Skip to main content

Cancer-associated fibroblasts as therapeutic targets for cancer: advances, challenges, and future prospects

Abstract

Research into cancer treatment has been mainly focused on developing therapies to directly target cancer cells. Over the past decade, extensive studies have revealed critical roles of the tumour microenvironment (TME) in cancer initiation, progression, and drug resistance. Notably, cancer-associated fibroblasts (CAFs) have emerged as one of the primary contributors in shaping TME, creating a favourable environment for cancer development. Many preclinical studies have identified promising targets on CAFs, demonstrating remarkable efficacy of some CAF-targeted treatments in preclinical models. Encouraged by these compelling findings, therapeutic strategies have now advanced into clinical evaluation. We aim to provide a comprehensive review of relevant subjects on CAFs, including CAF-related markers and targets, their multifaceted roles, and current landscape of ongoing clinical trials. This knowledge can guide future research on CAFs and advocate for clinical investigations targeting CAFs.

Introduction

The tumour microenvironment (TME) has emerged as a pivotal player in cancer development and drug resistance. With the introduction of the "seed and soil" theory in 1989 by Stephen Paget [1], the significance of TME has grown considerably. Substantial evidence now indicates that diverse cell populations within tumours create a supportive environment for the survival, growth, and metastasis of cancer cells. In line with the "seed and soil" theory, tumour cells ("seed") preferentially grow in organs with a suitable microenvironment ("soil"), leading to a non-random distribution of metastasis among organs. It is now widely accepted that cancer behaviours are regulated by both intrinsic factors and the intricate TME. The TME constitutes a complex and dynamic niche formed by various cellular and molecular components engaging in communication and interactions with cancer cells. Through these inter-TME dialogues and crosstalk with cancer cells, the TME provide a nurturing and protective environment for cancer cells.

The TME involves two major cellular components: immune cells and stromal cells. TME immune cells include myeloid-derived suppressor cells (MDSC), tumour-associated macrophages (TAMs), tumour-associated neutrophils, regulatory T (Treg) cells, natural killer cells, dendritic cells, B cells, effector T cells and T helper cells. The roles of these immune cells in the TME and cancer development have been thoroughly reviewed elsewhere [2,3,4,5,6], and will not be discussed here. The other large proportion of cells within TME are stromal cells, and the tumour-stroma ratio has shown promise as a prognostic biomarker for certain cancer types in clinical settings [7,8,9,10,11]. TME stromal cells include endothelial cells (blood vessels), as well as inter-related cancer-associated fibroblasts (CAFs), mesenchymal stromal cells (MSCs), and pericytes, which all share certain characteristics and plasticity, being able to interconvert and differentiate into different cell types [12, 13]. The roles of these broad tumour stromal cells have been comprehensively reviewed by Xu and colleagues [14].

CAFs, a subtype of activated fibroblasts, are the most abundant and prominent cell population within the tumour stroma. Research into CAFs has surged over the past decade, greatly advancing our understanding of their roles in the TME and potential as therapeutic targets. CAFs have been shown to play multiple essential roles in cancer initiation, progression, and metastasis, through interaction and communication with cancer cells or regulating extracellular matrix (ECM) remodelling and immune cell infiltration (Fig. 1) [15,16,17,18,19,20,21,22]. In this review, we provide a summary of markers for identifying CAFs and their subtypes. Importantly, we conduct a comprehensive review of both completed and ongoing clinical trials associated with CAF-targeted therapies. We also discuss the existing challenges in CAF-related studies and proposes a direction for future research.

Fig. 1
figure 1

CAFs regulate cancer development and progression through interactions and communication with various cell types within the tumour microenvironment by secreting a range of factors. Treg: regulatory T cell; MDSC: myeloid-derived suppressor cell; TAM: tumour-associated macrophage; DC: dendritic cells; NK cell: natural killer cell. The figure was generated using BioRender

CAF-related markers

Fibroblasts have long presented a challenge to define, given the absence of reliable markers specific to their cell lineage. Originating from the mesoderm during embryonic development, they share this mesenchymal lineage with adipocytes and bone cells including osteoblasts and chondrocytes. However, the few proteins that may infer a cell being a fibroblast are neither exclusive to fibroblasts alone nor uniformly present across all fibroblast subtypes [17, 23, 24]. Consequently, cells labelled as fibroblasts were often defined through negative selection—those lacking markers for epithelial, endothelial, and inflammatory cells are presumed to be fibroblasts. In addition to the absence of such markers, the morphology and location of fibroblasts are instrumental to identify them in the past. Fibroblasts exhibit an elongated, spindle-like, fusiform phenotype, characterised by a tapered structure at both ends and extended processes. Recent advancement in single-cell RNA sequencing (scRNA-seq) and imaging techniques have identified several proteins highly expressed in fibroblasts within the TME, serving as positive markers for identifying CAFs [25]. It is important to note, however, that these markers may not be universally expressed by all CAFs across different cancer types. The heterogeneity and plasticity of CAFs play a critical role in their diverse functions within tumours. CAFs can be derived or transformed from various cell types such as mesenchymal cells, normal fibroblasts, quiescent stellate cells in the pancreas and liver. Efforts to identify CAFs within the tumour stroma have led to identification and application of various CAF-related markers. These markers have diverse roles in host, CAFs, and cancer cells as shown in preclinical studies (Table 1).

Table 1 General CAF-related markers and their phenotypes upon target deficiency in cancer

Fibroblast activation protein (FAP)

FAP, a membrane-bound serine protease belonging to the dipeptidyl peptidase (DPP) family, is commonly expressed by CAFs and certain tumour cells such as sarcoma [26,27,28]. Additionally, FAP expression has been observed in fibroblasts participating in wound healing and chronic inflammatory conditions such as arthritis and cirrhosis [29,30,31,32]. The substantial upregulation of FAP is considered a biomarker for CAFs [33, 34], with potential as a unfavourable prognosis biomarker for various cancers [35,36,37,38]. However, contradictory findings suggest that high FAP expression may correlate with a better prognosis in some patients [39, 40].

