance of pancreatic cancer using a three-dimensional organoid-fibroblast co-culture system. J Exp Clin Cancer Res. 2022;41:312.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Hu M, Yao J, Carroll DK, Weremowicz S, Chen H, Carrasco D, et al. Regulation of in situ to invasive breast carcinoma transition. Cancer Cell. 2008;13:394–406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Goetz JG, Minguet S, Navarro-Lérida I, Lazcano JJ, Samaniego R, Calvo E, et al. Biomechanical remodeling of the microenvironment by stromal caveolin-1 favors tumor invasion and metastasis. Cell. 2011;146:148–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Di Modugno F, Colosi C, Trono P, Antonacci G, Ruocco G, Nisticò P. 3D models in the new era of immune oncology: focus on t cells, CAF and ECM. J Exp Clin Cancer Res. 2019;38:1–14.

    Article  Google Scholar 

  62. 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.

    Article  CAS  PubMed  Google Scholar 

  63. Winkler J, Abisoye-Ogunniyan A, Metcalf KJ, Werb Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat Commun. 2020;11:5120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Chiappori AA, Eckhardt SG, Bukowski R, Sullivan DM, Ikeda M, Yano Y, et al. A phase i pharmacokinetic and pharmacodynamic study of s-3304, a novel matrix metalloproteinase inhibitor, in patients with advanced and refractory solid tumors. Clin Cancer Res. 2007;13:2091–9.

    Article  CAS  PubMed  Google Scholar 

  65. Gaggioli C, Hooper S, Hidalgo-Carcedo C, Grosse R, Marshall JF, Harrington K, et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat Cell Biol. 2007;9:1392–400.

    Article  CAS  PubMed  Google Scholar 

  66. Ketteler J, Klein D. Caveolin-1, cancer and therapy resistance. Int J Cancer. 2018;143:2092–104.

    Article  CAS  PubMed  Google Scholar 

  67. Sanz-Moreno V, Gaggioli C, Yeo M, Albrengues J, Wallberg F, Viros A, et al. ROCK and JAK1 signaling cooperate to control actomyosin contractility in tumor cells and stroma. Cancer Cell. 2011;20:229–45.

    Article  CAS  PubMed  Google Scholar 

  68. Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, et al. Extracellular matrix remodeling in tumor progression and immune escape: From mechanisms to treatments. Mol Cancer. 2023;22:48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Shen Z, Tao L, Wang Y, Zhu Y, Pan H, Li Y, et al. Synergistic anticancer strategy targeting ECM stiffness: integration of matrix softening and mechanical signal transduction blockade in primary liver cancers. Adv Sci. 2025;12:2403040.

    Article  CAS  Google Scholar 

  70. Agrawal A, Javanmardi Y, Watson SA, Serwinski B, Djordjevic B, Li W, et al. Mechanical signatures in cancer metastasis. npj Biol Phys Mech. 2025;2:3.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Mai Z, Lin Y, Lin P, Zhao X, Cui L. Modulating extracellular matrix stiffness: a strategic approach to boost cancer immunotherapy. Cell Death Dis. 2024;15:307.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Najafi M, Farhood B, Mortezaee K. Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J Cell Biochem. 2019;120:2782–90.

    Article  CAS  PubMed  Google Scholar 

  73. Wang K, Ning S, Zhang S, Jiang M, Huang Y, Pei H, et al. Extracellular matrix stiffness regulates colorectal cancer progression via HSF4. J Exp Clin Cancer Res. 2025;44:30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Zheng J-H, Zhu Y-H, Yang J, Ji P-X, Zhao R-K, Duan Z-H, et al. A CLIC1 network coordinates matrix stiffness and the warburg effect to promote tumor growth in pancreatic cancer. Cell Reports. 2024;43.

  75. Maiques O, Sallan MC, Laddach R, Pandya P, Varela A, Crosas-Molist E, et al. Matrix mechano-sensing at the invasive front induces a cytoskeletal and transcriptional memory supporting metastasis. Nat Commun. 2025;16:1394.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Zheng H, An M, Luo Y, Diao X, Zhong W, Pang M, et al. PDGFR α+ ITGA11+ fibroblasts foster early-stage cancer lymphovascular invasion and lymphatic metastasis via ITGA11-SELE interplay. Cancer Cell. 2024;42:682–700.

    Article  CAS  PubMed  Google Scholar 

  77. Santos AM, Jung J, Aziz N, Kissil JL, Puré E, et al. Targeting fibroblast activation protein inhibits tumor stromagenesis and growth in mice. J Clin Invest. 2009;119:3613–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Jiang H, Torphy RJ, Steiger K, Hongo H, Ritchie AJ, Kriegsmann M, et al. Pancreatic ductal adenocarcinoma progression is restrained by stromal matrix. J Clin Investig. 2020;130:4704–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Wang Y, Liang Y, Xu H, Zhang X, Mao T, Cui J, et al. Single-cell analysis of pancreatic ductal adenocarcinoma identifies a novel fibroblast subtype associated with poor prognosis but better immunotherapy response. Cell Discov. 2021;7:36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Neesse A, Frese KK, Bapiro TE, Nakagawa T, Sternlicht MD, Seeley TW, et al. CTGF antagonism with mAb FG-3019 enhances chemotherapy response without increasing drug delivery in murine ductal pancreas cancer. Proc Natl Acad Sci. 2013;110:12325–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. LeSavage BL, Zhang D, Huerta-López C, Gilchrist AE, Krajina BA, Karlsson K, et al. Engineered matrices reveal stiffness-mediated chemoresistance in patient-derived pancreatic cancer organoids. Nat Mater. 2024;23:1138–49.

    Article  CAS  PubMed  Google Scholar 

  83. Davidson S, Coles M, Thomas T, Kollias G, Ludewig B, Turley S, et al. Fibroblasts as immune regulators in infection, inflammation and cancer. Nat Rev Immunol. 2021;21:704–17.

    Article  CAS  PubMed  Google Scholar 

  84. Saw PE, Chen J, Song E. Targeting CAFs to overcome anticancer therapeutic resistance. Trends Cancer. 2022;8:527–55.

    Article  CAS  PubMed  Google Scholar 

  85. Desbois M, Wang Y. Cancer-associated fibroblasts: Key players in shaping the tumor immune microenvironment. Immunol Rev. 2021;302:241–58.

    Article  CAS  PubMed  Google Scholar 

  86. Kim DK, Jeong J, Lee DS, Hyeon DY, Park GW, Jeon S, et al. PD-L1-directed PlGF/VEGF blockade synergizes with chemotherapy by targeting CD141+ cancer-associated fibroblasts in pancreatic cancer. Nat Commun. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-022-33991-6.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Feig C, Jones JO, Kraman M, Wells RJB, Deonarine A, Chan DS, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. PNAS. 2013;1:20212.

    Article  Google Scholar 

  88. Kato TOT, Noma K. Cancer-associated fibroblasts affect intratumoral CD8+ and FoxP3+ t cells via IL6 in the tumor microenvironment. Clin Cancer Res. 2018;24:4820.