High FAP expression has been correlated with enhanced tumour growth and metastatic potential [41,42,43,44]. FAP-positive (FAP+) CAFs produce ECM proteins that contribute to migration of tumour cells [45, 46]. In a transgenic mouse model, depletion of FAP-expressing cells led to rapid hypoxic necrosis mediated by interferon-gamma (IFN-γ) and tumour necrosis factor-alpha (TNF-α), both associated with CD8+ T cell-dependent tumour cell killing [47]. FAP+ CAF-induced immune suppression could also be mediated through the CXCL12-CXCR4 axis, and inhibition of CXCR4 resulted in eradication of cancer cells by increasing intratumoral CD8+ T cells [48]. Further insight into the role of FAP in the immunosuppressive TME was revealed by a murine liver tumour model, demonstrating that FAP+ CAFs mediate immunosuppression through STAT3-CCL2 signalling and recruitment of MDSCs [49]. Tumorigenesis and tumour growth were reduced in FAP knockout (Fap−/−) mice in both lung and colon cancer models [50]. Global FAP knockout delayed the onset of pancreatic tumours, increased tumour necrosis, impeded metastasis, and prolonged mice survival in the KPC (LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre) pancreatic cancer model [43]. Consistently, silencing FAP in CAFs was associated with impaired tumour-promoting effects in preclinical studies [51,52,53,54]. Interestingly, knockdown of FAP in cancer cells also led to reduced cell proliferation, invasion, and metastasis in oral squamous cell carcinoma (OSCC) and prostate cancer [55, 56]. These findings suggest that FAP may serve as a promising therapeutic target, in addition to its role as a CAF marker.

α-Smooth muscle actin (α-SMA)

α-SMA is another frequently used marker for activated fibroblasts. High expression of α-SMA in CAFs was associated with poor prognosis of cancer patients [57,58,59]. CAFs with high α-SMA expression can stimulate growth of luminal breast cancer cells, primarily through the secretion of osteopontin (OPN) [57]. Tumours harbouring CAFs with elevated α-SMA expression exhibited high metastatic potential [58]. Conversely, CAFs expressing low levels of α-SMA suppressed self-renewal and growth of stem-like cancer cells through the signalling molecule bone morphogenetic protein 4 (BMP4) [60]. The α-SMA+ CAFs can promote the generation and proliferation of CD44+CD24 breast cancer stem cells by secreting CXCL12 that activates CXCR4 on cancer cells [61]. Conditioned medium from α-SMA+ CAFs enhanced tumorigenicity in a co-culture assay of hepatocellular carcinoma (HCC) [62]. This effect was attributed to α-SMA+ CAF-derived hepatocyte growth factor (HGF), regulating the c-Met/FRA1/HEY1 signalling pathway in HCC cells [62]. Additionally, α-SMA+ CAFs secret a range of cytokines, such as M-CSF, IL-6, IL-8, IL-10, TGF-β, and CCL-2, inducing macrophage differentiation and M2 polarization that contributes to immunosuppressive TME [63,64,65]. These cytokines can also activate STAT3-PDL1 signalling in neutrophils [66], further contributing to the establishment of a suppressive TME. Although the tumour-promoting roles of α-SMA + CAFs have been demonstrated in many cancers, they appear to have the opposite effect in pancreatic cancer. In a pancreatic cancer animal model, the deletion of α-SMA + CAFs led to an increase in CD4 + Foxp3 + regulatory T cells within the tumours, resulting in accelerated tumour growth [67]. This may be due to the tumour-restricting role of the stroma, which acts as a physical barrier to limit the growth of pancreatic cancer cells and the infiltration of tumour-supporting immune cells. In fact, deletion of Sonic hedgehog (SHH) in a pancreatic ductal adenocarcinoma (PDAC) model also reduced stromal content and led to increased tumour growth [68]. These findings suggest that while eliminating α-SMA + CAFs could be a promising strategy to inhibit tumour growth in many cancer types, it should be carefully evaluated when treating pancreatic cancer.

Platelet-derived growth factor receptor α/β (PDGFRα/β)

PDGFRα/β, a tyrosine kinase receptor, functions through the formation of homodimers (αα or ββ) or heterodimers (αβ), each exhibiting distinct interactions with PDGF ligand dimers, ultimately leading to activation of various signalling pathways [69,70,71]. PDGFR signalling plays a crucial role in development of organs, such as lung and kidney [72,73,74]. As a less specific marker for CAFs, PDGFRα/β is also expressed in normal fibroblasts, smooth muscle cells, and pericytes [75, 76]. High expression of PDGFRβ in tumour stroma was associated with large tumour size, advanced stage, and high vessel density in prostate cancer [77]. Elevated levels of PDGFRβ were associated with an increased risk of recurrence in breast and colorectal cancers [78, 79]. However, in patients with epithelial ovarian cancer, high PDGFRα/β expression in both tumour and stromal cells did not show prognostic significance [80].

Increased PDGFRα/β activity was observed in sarcoma cancer stem-like cells, promoting migration, invasion, and chemoresistance [81]. PDGFRα can interact with integrin α5β1 to promote cell contraction and reorganization of the ECM, resulting in directional migration of prostate and pancreatic cancer cells [82]. Integrin α11 also binds to PDGFRβ on CAFs, leading to increased invasion of breast cancer cells [83]. PDGFRβ+ CAFs, when stimulated by PDGF, can enhance migration and invasion of co-cultured colorectal cancer cells in a stanniocalcin-1-dependent manner [84]. By interacting with TGFβR, PDGFRβ can induce differentiation of MSCs into CAFs [85]. In a pancreatic cancer mouse model, PDGFRα+ CAFs accelerated tumour proliferation, in contrast to normal pancreatic fibroblasts that impeded tumour progression. Further categorization of PDGFRα+ CAFs revealed that PDGFRα+/SAA3 (Serum Amyloid A3)+ CAFs could enhance PDAC progression, whereas PDGFRα+ CAFs without SAA expression suppress tumour growth, attributed to Mpp6 overexpression [86].