    Article  CAS  PubMed  Google Scholar 

  89. Song D, Wu Y, Li J, Liu J, Yi Z, Wang X, et al. Insulin-like growth factor 2 drives fibroblast-mediated tumor immunoevasion and confers resistance to immunotherapy. J Clin Invest. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI183366.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Özdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu C-C, Simpson TR, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25:719–34.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Neuzillet C, Tijeras-Raballand A, Ragulan C, Cros J, Patil Y, Martinet M, et al. Inter- and intra-tumoural heterogeneity in cancer-associated fibroblasts of human pancreatic ductal adenocarcinoma. J Pathol. 2019;248:51–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Gupta YH, Khanom A, Acton SE. Control of dendritic cell function within the tumour microenvironment. Front Immunol. 2022;13: 733800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Fan G, Gao R, Xie T, Li L, Tang L, Han X, et al. DKK1+ tumor cells inhibited the infiltration of CCL19+ fibroblasts and plasma cells contributing to worse immunotherapy response in hepatocellular carcinoma. Cell Death Dis. 2024;15:797.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Li X, Bu W, Meng L, Liu X, Wang S, Jiang L, et al. CXCL12/CXCR4 pathway orchestrates CSC-like properties by CAF recruited tumor associated macrophage in OSCC. Exp Cell Res. 2019;378:131–8.

    Article  CAS  PubMed  Google Scholar 

  95. Cho H, Seo Y, Loke KM, Kim S-W, Oh S-M, Kim J-H, et al. Cancer-stimulated CAFs enhance monocyte differentiation and protumoral TAM activation via IL6 and GM-CSF secretion. Clin Cancer Res. 2018;24:5407–21.

    Article  CAS  PubMed  Google Scholar 

  96. Cohen N, Shani O, Raz Y, Sharon Y, Hoffman D, Abramovitz L, et al. Fibroblasts drive an immunosuppressive and growth-promoting microenvironment in breast cancer via secretion of chitinase 3-like 1. Oncogene. 2017;36:4457–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Andersson P, Yang Y, Hosaka K, Zhang Y, Fischer C, Braun H, et al. Molecular mechanisms of IL-33–mediated stromal interactions in cancer metastasis. JCI Insight. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci.insight.122375.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Datta J, Dai X, Bianchi A, De Castro SI, Mehra S, Garrido VT, et al. Combined MEK and STAT3 inhibition uncovers stromal plasticity by enriching for cancer-associated fibroblasts with mesenchymal stem cell-like features to overcome immunotherapy resistance in pancreatic cancer. Gastroenterology. 2022;163:1593–612.

    Article  CAS  PubMed  Google Scholar 

  99. Yeh C-R, Slavin S, Da J, Hsu I, Luo J, Xiao G-Q, et al. Estrogen receptor α in cancer associated fibroblasts suppresses prostate cancer invasion via reducing CCL5, IL6 and macrophage infiltration in the tumor microenvironment. Mol Cancer. 2016;15:1–14.

    Article  Google Scholar 

  100. Mazur A, Holthoff E, Vadali S, Kelly T, Post SR. Cleavage of type i collagen by fibroblast activation protein-α enhances class a scavenger receptor mediated macrophage adhesion. PLoS ONE. 2016;11: e0150287.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Timperi E, Gueguen P, Molgora M, Magagna I, Kieffer Y, Lopez-Lastra S, et al. Lipid-associated macrophages are induced by cancer-associated fibroblasts and mediate immune suppression in breast cancer. Can Res. 2022;82:3291–306.

    Article  CAS  Google Scholar 

  102. Wang H, Liang Y, Liu Z, Zhang R, Chao J, Wang M, et al. POSTN+ cancer-associated fibroblasts determine the efficacy of immunotherapy in hepatocellular carcinoma. J Immunother Cancer. 2024;12: e008721.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Li Y, Zheng Y, Huang J, Nie R-C, Wu Q-N, Zuo Z, et al. CAF-macrophage crosstalk in tumour microenvironments governs the response to immune checkpoint blockade in gastric cancer peritoneal metastases. Gut. 2025;74:350–63.

    Article  CAS  PubMed  Google Scholar 

  104. Sun X, He X, Zhang Y, Hosaka K, Andersson P, Wu J, et al. Inflammatory cell-derived CXCL3 promotes pancreatic cancer metastasis through a novel myofibroblast-hijacked cancer escape mechanism. Gut. 2022;71:129–47.

    Article  CAS  PubMed  Google Scholar 

  105. Comito G, Giannoni E, Segura C, Barcellos-de-Souza P, Raspollini M, Baroni G, et al. Cancer-associated fibroblasts and M2-polarized macrophages synergize during prostate carcinoma progression. Oncogene. 2014;33:2423–31.

    Article  CAS  PubMed  Google Scholar 

  106. Cheng Y, Li H, Deng Y, Tai Y, Zeng K, Zhang Y, et al. Cancer-associated fibroblasts induce PDL1+ neutrophils through the IL6-STAT3 pathway that foster immune suppression in hepatocellular carcinoma. Cell Death Dis. 2018;9:422.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Song M, He J, Pan Q-Z, Yang J, Zhao J, Zhang Y-J, et al. Cancer-associated fibroblast-mediated cellular crosstalk supports hepatocellular carcinoma progression. Hepatology. 2021;73:1717–35.

    Article  CAS  PubMed  Google Scholar 

  108. Munir H, Jones JO, Janowitz T, Hoffmann M, Euler M, Martins CP, et al. Stromal-driven and amyloid β-dependent induction of neutrophil extracellular traps modulates tumor growth. Nat Commun. 2021;12:683.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Takesue S, Ohuchida K, Shinkawa T, Otsubo Y, Matsumoto S, Sagara A, et al. Neutrophil extracellular traps promote liver micrometastasis in pancreatic ductal adenocarcinoma via the activation of cancer-associated fibroblasts. Int J Oncol. 2019;56:596–605.

    PubMed  Google Scholar 

  110. 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.

    Article  CAS  PubMed  Google Scholar 

  111. Allaoui R, Bergenfelz C, Mohlin S, Hagerling C, Salari K, Werb Z, et al. Cancer-associated fibroblast-secreted CXCL16 attracts monocytes to promote stroma activation in triple-negative breast cancers. Nat Commun. 2016;7:13050.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Gabrilovich DI. Myeloid-derived suppressor cells. Cancer Immunol Res. 2017;5:3–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Mandruzzato S, Brandau S, Britten CM, Bronte V, Damuzzo V, Gouttefangeas C, et al. Toward harmonized phenotyping of human myeloid-derived suppressor cells by flow cytometry: Results from an interim study. Cancer Immunol Immunother. 2016;65:161–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Kumar V, Donthireddy L, Marvel D, Condamine T, Wang F, Lavilla-Alonso S, et al. Cancer-associated fibroblasts neutralize the anti-tumor effect of CSF1 receptor blockade by inducing PMN-MDSC infiltration of tumors. Cancer Cell. 2017;32:654–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Zou X, Tang X-Y, Qu Z-Y, Sun Z-W, Ji C-F, Li Y-J, et al. Targeting the PDGF/PDGFR signaling pathway for cancer therapy: A review. Int J Biol Macromol. 2022;202:539–57.

    Article  CAS  PubMed  Google Scholar 

  116. De Francesco EM, Lappano R, Santolla MF, Marsico S, Caruso A, Maggiolini M. HIF-1 α/GPER signaling mediates the expression of VEGF induced by hypoxia in breast cancer associated fibroblasts (CAFs). Breast Cancer Res. 2013;15:1–18.

    Article  Google Scholar 

  117. Crawford Y, Kasman I, Yu L, Zhong C, Wu X, Modrusan Z, et al. PDGF-c mediates the angiogenic and tumorigenic properties of fibroblasts associated with tumors refractory to anti-VEGF treatment. Cancer Cell. 2009;15:21–34.

    Article  CAS  PubMed  Google Scholar 

  118. Ahirwar DK, Nasser MW, Ouseph MM, Elbaz M, Cuitiño MC, Kladney RD, et al. Fibroblast-derived CXCL12 promotes breast cancer metastasis by facilitating tumor cell intravasation. Oncogene. 2018;37:4428–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Presta M, Chiodelli P, Giacomini A, Rusnati M, Ronca R. Fibroblast growth factors (FGFs) in cancer: FGF traps as a new therapeutic approach. Pharmacol Ther. 2017;179:171–87.

    Article  CAS  PubMed  Google Scholar 

  120. Unterleuthner D, Neuhold P, Schwarz K, Janker L, Neuditschko B, Nivarthi H, et al. Cancer-associated fibroblast-derived WNT2 increases tumor angiogenesis in colon cancer. Angiogenesis. 2020;23:159–77.

    Article  CAS  PubMed  Google Scholar 

  121. Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121:335–48.

    Article  CAS  PubMed  Google Scholar 

  122. Nordby Y, Richardsen E, Rakaee M, Ness N, Donnem T, Patel HR, et al. High expression of PDGFR-β in prostate cancer stroma is independently associated with clinical and biochemical prostate cancer recurrence. Sci Rep. 2017;7:43378.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Pietras K, Pahler J, Bergers G, Hanahan D. Functions of paracrine PDGF signaling in the proangiogenic tumor stroma revealed by pharmacological targeting. PLoS Med. 2008;5: e19.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Robertson KA, Nalepa G, Yang F-C, Bowers DC, Ho CY, Hutchins GD, et al. Imatinib mesylate for plexiform neurofibromas in patients with neurofibromatosis type 1: a phase 2 trial. Lancet Oncol. 2012;13:1218–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Wang F-T, Sun W, Zhang J-T, Fan Y-Z. Cancer-associated fibroblast regulation of tumor neo-angiogenesis as a therapeutic target in cancer. Oncol Lett. 2019;17:3055–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Li Z, Sun C, Qin Z. Metabolic reprogramming of cancer-associated fibroblasts and its effect on cancer cell reprogramming. Theranostics. 2021;11:8322.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Guido C, Whitaker-Menezes D, Capparelli C, Balliet R, Lin Z, Pestell RG, et al. Metabolic reprogramming of cancer-associated fibroblasts by TGF-β drives tumor growth: Connecting TGF-β signaling with “warburg-like” cancer metabolism and l-lactate production. Cell Cycle. 2012;11:3019–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Niu N, Shen X, Wang Z, Chen Y, Weng Y, Yu F, et al. Tumor cell-intrinsic epigenetic dysregulation shapes cancer-associated fibroblasts heterogeneity to metabolically support pancreatic cancer. Cancer Cell. 2024;42:869–84.

    Article  CAS  PubMed  Google Scholar 

  129. Demircioglu F, Wang J, Candido J, Costa AS, Casado P, de Luxan Delgado B, et al. Cancer associated fibroblast FAK regulates malignant cell metabolism. Nat Commun. 2020;11:1290.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Murthy D, Attri KS, Shukla SK, Thakur R, Chaika NV, He C, et al. Cancer-associated fibroblast-derived acetate promotes pancreatic cancer development by altering polyamine metabolism via the ACSS2–SP1–SAT1 axis. Nat Cell Biol. 2024;26:613–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Karakasheva TA, Lin EW, Tang Q, Qiao E, Waldron TJ, Soni M, et al. IL-6 mediates cross-talk between tumor cells and activated fibroblasts in the tumor microenvironment. Can Res. 2018;78:4957–70.

    Article  CAS  Google Scholar 

  132. Guo Z, Zhang H, Fu Y, Kuang J, Zhao B, Zhang L, et al. Cancer-associated fibroblasts induce growth and radioresistance of breast cancer cells through paracrine IL-6. Cell Death Discov. 2023;9:6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Tyan S-W, Kuo W-H, Huang C-K, Pan C-C, Shew J-Y, Chang K-J, et al. Breast cancer cells induce cancer-associated fibroblasts to secrete hepatocyte growth factor to enhance breast tumorigenesis. PLoS ONE. 2011;6: e15313.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Suh J, Kim D-H, Lee Y-H, Jang J-H, Surh Y-J. Fibroblast growth factor-2, derived from cancer-associated fibroblasts, stimulates growth and progression of human breast cancer cells via FGFR1 signaling. Mol Carcinog. 2020;59:1028–40.

    Article  CAS  PubMed  Google Scholar 

  135. Chan T-S, Hsu C-C, Pai VC, Liao W-Y, Huang S-S, Tan K-T, et al. Metronomic chemotherapy prevents therapy-induced stromal activation and induction of tumor-initiating cells. J Exp Med. 2016;213:2967.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Chen B, Sang Y, Song X, Zhang D, Wang L, Zhao W, et al. Exosomal miR-500a-5p derived from cancer-associated fibroblasts promotes breast cancer cell proliferation and metastasis through targeting USP28. Theranostics. 2021;11:3932.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Wang H, Wei H, Wang J, Li L, Chen A, Li Z. MicroRNA-181d-5p-containing exosomes derived from CAFs promote EMT by regulating CDX2/HOXA5 in breast cancer. Mol Therapy-Nucleic Acids. 2020;19:654–67.

    Article  CAS  Google Scholar 

  138. Yoshida T, Ishii G, Goto K, Neri S, Hashimoto H, Yoh K, et al. Podoplanin-positive cancer-associated fibroblasts in the tumor microenvironment induce primary resistance to EGFR-TKIs in lung adenocarcinoma with EGFR mutation. Clin Cancer Res. 2015;21:642–51.

    Article  CAS  PubMed  Google Scholar 

  139. Chan JSK, Sng MK, Teo ZQ, Chong HC, Twang JS, Tan NS. Targeting nuclear receptors in cancer-associated fibroblasts as concurrent therapy to inhibit development of chemoresistant tumors. Oncogene. 2018;37:160–73.