The immunomodulatory effects of PDGFRα/β+ CAFs have also been well-documented in several studies. PDGFRα+ CAFs secrete Chitinase 3-like 1 to induce macrophage recruitment and M2 polarization in breast cancer [87]. In a co-culture assay, T cells, in the presence of PDGFRα/β+PDPN (podoplanin) + CAFs, exhibited low cytotoxicity towards co-cultured tumour cells [88]. The reduced T cell cytotoxicity could be resulted from increased apoptosis of FAS-expressing CD8+ T cells, a process mediated through the expression of FAS ligand and programmed death-ligand 2 (PD-L2) by CAFs [88]. Interestingly, high PDGFRα expression in CAFs was also associated with increased immune infiltration, potent T cell cytotoxicity, and prolonged survival in PDAC [89], highlighting the complex roles of PDGFRα/β in different contexts. Therefore, PDGFRα/β-targeted monotherapy may not be suitable for treating PDAC. In addition to its interesting roles in CAFs, PDGFR also plays a direct role in cancer cells. Knockdown or knockout of PDGFRA in gastrointestinal (GI) and glioblastoma (GBM) cancer cells suppressed tumour proliferation [90,91,92]. In BRCA1-deficient breast cancer cells, deletion of PDGFRβ promoted cell death and inhibited tumorigenesis [93]. PDGFRα/β could therefore serve as promising targets for direct anti-cancer therapy in treating these cancers.

Vimentin

Vimentin, a type III intermediate filament protein, serves as a major component of the cytoskeleton in non-epithelial cells, particularly mesenchymal cells. While high vimentin expression was observed in CAFs, normal fibroblasts (NFs) also exhibited similar levels of vimentin [94]. Presence of α-SMAVimentin+ CAFs was associated with poor survival in PDAC patients [95], and high vimentin expression in tumour stroma was linked to high malignant potential and disease recurrence in colorectal cancer (CRC) patients [96]. Interestingly, another study found that low vimentin expression in stroma and high vimentin expression in cancer cells was associated with prolonged overall survival (OS) in patients with ovarian tumours [97].

Vimentin plays diverse roles in EMT, focal adhesion, migration, invasion, and metastasis of cancer cells [98, 99], but knowledge on the function of vimentin in CAFs is limited. In a preclinical study employing a Cre-dependent LSL-KrasG12D/Lkb1fl/fl lung cancer model, vimentin was expressed in CAFs surrounding collective invasion packs of epithelial tumour cells, and whole-body vimentin knockout led to a reduction of invasion packs [100]. In a non-small cell lung cancer (NSCLC) model induced by LSL-KrasG12D/Tp53fl/fl, whole-body knockout of vimentin attenuated cancer-associated cachexia symptoms, inhibited tumour growth, and led to improved survival [101]. Vimentin also plays a direct role in cancer cells, as demonstrated by the observation of reduced cell motility in cancer cells upon vimentin knockdown [102, 103]. Future studies on the detailed function of Vimentin+ CAFs will be beneficial to understand their specific roles in cancer development.

Podoplanin (PDPN)

Podoplanin (PDPN) is a mucin-type protein with diverse physiological and pathological functions. PDPN-deficient mice displayed defects in blood-lymphatic vascular separation, impacting proper regulation of lymph flow [104, 105]. While high PDPN expression was predominantly found in lymphatic endothelium and often utilised as a marker for lymphatic vessels [106], elevated PDPN expression has been reported in CAFs and associated with poor outcomes in various cancer types, including lung [107], breast [108], and pancreatic cancers [109]. The roles of PDPN+ CAFs have been explored in several studies. In a collagen invasion assay involving co-cultured cancer cells and CAFs, PDPN+ CAFs created invasion tracks for lung cancer cells, and knockdown of PDPN in CAFs decreased invasion of both CAFs and cancer cells [110]. However, ectopic expression of PDPN in human fibroblasts did not affect the migratory and invasive properties of co-cultured breast cancer cells [111]. PDPN+ CAFs showed high expression of TGF-β and were associated with CD204+ TAM infiltration in stage-I lung squamous cell carcinoma, leading to the immunosuppressive TME [107]. Interestingly, PDPN+ CAFs also exhibited a tumour-inhibitory effect by suppressing the proliferation of small cell lung cancer (SCLC) cells in a co-culture assay [112]. Another study suggested an association between PDPN+ CAFs and prolonged disease-free survival (DFS) in CRC patients [113]. PDPN could also act as a co-inhibitory receptor on T cells, and T cell specific PDPN conditional knockout mice exhibited delayed tumour growth [114]. Macrophage-specific PDPN conditional knockout mice showed reduced lymph angiogenesis and lymph invasion in breast cancer [115]. Knockdown of PDPN in cancer cells also resulted in reduced cell proliferation, migration, and invasion [116,117,118], suggesting the intricate roles of PDPN in cancer development.

Fibroblast-specific protein 1 (FSP-1)

FSP-1, also known as S100A4, is a well-established marker for fibroblasts involved in tissue remodelling [119, 120]. Although FSP-1 is expressed in both CAFs and NFs, CAFs from cancer tissues generally exhibit more abundant FSP-1 expression than NFs from adjacent normal tissues [121]. Increased FSP-1 expression in CAFs was linked to EMT [122], and its presence was detected in inflammatory macrophages [123]. In CRC patients, high FSP-1 expression in CAFs was associated with tumour invasion [124]. Intriguingly, tumoral FSP-1 positivity and stromal FSP-1 negativity was correlated to short DFS and OS in patients with invasive lobular carcinoma [125].

FSP-1+ CAFs promote tumour metastasis by secreting factors such as VEGF-A and Tenascin-C, establishing an angiogenic microenvironment at metastatic sites and providing protection from apoptosis [126]. In addition, monocyte chemotactic protein-1 derived from FSP-1+ CAFs increased monocyte recruitment and inflammatory responses in a skin tumour model [127]. Mice with FSP-1 deficiency had decreased tumour incidence, and co-injection of FSP-1+ CAFs with mouse mammary carcinoma cells partially restored tumour development and metastasis [128]. However, depletion of FSP-1+ stromal cells did not prevent the development of hepatocellular carcinoma (HCC), although it reduced the stemness phenotype of tumours [129]. Head and neck squamous cell carcinoma (HNSCC) cells with FSP-1 knockdown exhibited reduced expression of matrix metalloproteinase 3 (MMP3), resulting in decreased invasiveness and metastasis in vivo [130]. The loss of FSP-1 in cancer cells resulted in increased ferroptosis and cell death upon the treatment of ferroptosis-inducing agent [131]. These findings suggest that FSP-1 could be a promising anti-tumour target, given its tumour-promoting roles in both CAFs and cancer cells.