    Article  CAS  PubMed  Google Scholar 

  140. Brabletz T, Kalluri R, Nieto MA, Weinberg RA. EMT in cancer. Nat Rev Cancer. 2018;18:128–34.

    Article  CAS  PubMed  Google Scholar 

  141. Ligorio M, Sil S, Malagon-Lopez J, Nieman LT, Misale S, Di Pilato M, et al. Stromal microenvironment shapes the intratumoral architecture of pancreatic cancer. Cell. 2019;178:160–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Fiori ME, Di Franco S, Villanova L, Bianca P, Stassi G, De Maria R. Cancer-associated fibroblasts as abettors of tumor progression at the crossroads of EMT and therapy resistance. Mol Cancer. 2019;18:1–16.

    Article  Google Scholar 

  143. Yu Y, Xiao C, Tan L, Wang Q, Li X, Feng Y. Cancer-associated fibroblasts induce epithelial–mesenchymal transition of breast cancer cells through paracrine TGF-β signalling. Br J Cancer. 2014;110:724–32.

    Article  CAS  PubMed  Google Scholar 

  144. Shan T, Chen S, Chen X, Lin WR, Li W, Ma J, et al. Prometastatic mechanisms of CAF-mediated EMT regulation in pancreatic cancer cells. Int J Oncol. 2017;50:121–8.

    Article  CAS  PubMed  Google Scholar 

  145. Yi Y, Zeng S, Wang Z, Wu M, Ma Y, Ye X, et al. Cancer-associated fibroblasts promote epithelial-mesenchymal transition and EGFR-TKI resistance of non-small cell lung cancers via HGF/IGF-1/ANXA2 signaling. Biochimica et Biophysica Acta BBA-Mol Basis Dis. 2018;1864:793–803.

    Article  CAS  Google Scholar 

  146. Zhuang J, Lu Q, Shen B, Huang X, Shen L, Zheng X, et al. TGF β 1 secreted by cancer-associated fibroblasts induces epithelial-mesenchymal transition of bladder cancer cells through lncRNA-ZEB2NAT. Sci Rep. 2015;5:11924.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Ren Y, Jia H, Xu Y, Zhou X, Zhao X, Wang Y, et al. Paracrine and epigenetic control of CAF-induced metastasis: the role of HOTAIR stimulated by TGF-ss1 secretion. Mol Cancer. 2018;17:1–14.

    Article  CAS  Google Scholar 

  148. Zhou X, Han J, Zuo A, Ba Y, Liu S, Xu H, et al. THBS2+ cancer-associated fibroblasts promote EMT leading to oxaliplatin resistance via COL8A1-mediated PI3K/AKT activation in colorectal cancer. Mol Cancer. 2024;23:282.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Fuchs Y, Steller H. Programmed cell death in animal development and disease. Cell. 2011;147:742–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Ouyang L, Shi Z, Zhao S, Wang F-T, Zhou T-T, Liu B, et al. Programmed cell death pathways in cancer: a review of apoptosis, autophagy and programmed necrosis. Cell Prolif. 2012;45:487–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Zhang H, Deng T, Liu R, Ning T, Yang H, Liu D, et al. CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Mol Cancer. 2020;19:1–17.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Su S, Chen J, Yao H, Liu J, Yu S, Lao L, et al. CD10+ GPR77+ cancer-associated fibroblasts promote cancer formation and chemoresistance by sustaining cancer stemness. Cell. 2018;172:841–56.

    Article  CAS  PubMed  Google Scholar 

  154. Hasegawa T, Yashiro M, Nishii T, Matsuoka J, Fuyuhiro Y, Morisaki T, et al. Cancer-associated fibroblasts might sustain the stemness of scirrhous gastric cancer cells via transforming growth factor-β signaling. Int J Cancer. 2014;134:1785–95.

    Article  CAS  PubMed  Google Scholar 

  155. Cazet AS, Hui MN, Elsworth BL, Wu SZ, Roden D, Chan C-L, et al. Targeting stromal remodeling and cancer stem cell plasticity overcomes chemoresistance in triple negative breast cancer. Nat Commun. 2018;9:2897.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Biffi G, Oni TE, Spielman B, Hao Y, Elyada E, Park Y, et al. IL-1-induced JAK/STAT signaling is antagonized by TGF-β to shape CAF heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov. 2019;9:282–301.

    Article  PubMed  Google Scholar 

  157. Sheng N, Shindo K, Ohuchida K, Shinkawa T, Zhang B, Feng H, et al. TAK1 promotes an immunosuppressive tumor microenvironment through cancer-associated fibroblast phenotypic conversion in pancreatic ductal adenocarcinoma. Clin Cancer Res. 2024;30:5138–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Lee JJ, Bernard V, Semaan A, Monberg ME, Huang J, Stephens BM, et al. Elucidation of tumor-stromal heterogeneity and the ligand-receptor interactome by single-cell transcriptomics in real-world pancreatic cancer biopsies. Clin Cancer Res. 2021;27:5912–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Sebastian A, Hum NR, Martin KA, Gilmore SF, Peran I, Byers SW, et al. Single-cell transcriptomic analysis of tumor-derived fibroblasts and normal tissue-resident fibroblasts reveals fibroblast heterogeneity in breast cancer. Cancers. 2020;12:1307.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Scanlan MJ, Raj B, Calvo B, Garin-Chesa P, Sanz-Moncasi MP, Healey JH, et al. Molecular cloning of fibroblast activation protein alpha, a member of the serine protease family selectively expressed in stromal fibroblasts of epithelial cancers. Proc Natl Acad Sci. 1994;91:5657–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Garin-Chesa P, Old LJ, Rettig WJ. Cell surface glycoprotein of reactive stromal fibroblasts as a potential antibody target in human epithelial cancers. Proc Natl Acad Sci. 1990;87:7235–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Henry LR, Lee H-O, Lee JS, Klein-Szanto A, Watts P, Ross EA, et al. Clinical implications of fibroblast activation protein in patients with colon cancer. Clin Cancer Res. 2007;13:1736–41.

    Article  CAS  PubMed  Google Scholar 

  163. Costa A, Kieffer Y, Scholer-Dahirel A, Pelon F, Bourachot B, Cardon M, et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell. 2018;33:463–79.

    Article  CAS  PubMed  Google Scholar 

  164. Cremasco V, Astarita JL, Grauel AL, Keerthivasan S, Macisaac KD, Woodruff MC, et al. FAP delineates heterogeneous and functionally divergent stromal cells in immune-excluded breast tumors. Cancer Immunol Res. 2018;6:1472.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Friedman G, Levi-Galibov O, David E, Bornstein C, Giladi A, Dadiani M, et al. Cancer-associated fibroblast compositions change with breast cancer progression linking the ratio of S100A4+ and PDPN+ CAFs to clinical outcome. Nat Cancer. 2020;1:692–708.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. McAndrews KM, Chen Y, Darpolor JK, Zheng X, Yang S, Carstens JL, et al. Identification of functional heterogeneity of carcinoma-associated fibroblasts with distinct IL6-mediated therapy resistance in pancreatic cancer. Cancer Discov. 2022;12:1580–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Zhang S, Fang W, Zhou S, Zhu D, Chen R, Gao X, et al. Single cell transcriptomic analyses implicate an immunosuppressive tumor microenvironment in pancreatic cancer liver metastasis. Nat Commun. 2023;14:5123.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Grauel AL, Nguyen B, Ruddy D, Laszewski T, Schwartz S, Chang J, et al. TGF β-blockade uncovers stromal plasticity in tumors by revealing the existence of a subset of interferon-licensed fibroblasts. Nat Commun. 2020;11:6315.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Song H, Lu T, Han D, Zhang J, Gan L, Xu C, et al. YAP1 inhibition induces phenotype switching of cancer-associated fibroblasts to tumor suppressive in prostate cancer. Can Res. 2024;84:3728–42.