Tenascin-C (TN-C)

TN-C, a glycoprotein interacting with ECM molecules like fibronectin [132], is abundantly expressed by CAFs and solid malignant tumours [133,134,135]. TN-C expression in the stroma of prostate cancer showed correlation with the expression of other CAF markers, including FSP-1, α-SMA, and vimentin [135]. In pancreatic cancer, TN-C in CAFs can enhance epithelial-to-mesenchymal transition and is associated with resistance to immune checkpoint inhibitors in patients [136]. High tumoral TN-C expression could be associated with tumour progression, metastasis, and poor prognosis in different cancer types [137,138,139,140]. TN-C produced by CAFs promoted metastasis of colon cancer cells in response to TGF-β signalling [133]. In an osteosarcoma xenograft model, TN-C contributed to lung metastasis by interacting with its receptor integrin α9β1 [141]. TN-C also promoted immune suppression by immobilising infiltrated T lymphocytes through chemokine (C-X-C motif) ligand 12 (CXCL12) signalling [142]. Moreover, TN-C increased infiltration of Treg cells, and ablation of TN-C inhibited immune-suppressive stromal properties in an OSCC model [143]. TN-C knockout mice exhibited increased immune cell infiltration and reduced tumorigenesis, tumour size, and tumour metastasis compared to wild type mice [143, 144]. Knockdown of TN-C in CAFs led to increased endothelial tubulogenesis of glioblastoma [145], and TN-C knockout in tumour cells reduces lymphoid immune suppression, migration, and invasion of osteosarcoma and OSCC [140, 143]. A recent study found that reducing TN-C expression in cancer cells can enhance the efficacy of inhibitors targeting the ErbB3, PI3K-AKT, Ras, and MAPK signalling pathways in oesophageal squamous cell carcinoma [146]. More in-depth exploration of TN-C roles in cancer has been reviewed by others [147, 148].

Periostin (POSTN)

POSTN is a secreted cell adhesion glycoprotein that serves as a ligand for integrins αVβ3 and αVβ5. High expression of POSTN in CAFs was associated with poor prognosis in various cancers, including breast [149], cervical [150], CRC [151, 152], oesophageal cancers (EAC) [153], and PDAC [154]. The colony number and spheroid size of CRC were significantly larger when co-cultured with Postn+/+ fibroblasts than when co-cultured with POSTN knockdown or knockout fibroblasts [151, 155]. When binding to integrin αVβ3, CAF-derived POSTN can activate PI3K/AKT signalling pathway, promoting EMT, migration and invasion of ovarian cancers and EAC [153, 156]. The ERK pathway can also be activated by CAF-derived POSTN, leading to enhanced proliferation, migration, and EMT of NSCLC and gastric cancer cells [157, 158]. Downregulating POSTN in the TME of PDAC reduced proliferation, metastasis, and clonality of PDAC cells [159]. POSTN also showed interaction with protein tyrosine kinase 7 in HNSCC to promote cancer stemness [160]. Through bindings to integrins αVβ3 and αVβ5, POSTN can activate the ERK/NF-κB signalling pathway in ovarian cancer cells, leading to increased expression of cytokines that promote macrophage mobility and polarization toward the M2 phenotype [161]. POSTN also induced expression of Programmed Cell Death Protein 1 (PD-1) on TAMs through integrin-ILK-NF-κB signalling, and PD-1+ TAMs, in turn, produced IL-6 and IFN-γ, leading to induction of Programmed Cell Death Ligand 1 (PD-L1) expression on CRC cells [162]. POSTN knockout (Postn−/−) mice exhibited reduced infiltration of PD-1-positive TAMs in CRC tumours [162] and displayed a lower tumorigenic potential [163, 164]. Intriguingly, Postn−/− mice demonstrated impaired capsule formation and enhanced tumour growth in another study [165]. Tumoural POSTN may also contribute to tumour growth and knockdown of POSTN in lung cancer cells repressed tumour growth in vivo [166]. These studies suggest the diverse roles of POSTN, underscoring its potential as a therapeutic target.

Galectin-1 (Gal-1)

Gal-1, a member β-galactoside-binding protein family, is ubiquitously expressed both intracellularly and extracellularly, despite lacking a secretion signal peptide [167]. Gal-1 plays a crucial role in cell–cell and cell–matrix adhesion in the TME, and CAF-derived Gal-1 induced metastasis, EMT, and angiogenesis in gastric cancer [168, 169]. TGF-β secreted from gastric cancer cells could transform NFs into CAFs by upregulating Gal-1 and α-SMA expression in fibroblasts [170]. Elevated Gal-1 expression in CAFs contributed to adaptive resistance to tyrosine kinase inhibitors (TKIs) of anaplastic lymphoma kinase in NSCLC [171]. Knockdown of Gal-1 in CAFs reduced the expression of monocyte chemoattractant protein-1 (MCP-1) and inhibited the progression of OSCC in vivo [172]. Gal-1 knockdown in CAFs also reduced migration and invasion of breast cancer cells by downregulating MMP9 expression [173]. Interestingly, tumour-derived Gal-1 increased frequency of CD4+CD25+Foxp3+ Treg cells in breast cancer [174], leading to an immunosuppressive TME. In addition, knockdown of Gal-1 in cancer cells reduced migration, invasion, and tumour growth [175, 176]. Gal-1 deficient mice exhibited impaired tumour growth due to inadequate tumour angiogenesis and a less immunosuppressive TME [177, 178]. In summary, Gal-1 serves as a marker for CAFs and presents itself as a promising target for cancer treatment.

Caveolin 1 (CAV1)

CAV1, a scaffolding protein crucial for the formation of caveolae, is involved in processes like endocytosis and receptor internalization [179], exhibiting both tumour-suppressive and tumour-promoting properties [180]. In the transition of NFs into CAFs, CAV1 expression was significantly downregulated, making it a negative marker for CAFs [181]. Low expression of CAV1 in CAFs was an independent predictor of poor prognosis in gastric cancer patients [182]. CAFs with reduced CAV1 expression exhibited an enhanced glycolytic phenotype, promoting migration and progression of PDAC [183]. Knockdown of CAV1 in fibroblasts not only promoted tumour growth but also increased chemoresistance in PDAC and HCC [184, 185]. CAV1 deficiency in CAFs also led to increased production and secretion of pro-inflammatory and tumour-enhancing cytokines, contributing to proliferation and invasion of gastric cancer cells [186]. Moreover, CAV1-deficient mice exhibited increased tumour permeability, angiogenesis, and growth in different tumour models [187, 188]. Interestingly, knockdown of CAV1 in cancer cells resulted in attenuated tumour growth, decreased proliferation, and impaired migration and invasion [189, 190], suggesting a tumour-promoting role of CAV1 in cancer cells. However, the detailed mechanisms of how CAV1 regulates cancer cells remain to be investigated.

Ephs/ephrins

Eph receptors and their membrane bound ephrin ligands control cell–cell interactions during development, including tissue boundary formation and patterning of the neural and vascular systems, and are often up-regulated in tumours and the TME, including on CAFs [191, 192]. Several Eph receptors have been identified as elevated in stromal cells from human gastric tumours compared to those from normal tissues, and expression of EphA2 was associated with poor prognosis [193]. In co-culture assays including CAFs and cancer cells, tyrosine phosphorylation of EphA2 on CAFs was increased, which led to enhanced invasiveness of cancer cells [194, 195]. Similarly, ephrin-B on fibroblasts was found to increase invasiveness of EphB3/4-expressing prostate cancer cells [196]. EphA3 was identified to be widely expressed in the stroma of diverse cancer types, present on MSCs and specific CAF subtypes in human and mouse tumours [197, 198]. Antibody targeting [197] or knock-down of TME-expressed EphA3 [198] decreased angiogenesis and tumour growth. In breast cancer, EphA3 was identified on both cancer cells (upregulated by RAGE signalling), and on CAFs, and its activity promoted invasion, which was blocked by a specific EphA3 inhibitor [199]. Ephrin-A5 expression was identified on pancreatic CAFs and thought to mediate interaction with EphA receptors on cancer cells, as well as on other CAFs, and to promote collagen synthesis [200]. Ephrin-B2 expressed on lung and pancreatic myofibroblasts was found to be shed by the transmembrane metalloprotease ADAM10, leading to fibroblast activation and fibrosis, and inducing EphB4 signalling in pancreatic cancer cells [201, 202]. Multiple ephrin-Bs were similarly found to be elevated in prostate CAFs and to promote CAF activation, cancer cell proliferation, and tumorigenicity in vivo [203]. Thus, while Eph and ephrin expression in tumour cells can have both tumour suppressive and promoting roles [191], their expression in CAFs appears exclusively tumour-promoting.

CAF subtypes

The heterogeneity of CAFs is supported by several key findings. Firstly, the molecular markers employed for CAF identification are diverse and lack complete specificity, often failing to encompass the entire CAF population. Minimal co-localisation of commonly used CAF markers, such as FSP1, αSMA, and PDGFRβ, was observed in tumour stroma of pancreatic and breast cancer mouse models, highlighting the inability of these markers to represent all CAFs in isolation [212]. Secondly, attempts to antagonise CAFs to reduce tumour burden have yielded contradictory outcomes, emphasising the intricate roles of CAFs, which are potentially associated to their heterogeneity. The growing recognition of CAF heterogeneity has encouraged extensive investigations on CAF subtypes that play tumour-suppressive and tumour-promoting roles in the TME (Fig. 2). The advance of scRNA-seq technology has also facilitated the identification and stratification of CAF subtypes in different cancers (Table 2). It is important to note, however, that there are no definitive factors to clearly stratify the pro-tumour or anti-tumour functions of CAFs.

Fig. 2
figure 2

Tumour-suppressive and tumour-promoting roles of CAF subtypes in TME. CAF: cancer-associated fibroblast; myCAF: myofibroblastic CAF; iCAF: inflammatory CAF; apCAF: antigen-presenting CAF; Treg: regulatory T cell; NK cell: natural killer cell; CSC: cancer stem cell; ECM: extracellular matrix. The figure was generated using BioRender

Table 2 Major CAF subtypes in different cancers

Myofibroblastic CAFs (myCAFs)

Öhlund et al. firstly identified two distinct subpopulations of CAFs, including myCAFs and inflammatory CAFs (iCAFs) in pancreatic cancer [213]. The myCAFs, characterised by high α-SMA expression and low IL-6 expression (α-SMAhighIL -6low) phenotype, were in close proximity to neoplastic cells, forming a structural ring surrounding clusters of cancer cells. These myCAFs exhibited an upregulation of TGF-β response targets such as CTGF and COL1A1. A subsequent study found that TGF-β secreted by PDAC cells contributes to the generation of myCAFs by downregulating IL1R1 expression [214].

One distinctive feature of myCAFs is their high contractility and ability to synthesise key ECM proteins like collagens [215]. This unique trait is believed to contribute to tumour tissue stiffness, creating a physical barrier that constrain tumour growth and impact treatment efficacy. Depletion of α-SMA+ myCAFs in a transgenic murine PDAC model resulted in increased invasion, induction of EMT, emergence of stem-like properties, reduced overall survival (OS), and elevated presence of CD4+Foxp3+ Treg cells [67]. Absence of α-SMA+ myCAFs in the PDAC model also rendered tumours unresponsive to gemcitabine treatment. However, administration of anti-CTLA4 therapy showed potential in slowing disease progression and extend survival in the α-SMA+ myCAFs depleted model. Hedgehog (HH) signalling is generally activated in myCAFs, and depletion of SHH (a HH ligand) in PDAC tumours led to reduced stroma content but more aggressive cancer with increased vascularity [68]. These studies highlight the intricate relationship between myCAFs and tumour growth in PDAC. Although the ECM established by myCAFs may impede drug delivery, the physical barrier and stiffness could act as constraints against tumour growth. Consequently, therapeutic strategies targeting myCAFs in PDAC should be carefully considered.