    Article  CAS  Google Scholar 

  170. Reardon DA, Akabani G, Coleman RE, Friedman AH, Friedman HS, Herndon JE, et al. Salvage radioimmunotherapy with murine iodine-131–labeled antitenascin monoclonal antibody 81C6 for patients with recurrent primary and metastatic malignant brain tumors: Phase II study results. J Clin Oncol. 2006;24:115–22.

    Article  CAS  PubMed  Google Scholar 

  171. Shum M, al. et. Early results from an open-label phase 1b/II study of eribulin mesylate (EM) + pegvorhyaluronidase alfa (PEGHP20) combination for the treatment of patients with HER2-negative, high-hyaluronan (HA) metastatic breast cancer (MBC). Ann Oncol. 2018;29:99.

    Article  Google Scholar 

  172. Yao Y, Guo Q, Cao Y, Qiu Y, Tan R, Yu Z, et al. Artemisinin derivatives inactivate cancer-associated fibroblasts through suppressing TGF-β signaling in breast cancer. J Exp Clin Cancer Res. 2018;37:1–14.

    Article  Google Scholar 

  173. Zboralski D, Osterkamp F, Christensen E, Bredenbeck A, Schumann A, Hoehne A, et al. Fibroblast activation protein targeted radiotherapy induces an immunogenic tumor microenvironment and enhances the efficacy of PD-1 immune checkpoint inhibition. Eur J Nucl Med Mol Imaging. 2023;50:2621–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Chen M, Xiang R, Wen Y, Xu G, Wang C, Luo S, et al. A whole-cell tumor vaccine modified to express fibroblast activation protein induces antitumor immunity against both tumor cells and cancer-associated fibroblasts. Sci Rep. 2015;5:14421.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Wen Y, Wang C-T, Ma T-T, Li Z-Y, Zhou L-N, Mu B, et al. Immunotherapy targeting fibroblast activation protein inhibits tumor growth and increases survival in a murine colon cancer model. Cancer Sci. 2010;101:2325–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Narra K, Mullins SR, Lee H-O, Strzemkowski-Brun B, Magalong K, Christiansen VJ, et al. Phase II trial of single agent val-boroPro (talabostat) inhibiting fibroblast activation protein in patients with metastatic colorectal cancer. Cancer Biol Ther. 2007;6:1691–9.

    Article  CAS  PubMed  Google Scholar 

  177. Jacobetz MA, Chan DS, Neesse A, Bapiro TE, Cook N, Frese KK, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut. 2013;62:112–20.

    Article  CAS  PubMed  Google Scholar 

  178. Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21:418–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Hingorani SR, Harris WP, Beck JT, Berdov BA, Wagner SA, Pshevlotsky EM, et al. Phase ib study of PEGylated recombinant human hyaluronidase and gemcitabine in patients with advanced pancreatic cancer. Clin Cancer Res. 2016;22:2848–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Hingorani SR, Zheng L, Bullock AJ, Seery TE, Harris WP, Sigal DS, et al. HALO 202: Randomized phase II study of PEGPH20 plus nab-paclitaxel/gemcitabine versus nab-paclitaxel/gemcitabine in patients with untreated, metastatic pancreatic ductal adenocarcinoma. J Clin Oncol. 2018;36:359–66.

    Article  CAS  PubMed  Google Scholar 

  181. Ramanathan RK, McDonough SL, Philip PA, Hingorani SR, Lacy J, Kortmansky JS, et al. Phase IB/II randomized study of FOLFIRINOX plus pegylated recombinant human hyaluronidase versus FOLFIRINOX alone in patients with metastatic pancreatic adenocarcinoma: SWOG S1313. J Clin Oncol. 2019;37:1062–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Van Cutsem E, Tempero MA, Sigal D, Oh D-Y, Fazio N, Macarulla T, et al. Randomized phase III trial of pegvorhyaluronidase alfa with nab-paclitaxel plus gemcitabine for patients with hyaluronan-high metastatic pancreatic adenocarcinoma. J Clin Oncol. 2020;38:3185–94.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Sica DA, Gehr TW, Ghosh S. Clinical pharmacokinetics of losartan. Clin Pharmacokinet. 2005;44:797–814.

    Article  CAS  PubMed  Google Scholar 

  184. Chauhan VP, Martin JD, Liu H, Lacorre DA, Jain SR, Kozin SV, et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat Commun. 2013;4:2516.

    Article  PubMed  Google Scholar 

  185. Hu C, Liu X, Ran W, Meng J, Zhai Y, Zhang P, et al. Regulating cancer associated fibroblasts with losartan-loaded injectable peptide hydrogel to potentiate chemotherapy in inhibiting growth and lung metastasis of triple negative breast cancer. Biomaterials. 2017;144:60–72.

    Article  CAS  PubMed  Google Scholar 

  186. Diop-Frimpong B, Chauhan VP, Krane S, Boucher Y, Jain RK. Losartan inhibits collagen i synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. Proc Natl Acad Sci. 2011;108:2909–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Murphy JE, Wo JY, Ryan DP, Clark JW, Jiang W, Yeap BY, et al. Total neoadjuvant therapy with FOLFIRINOX in combination with losartan followed by chemoradiotherapy for locally advanced pancreatic cancer: A phase 2 clinical trial. JAMA Oncol. 2019;5:1020–7.

    Article  PubMed  PubMed Central  Google Scholar 

  188. Darakhshan S, Pour AB. Tranilast: A review of its therapeutic applications. Pharmacol Res. 2015;91:15–28.

    Article  CAS  PubMed  Google Scholar 

  189. Panagi M, Voutouri C, Mpekris F, Papageorgis P, Martin MR, Martin JD, et al. TGF-β inhibition combined with cytotoxic nanomedicine normalizes triple negative breast cancer microenvironment towards anti-tumor immunity. Theranostics. 2020;10:1910.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Papageorgis P, Polydorou C, Mpekris F, Voutouri C, Agathokleous E, Kapnissi-Christodoulou CP, et al. Tranilast-induced stress alleviation in solid tumors improves the efficacy of chemo-and nanotherapeutics in a size-independent manner. Sci Rep. 2017;7:46140.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Incio J, Suboj P, Chin SM, Vardam-Kaur T, Liu H, Hato T, et al. Metformin reduces desmoplasia in pancreatic cancer by reprogramming stellate cells and tumor-associated macrophages. PLoS ONE. 2015;10: e0141392.