Precision in therapeutic approaches targeting CAFs is crucial, as strategies focusing on CAF depletion may inadvertently lead to loss of other tumour-suppressive cells, potentially exacerbating tumour aggressiveness. Krishnamurty et al. revealed that markers like α-SMA and FAP were expressed in multiple stromal cell types, including fibroblasts and pericytes in both murine PDAC tumours and normal tissues [216]. Recently, a study demonstrated that leucine rich repeat containing 15 (LRRC15) displayed a more specific expression pattern in myCAFs, and targeted depletion of LRRC15+ myCAFs resulted in a substantial 70% reduction in overall PDPN+ CAFs, significantly attenuating PDAC tumour growth [216]. This selective depletion prompted a transformation of the remaining CAFs within PDAC tumours into a more universally fibroblast-like state. Moreover, elimination of LRRC15+ myCAFs enhanced the function of CD8+ T cells, rendering them more effective in response to anti-PD-L1 treatment. Taken together, myCAFs remain to be a promising and feasible therapeutic target for PDAC when approached with precision in target selection.

The presence of myCAFs has also been documented in breast cancer, and these myCAFs may contribute to immunosuppression and resistance to immunotherapy [217,218,219]. The myCAFs in breast cancer exhibited increased secretion and alignment of collagens, which could promote tumour growth and invasion [217]. A recent study suggested that myCAFs in breast cancer may originate from a specific subset of fibroblasts known as CD26 NFs [220]. The roles of myCAFs in many other cancers have also been studied, revealing diverse functions in different cancer types [221,222,223,224,225]. In castration-resistant prostate cancer (CRPC), SPP1 (Secreted Phosphoprotein 1)+ myCAFs were shown to promote resistance to androgen deprivation therapy via paracrine activation of the ERK signalling pathway [226]. In lung cancer, myCAFs marked by FAP and α-SMA expression exhibited a high level of fibrillar collagens, contributing to the formation of a dense ECM that can restrict the motility of T cells [225]. In gastric cancer, a specific subtype of myCAFs characterised by IGFBP7 expression enhanced cancer cell metastasis and stemness [227]. In squamous cell carcinoma (SCC), both myCAFs and iCAFs were involved in secretion of collagens and fibronectin 1, which can interact with CD44 on SCC keratinocytes and lead to increased cancer cell proliferation and invasion by activating PI3K/AKT and Src/MAPK signalling pathways [228]. Using scRNA-Seq and spatial analysis, Davidson et al. demonstrated that myCAFs were in close proximity to and strongly interacted with mesenchymal-like clear cell renal cell carcinoma (ccRCC) within primary tumours and metastatic sites [229]. This interaction promoted cancer invasion through secretion of multiple ligands acting on cancer cells. In liver cancer, myCAFs secrete hyaluronan by overexpressing hyaluronan synthase 2 (HAS2), leading to increased tumour growth [230]. Conditional knockout of Has2 in CAFs resulted in reduced hyaluronan production and, consequently, smaller tumour sizes in preclinical models. Interestingly, blocking tumoral CD44 receptor for hyaluronan did not inhibit cancer development, suggesting potential interactions of hyaluronan with non-tumour cells or other receptors [230]. In another study, depletion of myCAFs was found to reduce tumour growth and mortality in desmoplastic CRC and pancreatic metastasis [231]. In summary, myCAFs demonstrated both tumour-promoting and tumour-suppressive effects. Future studies should focus on developing treatments that can diminish their tumour-promoting effects while preserving tumour-suppressive functions.

Inflammatory CAFs (iCAFs)

In PDAC, iCAFs were characterized by low α-SMA expression and high levels of inflammatory cytokines, such as IL-6 and leukemia inhibitory factor [213]. These cells exhibited a loss of myofibroblastic features and are typically situated at a distance from cancer cells [213]. Activation of the JAK/STAT pathway by tumour-derived IL-1 is recognised as a driver for the formation of iCAFs [214]. Hypoxia in the TME could also contribute to the generation of iCAFs, resulting in their enrichment in hypoxic tumour regions [232, 233]. In addition, the formation of iCAFs can be induced by IL-17A derived from a specific subpopulation of CD8+ T cells, known as Tc17 cells [234]. Compared to untreated patients, pancreatic cancer patients resistant to chemotherapy exhibited high levels of iCAFs in tumour stroma, indicating a role of iCAFs in chemoresistance [235]. Zhang et al. also reported an increased population of iCAFs in chemo-resistant PDAC patients following chemotherapy, while the abundance of myCAFs remained unchanged [236]. Despite the predominant pro-tumorigenic roles attributed to iCAFs in various studies, a cluster of tumour-restrictive iCAFs characterised by high expression of osteoglycin was identified [237]. In PDAC patients, iCAF-derived osteoglycin serves as a favourable prognostic biomarker for OS [237].

The pro-tumorigenic roles of iCAFs extend beyond pancreatic cancer and are reported in other cancer types. In breast cancer, iCAFs recruit myeloid cells in a CXCL12-dependent manner and enhance MMP activity, ultimately leading to increased tumour invasion [220]. The spatial distribution of iCAFs in breast cancer mirrored that observed in pancreatic cancer, positioning iCAFs relatively distal to the invasive tumour surface [217]. The abundance of iCAFs in breast tumour tissues correlated with the infiltration of Treg cells as well as dysfunction of cytotoxic T-lymphocytes [217]. Interestingly, the iCAF-like fibroblasts characterised by PDGFRβ+α-SMAlowCD34highCD146 was also abundantly detected in the surrounding ductal regions of healthy breast tissue [217]. In liver cancer, iCAFs showed high expression of HGF, promoting tumour growth via the HGF-MET axis [230]. Conditional depletion of HGF in CAFs resulted in decreased development of liver cancer, and depletion of the HGF receptor MET in hepatocytes or tumour compartments reduced tumour growth [230]. In gastric cancer, iCAFs were enriched with pro-stemness-associated pathways, including NF-κB signalling, TNF signalling, and cytokine-receptor interaction pathways, implying their involvement in cancer stemness [238]. Moreover, iCAFs showed interaction with surrounding T cells by secreting IL-6 and CXCL12, leading to establishment of a tumour-favourable microenvironment in gastric cancer [239]. In CRC, fibroblast growth factor 19 (FGF19) derived from tumour cells can induce the formation of iCAFs through the FGFR4-JAK2-STAT3 pathway [240]. These iCAFs subsequently promoted liver metastasis by increasing neutrophil infiltration and the formation of neutrophil extracellular traps in liver metastatic niches. Some iCAFs showed high expression of IL1R1 and addition of an IL-1-inhbiting antibody effectively reduced tumour spheroid growth [241]. Elevated levels of IL1R1 in CRC cancer patients were correlated with an increased expression of T cell exhaustion markers like LAG3, as well as immunoregulatory proteins such as PD-L1 and PD-L2 [241]. In bladder cancer, iCAFs express a variety of growth factors that contribute to angiogenesis, cancer cell proliferation, and chemoresistance [242]. In preclinical models of PDAC, while ablation of the tumour-restrictive α-SMA+ CAFs reduced survival, depletion of FAP+ CAFs significantly improved survival and enhanced the efficacy of immune checkpoint inhibitors (ICIs) [243]. Further research is needed to assess the efficacy of iCAF-targeted therapies.

Antigen-presenting CAFs (apCAFs)

In addition to the two predominant CAF subtypes described above, scRNA-seq studies have identified a less common cluster of CAFs characterised by high expression of MHC-II genes and CD74 [244]. These distinct CAFs were designated as apCAFs due to their unique ability to activate CD4+ T cells in an antigen-specific manner. It should be noted apCAFs are not abundant and only sporadically detected in most cancers. In pancreatic cancer, apCAFs may originate from mesothelial cells, a transformation induced by IL-1 and TGF-β. These apCAFs can directly interact with naive CD4+ T cells, resulting in formation of Treg cells [245]. The use of a blocking antibody targeting mesothelin, a marker associated with mesothelial cells, can effectively inhibit the transition of mesothelial cells into apCAFs, leading to reduced Treg cells and attenuated tumour growth [245]. However, in lung cancer, apCAFs showed a distinct role by directly activating T cell receptors on adjacent effector CD4+ T cells and producing C1q to rescue these T cells from apoptosis [246]. Deletion of MHC-II to reduce apCAFs led to increased tumour burden, reduced survival rates, and fewer infiltrated T cells, implying a tumour-suppressive effect of apCAFs in lung cancer [246]. Due to the low abundance of apCAFs, their roles in cancer development have not been extensively studied. More studies will be required to examine their therapeutic potential.

Other CAF subtypes

Despite the initial classification of myCAF, iCAF, and apCAF subtypes in pancreatic cancer, researchers have also identified different CAF subtypes. In PDAC, CAFs expressing Meflin were associated with a better response to chemotherapy, and inducing Meflin expression in CAFs could enhance sensitivity of PDAC tumours to gemcitabine [247]. These Meflin+ CAFs were referred as "rCAFs" due to their capacity to restrain tumour growth. Further investigations revealed that Meflin directly inhibits lysyl oxidase, an enzyme responsible for crosslinking collagen and elastin, contributing to tissue stiffness and increased interstitial pressure [247]. The advancements in scRNA-seq technology have facilitated a more comprehensive analysis of CAF subtypes, especially in cases where a sufficient number of stromal cells are available. For instance, Cords et al. conducted an in-depth analysis of CAF subtypes by employing scRNA-seq on over 16,000 stromal cells obtained from 14 breast cancer patients [248], leading to identification of nine distinct CAF subtypes, each characterized by unique molecular signatures and functions. In cribriform prostate cancer, a specific subtype of CAFs characterized by the CTHRC1+ASPN+FAP+ENG+ signature was referred to as "CAFÉ CAFs", which was associated with an immunosuppressive TME [249]. In lung cancer, a subpopulation CAFs characterised by ZIP1+FSP1+CX43high, known as "zCAFs", can absorb and transfer Zn2+ to neighbouring cancer cells via gap junctions, leading to chemoresistance [250]. Another subtype of CAFs with MYH11+α-SMA+CD34+FAPADH1B signature was associated with reduced infiltration of CD3+ and CD8+ T cells, contributing to immune exclusion within tumour nests [225]. Several other CAF subtypes have been characterized in different cancers, and these subtypes have been reviewed extensively by other researchers [251,252,253].

Normal fibroblasts (NFs)

NFs are widely distributed in many healthy organs and tissues, where they play crucial roles in development, homeostasis, injury repair, and normal signalling. NFs secrete structural macromolecules, such as collagen, contributing to the synthesis, remodelling, and maintenance of the ECM [254]. In addition, NFs serve as a rich source of signalling molecules, including growth factors, cytokines, and chemokines, which act on other cells to regulate development and other biological processes [255]. In response to tissue damage, NFs can rapidly expand to produce more ECM-secreting fibroblasts that are critical to tissue synthesis, as well as myofibroblasts with high expression of contractile proteins such as α-SMA [256]. Tissue-specific fibroblasts have been found organs such as skin, lung, colon, skeletal muscle, and heart, where they support organ development and homeostasis [257, 258]. Various molecular markers have been reported to identify NFs, with widely used pan-fibroblast markers including CD90, PDGFRα/β, vimentin, and collagens [259]. Markers and genes enriched in specific NF subtypes have been reviewed by elsewhere [259,260,261]. It is important to note, however, that some NF markers are also expressed by other cell types, such as the high PDGFRβ expression found in pericytes and smooth muscle cells [262]. Given that CAFs can originate from NFs, there is substantial overlap in markers between these two fibroblast types, making it critical to approach CAF analysis with caution to avoid NF contamination.