    Article  PubMed  PubMed Central  Google Scholar 

  192. Xu S, Yang Z, Jin P, Yang X, Li X, Wei X, et al. Metformin suppresses tumor progression by inactivating stromal fibroblasts in ovarian cancer. Mol Cancer Ther. 2018;17:1291–302.

    Article  CAS  PubMed  Google Scholar 

  193. Kozono S, Ohuchida K, Eguchi D, Ikenaga N, Fujiwara K, Cui L, et al. Pirfenidone inhibits pancreatic cancer desmoplasia by regulating stellate cells. Can Res. 2013;73:2345–56.

    Article  CAS  Google Scholar 

  194. Polydorou C, Mpekris F, Papageorgis P, Voutouri C, Stylianopoulos T. Pirfenidone normalizes the tumor microenvironment to improve chemotherapy. Oncotarget. 2017;8:24506.

    Article  PubMed  PubMed Central  Google Scholar 

  195. Takai K, Le A, Weaver VM, Werb Z. Targeting the cancer-associated fibroblasts as a treatment in triple-negative breast cancer. Oncotarget. 2016;7:82889.

    Article  PubMed  PubMed Central  Google Scholar 

  196. Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, et al. Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324:1457–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Ko AH, LoConte N, Tempero MA, Walker EJ, Kelley RK, Lewis S, et al. A phase i study of FOLFIRINOX plus IPI-926, a hedgehog pathway inhibitor, for advanced pancreatic adenocarcinoma. Pancreas. 2016;45:370–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Jimeno A, Weiss GJ, Miller WH Jr, Gettinger S, Eigl BJ, Chang ALS, et al. Phase i study of the hedgehog pathway inhibitor IPI-926 in adult patients with solid tumors. Clin Cancer Res. 2013;19:2766–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Bowles D, Keysar S, Glogowska M, Le P, Pittman M, Wang G, et al. A pilot study of cetuximab and the hedgehog inhibitor IPI-926 in recurrent/metastatic (r/m) head-and-neck squamous cell carcinoma: Molecular biology and therapeutics. Int J Radiat Oncol Biol Phys. 2014;88:511.

    Article  Google Scholar 

  200. Stephenson J, Richards D, Wolpin B, Becerra C, Hamm J, Messersmith W, et al. The safety of IPI-926, a novel hedgehog pathway inhibitor, in combination with gemcitabine in patients (pts) with metastatic pancreatic cancer. J Clin Oncol. 2011;29:4114–24.

    Article  Google Scholar 

  201. Catenacci DV, Junttila MR, Karrison T, Bahary N, Horiba MN, Nattam SR, et al. Randomized phase ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J Clin Oncol. 2015;33:4284–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Kim EJ, Sahai V, Abel EV, Griffith KA, Greenson JK, Takebe N, et al. Pilot clinical trial of hedgehog pathway inhibitor GDC-0449 (vismodegib) in combination with gemcitabine in patients with metastatic pancreatic adenocarcinoma. Clin Cancer Res. 2014;20:5937–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Lee JJ, Perera RM, Wang H, Wu D-C, Liu XS, Han S, et al. Stromal response to hedgehog signaling restrains pancreatic cancer progression. Proc Natl Acad Sci. 2014;111:E3091–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Neri S, Ishii G, Hashimoto H, Kuwata T, Nagai K, Date H, et al. Podoplanin-expressing cancer-associated fibroblasts lead and enhance the local invasion of cancer cells in lung adenocarcinoma. Int J Cancer. 2015;137:784–96.

    Article  CAS  PubMed  Google Scholar 

  205. Whatcott CJ, Ng S, Barrett MT, Hostetter G, Von Hoff DD, Han H. Inhibition of ROCK1 kinase modulates both tumor cells and stromal fibroblasts in pancreatic cancer. PLoS ONE. 2017;12: e0183871.

    Article  PubMed  PubMed Central  Google Scholar 

  206. Vennin C, Chin VT, Warren SC, Lucas MC, Herrmann D, Magenau A, et al. Transient tissue priming via ROCK inhibition uncouples pancreatic cancer progression, sensitivity to chemotherapy, and metastasis. Sci Transl Med. 2017;9:eaai8504.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Pein M, Insua Rodrı́guez J, Hongu T, Riedel A, Meier J, Wiedmann L, et al. Metastasis-initiating cells induce and exploit a fibroblast niche to fuel malignant colonization of the lungs. Nat Commun. 2020;11:1494.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGF β attenuates tumour response to PD-L1 blockade by contributing to exclusion of t cells. Nature. 2018;554:544–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Zhang Q, Hou X, Evans BJ, VanBlaricom JL, Weroha SJ, Cliby WA. LY2157299 monohydrate, a TGF-β R1 inhibitor, suppresses tumor growth and ascites development in ovarian cancer. Cancers. 2018;10:260.

    Article  PubMed  PubMed Central  Google Scholar 

  210. Yap TA, Vieito M, Baldini C, Sepúlveda-Sánchez JM, Kondo S, Simonelli M, et al. First-in-human phase i study of a next-generation, oral, TGF β receptor 1 inhibitor, LY3200882, in patients with advanced cancer. Clin Cancer Res. 2021;27:6666–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Hurwitz HI, Uppal N, Wagner SA, Bendell JC, Beck JT, Wade SM III, et al. Randomized, double-blind, phase II study of ruxolitinib or placebo in combination with capecitabine in patients with metastatic pancreatic cancer for whom therapy with gemcitabine has failed. J Clin Oncol. 2015;33:4039–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Gil-Martin M, Gomez Pardo P, Lopez-Tarruella S, Manso L, Perez-Fidalgo JA, Ademuyiwa FO, et al. Phase i study of the combination of balixafortide (CXCR4 inhibitor) and eribulin in HER2-negative metastatic breast cancer (MBC) patients (pts). J Clin Oncol. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1200/JCO.2017.35.15_suppl.2555.

    Article  Google Scholar 

  213. Mazzocca A, Fransvea E, Dituri F, Lupo L, Antonaci S, Giannelli G. Down-regulation of connective tissue growth factor by inhibition of transforming growth factor β blocks the tumor–stroma cross-talk and tumor progression in hepatocellular carcinoma. Hepatology. 2010;51:523–34.

    Article  CAS  PubMed  Google Scholar 

  214. Lee EQ, Duda DG, Muzikansky A, Gerstner ER, Kuhn JG, Reardon DA, et al. Phase i and biomarker study of plerixafor and bevacizumab in recurrent high-grade glioma. Clin Cancer Res. 2018;24:4643–9.

    Article  CAS  PubMed  Google Scholar 

  215. Ham I-H, Oh HJ, Jin H, Bae CA, Jeon S-M, Choi KS, et al. Targeting interleukin-6 as a strategy to overcome stroma-induced resistance to chemotherapy in gastric cancer. Mol Cancer. 2019;18:1–14.

    Article  Google Scholar 

  216. Shi Y, Gao W, Lytle NK, Huang P, Yuan X, Dann AM, et al. Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring. Nature. 2019;569:131–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Fernández-Nogueira P, Mancino M, Fuster G, López-Plana A, Jauregui P, Almendro V, et al. Tumor-associated fibroblasts promote HER2-targeted therapy resistance through FGFR2 activation. Clin Cancer Res. 2020;26:1432–48.

    Article  PubMed  Google Scholar 

  218. Damodaran S, André F, Unni N, Ferreira M, Giridhar K, Daniel B, et al. Abstract RF01–04: final results from the phase 2, open-label FOENIX-MBC2 study: Efficacy and safety of futibatinib in adult patients with locally advanced/metastatic HR+/HER2- breast cancer harboring high-level FGFR1 gene amplification. Cancer Res. 2024;84:01–4.

    Article  Google Scholar 

  219. Zervantonakis IK, Poskus MD, Scott AL, Selfors LM, Lin J-R, Dillon DA, et al. Fibroblast–tumor cell signaling limits HER2 kinase therapy response via activation of MTOR and antiapoptotic pathways. Proc Natl Acad Sci. 2020;117:16500–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Duluc C, Moatassim-Billah S, Chalabi-Dchar M, Perraud A, Samain R, Breibach F, et al. Pharmacological targeting of the protein synthesis mTOR/4E-BP 1 pathway in cancer-associated fibroblasts abrogates pancreatic tumour chemoresistance. EMBO Mol Med. 2015;7:735–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Liu Z, Hu S, Zhao X, Xu W, Ju F, Gu M, et al. AKAP12 positive fibroblast determines immunosuppressive contexture and immunotherapy response in patients with TNBC by promoting macrophage M2 polarization. J Immunother Cancer. 2024;12: e009877.

    Article  PubMed  PubMed Central  Google Scholar 

  222. Jiang Y, Wang Y, Chen G, Sun F, Wu Q, Huang Q, et al. Nicotinamide metabolism face-off between macrophages and fibroblasts manipulates the microenvironment in gastric cancer. Cell Metab. 2024;36(8):1806–22.

    Article  CAS  PubMed  Google Scholar 

  223. Picozzi V, Rocha F, Winter J, Pishvaian M, Mody K, Glaspy J, et al. Gemcitabine/nab-paclitaxel and pamrevlumab in locally advanced pancreatic cancer (LAPC): A phase 1/2 trial. 2019.

  224. Picozzi VJ, Khan KH, Hammel P, Reni M, Lin D, Lee WJ, et al. LAPIS: Randomized phase 3 trial of chemotherapy (CTX) with and without pamrevlumab (PAM) for locally advanced pancreatic cancer (LAPC). 2025.

  225. Sharma M, Turaga RC, Yuan Y, Satyanarayana G, Mishra F, Bian Z, et al. Simultaneously targeting cancer-associated fibroblasts and angiogenic vessel as a treatment for TNBC. J Exp Med. 2021;218: e20200712.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Hofheinz R-D, Al-Batran S-E, Hartmann F, Hartung G, Jäger D, Renner C, et al. Stromal antigen targeting by a humanised monoclonal antibody: An early phase II trial of sibrotuzumab in patients with metastatic colorectal cancer. Oncol Res Treat. 2003;26:44–8.

    Article  CAS  Google Scholar 

  227. Wang L-CS, Lo A, Scholler J, Sun J, Majumdar RS, Kapoor V, et al. Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor t cells can inhibit tumor growth and augment host immunity without severe toxicity. Cancer Immunol Res. 2014;2:154–66.

    Article  CAS  PubMed  Google Scholar 

  228. Chen M, Xiao L, Jia H, Wang S, Jiang X, Lei X, et al. Stereotactic ablative radiotherapy and FAP α-based cancer vaccine suppresses metastatic tumor growth in 4T1 mouse breast cancer. Radiother Oncol. 2023;189: 109946.

    Article  CAS  PubMed  Google Scholar 

  229. Fang J, Xiao L, Joo K-I, Liu Y, Zhang C, Liu S, et al. A potent immunotoxin targeting fibroblast activation protein for treatment of breast cancer in mice. Int J Cancer. 2016;138:1013–23.

    Article  CAS  PubMed  Google Scholar 

  230. Ji T, Zhao Y, Ding Y, Wang J, Zhao R, Lang J, et al. Transformable peptide nanocarriers for expeditious drug release and effective cancer therapy via cancer-associated fibroblast activation. Angew Chem Int Ed. 2016;55:1050–5.

    Article  CAS  Google Scholar 

  231. Scott AM, Wiseman G, Welt S, Adjei A, Lee F-T, Hopkins W, et al. A phase i dose-escalation study of sibrotuzumab in patients with advanced or metastatic fibroblast activation protein-positive cancer. Clin Cancer Res. 2003;9:1639–47.

    CAS  PubMed  Google Scholar 

  232. Liu Y, Sun Y, Wang P, Li S, Dong Y, Zhou M, et al. FAP-targeted CAR-t suppresses MDSCs recruitment to improve the antitumor efficacy of claudin18. 2-targeted CAR-t against pancreatic cancer. J Transl Med. 2023;21:255.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Fabre M, Ferrer C, Domı́nguez-Hormaetxe S, Bockorny B, Murias L, Seifert O, et al. OMTX705, a novel FAP-targeting ADC demonstrates activity in chemotherapy and pembrolizumab-resistant solid tumor models. Clin Cancer Res. 2020;26:3420–30.

    Article  CAS  PubMed  Google Scholar 

  234. Zhen Z, Tang W, Wang M, Zhou S, Wang H, Wu Z, et al. Protein nanocage mediated fibroblast-activation protein targeted photoimmunotherapy to enhance cytotoxic t cell infiltration and tumor control. Nano Lett. 2017;17:862–9.

    Article  CAS  PubMed  Google Scholar 

  235. Zhou S, Zhen Z, Paschall AV, Xue L, Yang X, Bebin Blackwell A-G, et al. FAP-targeted photodynamic therapy mediated by ferritin nanoparticles elicits an immune response against cancer cells and cancer associated fibroblasts. Adv Func Mater. 2021;31:2007017.

    Article  CAS  Google Scholar 

  236. Watanabe S, Noma K, Ohara T, Kashima H, Sato H, Kato T, et al. Photoimmunotherapy for cancer-associated fibroblasts targeting fibroblast activation protein in human esophageal squamous cell carcinoma. Cancer Biol Ther. 2019;20:1234–48.

    Article  PubMed  PubMed Central  Google Scholar 

  237. Li L, Zhou S, Lv N, Zhen Z, Liu T, Gao S, et al. Photosensitizer-encapsulated ferritins mediate photodynamic therapy against cancer-associated fibroblasts and improve tumor accumulation of nanoparticles. Mol Pharm. 2018;15:3595–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Assouline B, Kahn R, Hodali L, Condiotti R, Engel Y, Elyada E, et al. Senescent cancer-associated fibroblasts in pancreatic adenocarcinoma restrict CD8+ t cell activation and limit responsiveness to immunotherapy in mice. Nat Commun. 2024;15:6162.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Liu L, Huang H, Cheng B, Xie H, Peng W, Cui H, et al. Revealing the role of cancer-associated fibroblast senescence in prognosis and immune landscape in pancreatic cancer. iScience. 2025;28.

  240. Lawrence JA, Akman SA, Melin SA, Case LD, Schwartz GG. Oral paricalcitol (19-nor-1, 25-dihydroxyvitamin D2) in women receiving chemotherapy for metastatic breast cancer: A feasibility trial. Cancer Biol Ther. 2013;14:476–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. Hanley CJ, Mellone M, Ford K, Thirdborough SM, Mellows T, Frampton SJ, et al. Targeting the myofibroblastic cancer-associated fibroblast phenotype through inhibition of NOX4. JNCI: J Nat Cancer Inst. 2018;110:109–20.

    Article  CAS  PubMed  Google Scholar 

  242. Miao L, Liu Q, Lin CM, Luo C, Wang Y, Liu L, et al. Targeting tumor-associated fibroblasts for therapeutic delivery in desmoplastic tumors. Can Res. 2017;77:719–31.

    Article  CAS  Google Scholar 

  243. Froeling FE, Feig C, Chelala C, Dobson R, Mein CE, Tuveson DA, et al. Retinoic acid–induced pancreatic stellate cell quiescence reduces paracrine wnt–β-catenin signaling to slow tumor progression. Gastroenterology. 2011;141:1486–97.

    Article  CAS  PubMed  Google Scholar 

  244. Ene-Obong A, Clear AJ, Watt J, Wang J, Fatah R, Riches JC, et al. Activated pancreatic stellate cells sequester CD8+ t cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology. 2013;145:1121–32.

    Article  CAS  PubMed  Google Scholar 

  245. Carapuça EF, Gemenetzidis E, Feig C, Bapiro TE, Williams MD, Wilson AS, et al. Anti-stromal treatment together with chemotherapy targets multiple signalling pathways in pancreatic adenocarcinoma. J Pathol. 2016;239:286–96.

    Article  PubMed  PubMed Central  Google Scholar 

  246. Han X, Li Y, Xu Y, Zhao X, Zhang Y, Yang X, et al. Reversal of pancreatic desmoplasia by re-educating stellate cells with a tumour microenvironment-activated nanosystem. Nat Commun. 2018;9:3390.

    Article  PubMed  PubMed Central  Google Scholar 

  247. Dauer P, Zhao X, Gupta VK, Sharma N, Kesh K, Gnamlin P, et al. Inactivation of cancer-associated-fibroblasts disrupts oncogenic signaling in pancreatic cancer cells and promotes its regression. Can Res. 2018;78:1321–33.

    Article  CAS  Google Scholar 

  248. Banerjee S, Modi S, McGinn O, Zhao X, Dudeja V, Ramakrishnan S, et al. Impaired synthesis of stromal components in response to minnelide improves vascular function, drug delivery, and survival in pancreatic cancer. Clin Cancer Res. 2016;22:415–25.

    Article  CAS  PubMed  Google Scholar 

  249. Sherman MH, Ruth TY, Engle DD, Ding N, Atkins AR, Tiriac H, et al. Vitamin d receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell. 2014;159:80–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Kim DJ, Dunleavey JM, Xiao L, Ollila DW, Troester MA, Otey CA, et al. Suppression of TGF β-mediated conversion of endothelial cells and fibroblasts into cancer associated (myo) fibroblasts via HDAC inhibition. Br J Cancer. 2018;118:1359–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Zhu Y, Zhang L, Zha H, Yang F, Hu C, Chen L, et al. Stroma-derived fibrinogen-like protein 2 activates cancer-associated fibroblasts to promote tumor growth in lung cancer. Int J Biol Sci. 2017;13:804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Haubeiss S, Schmid JO, Mürdter TE, Sonnenberg M, Friedel G, van der Kuip H, et al. Dasatinib reverses cancer-associated fibroblasts (CAFs) from primary lung carcinomas to a phenotype comparable to that of normal fibroblasts. Mol Cancer. 2010;9:1–8.

    Article  Google Scholar 

  253. Zhang X, Lao M, Yang H, Sun K, Dong Y, He L, et al. Targeting cancer-associated fibroblast autophagy renders pancreatic cancer eradicable with immunochemotherapy by inhibiting adaptive immune resistance. Autophagy. 2024;20:1314–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  254. Mazzeo L, Ghosh S, Di Cicco E, Isma J, Tavernari D, Samarkina A, et al. ANKRD1 is a mesenchymal-specific driver of cancer-associated fibroblast activation bridging androgen receptor loss to AP-1 activation. Nat Commun. 2024;15:1038.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  255. Ma H, Srivastava S, Ho SWT, Xu C, Lian BSX, Ong X, et al. Spatially resolved tumor ecosystems and cell states in gastric adenocarcinoma progression and evolution. Cancer Discovery. 2025;OF1–26.

  256. Lin J-R, Wang S, Coy S, Chen Y-A, Yapp C, Tyler M, et al. Multiplexed 3D atlas of state transitions and immune interaction in colorectal cancer. Cell. 2023;186:363–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  257. Jovic D, Liang X, Zeng H, Lin L, Xu F, Luo Y. Single-cell RNA sequencing technologies and applications: A brief overview. Clin Transl Med. 2022;12: e694.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Braxton AM, Kiemen AL, Grahn MP, Forjaz A, Parksong J, Mahesh Babu J, et al. 3D genomic mapping reveals multifocality of human pancreatic precancers. Nature. 2024;629:679–87.

    Article  CAS  PubMed  Google Scholar 

  259. Kiemen AL, Braxton AM, Grahn MP, Han KS, Babu JM, Reichel R, et al. CODA: Quantitative 3D reconstruction of large tissues at cellular resolution. Nat Methods. 2022;19:1490–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Onder L, Papadopoulou C, Lütge A, Cheng H-W, Lütge M, Perez-Shibayama C, et al. Fibroblastic reticular cells generate protective intratumoral t cell environments in lung cancer. Cell. 2025;188:430–46.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank Dr. Julian Heng (ORCID: 0000-0002-0378-7078) for his professional English-language editing of this manuscript.

Funding

This work was supported by Chengdu Technology Bureau (2024-YF05-02230-SN), Sichuan Cancer Hospital Outstanding Youth Funding (Grant No. YB2021031) and Health Commission of Sichuan Province (24QNMP038).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, H. J. and M.C.; writing-original draft preparation, H,J. and X, C..; writing-review and editing, L. Z. and M.C..

Corresponding author

Correspondence to Meihua Chen.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jia, H., Chen, X., Zhang, L. et al. Cancer associated fibroblasts in cancer development and therapy. J Hematol Oncol 18, 36 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-025-01688-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13045-025-01688-0

Keywords