Directly targeting CAFs

Given the pivotal roles of CAFs in cancer, various strategies have been proposed to develop therapeutic interventions targeting CAFs. These approaches primarily involve CAF elimination, reprogramming, and targeting functional factors originating from CAFs. It is essential to note that many treatments targeting CAFs do not exhibit direct inhibitory effects on cancer cells. Consequently, CAF-targeted therapies are often combined with other approaches against tumours, aiming to synergistically enhance their therapeutic efficacy. Proteins that are highly expressed by CAFs and play tumour-promoting roles are considered as attractive therapeutic targets. A common treatment strategy is to inhibit functions of these targets by using small molecular inhibitors or blocking antibodies (Fig. 3; Table 3).

Fig. 3
figure 3

Treatments directly targeting CAFs currently in clinical trials. CAF, cancer-associated fibroblasts. Cell surface proteins and intracellular proteins highly expressed by CAFs have been targeted by various drugs. The figure was generated using BioRender

Table 3 Clinical trials for therapies directly targeting CAFs

FAP

Besides serving as a marker for CAFs, FAP is one of the most promising targets on CAFs, owing to its important roles and high expression in both CAFs and epithelial cells. Therapeutic treatments targeting FAP+ CAFs have shown capability to alleviate immunosuppression and enhance responses to ICIs. For instance, an adenoviral-vector vaccine designed to eliminate FAP+ cells reduced the number and suppressive function of immunosuppressive cells within tumours, concurrently inducing a robust CD8+ T cell response [263]. Talabostat, a small molecule dipeptidyl peptidase inhibitor of FAP, exhibited anti-tumour activity primarily through induction of tumour-specific cytotoxic T lymphocytes [264]. The introduction of CAR-T cells designed to target FAP also showed promising therapeutic outcomes in murine models [243, 265,266,267]. In addition, several studies explored targeted delivery of radioisotopes or drugs to tumours by using anti-FAP antibodies, resulting in therapeutic regressions in preclinical cancer models [268,269,270].

A number of treatments targeting FAP+ CAFs have progressed into clinical trials. The humanized murine anti-FAP monoclonal antibody F19 [271], known as Sibrotuzumab, showed a specific FAP targeting effect in cancer patients [272]. However, this antibody treatment alone did not demonstrate substantial therapeutic benefit in patients with metastatic CRC [273]. The FAP inhibitor talabostat also did not achieve significant therapeutic effect in the clinic either as a monotherapy or in combination with chemotherapy [274, 275]. Currently, an ongoing clinical investigation is exploring the combination of talabostat with immunotherapy [276, 277]. An anti-FAP bispecific antibody linked to IL-2v (RO6874281) has been assessed in a Phase I trial, showing objective responses in some patients [278]. Another bispecific antibody, targeting both FAP and DR5, displayed strong anti-tumour efficacy in preclinical models [279] and is currently under clinical evaluation [280]. Other anti-FAP bispecific antibodies featured with FAP-targeting and immunomodulatory effects have also been developed [281]. The exploration of CAR-T cell therapies target FAP is underway in clinical trials too [282].

Recent developments in targeted delivery of radionuclide to tumours by anti-FAP peptides or inhibitors have shown great promise [283, 284]. Several peptide-based FAP inhibitors (FAPI) with high affinities and selective binding to FAP-expressing tumours have been developed. Radiolabelled FAPI with 177Lu exhibited promising efficacy in preclinical cancer models [285,286,287], leading to the assessment of 177Lu-FAPI in clinical trials. Safety profiles of 177Lu-FAPI have been established in several phase I studies, and anti-tumour effects were observed in cancer patients [284, 288]. In a dose-escalation study for treating patients with metastatic radioiodine refractory thyroid cancer, 177Lu-FAPI demonstrated promising therapeutic efficacy, with a disease control rate (DCR) of 83% and objective response rate (ORR) of 25% [289]. More ongoing clinical trials are in progress to evaluate the efficacy of different 177Lu-FAPI treatments. In the future, it will be interesting to explore the therapeutic effect of combining 177Lu-FAPI with established anti-tumour therapies like immunotherapy and chemotherapy.

PDGFRα/β

High expression of PDGFRα/β has been observed in CAFs, vascular cells, and malignant cells, where these receptors play crucial roles in shaping an immunosuppressive TME, promoting angiogenesis, facilitating tumour growth, and fostering metastasis [290]. Therapeutic interventions targeting PDGFRα/β demonstrated potential to enhance immunotherapies in murine models, prompting further exploration in clinical settings [291, 292].

Several small molecule inhibitors targeting the PDGF/PDGFR pathway have been developed. Imatinib, a TKI targeting PDGFR, c-Kit, and BCR-ABL, has been approved by the U.S. Food and Drug Administration (FDA) for treating several cancers. Clinical trials are currently exploring the combined use of imatinib with immunotherapies. While a combination of imatinib and ipilimumab was well-tolerated in cancer patients, it did not exhibit a synergistic effect [293]. Efficacy studies involving the combination of imatinib with other ICIs, such as atezolizumab and pembrolizumab, are currently underway [294]. Other multi-target TKIs for PDGFR, including regorafenib, sunitinib, ripretinib, and avapritinib, have received FDA approval for treating gastrointestinal stromal tumours (GIST) [295]. These TKIs are also being evaluated as combination treatments with immunotherapies or targeted therapies in different cancers.

In addition to small molecule inhibitors, there is ongoing development of antibodies targeting PDGFR. The combination of the anti-PDGFRα antibody olaratumab with doxorubicin did not yield a significant improvement in OS for sarcoma patients compared to the placebo plus doxorubicin treatment [296]. The combination of olaratumab with pembrolizumab was well-tolerated, with a reported disease control rate (DCR) of 53.6% in a phase I trial [297]. Bispecific antibodies concurrently binding to PDGFR and other targets also showed promising results in preclinical studies [298, 299], while their clinical effectiveness remain to be investigated.

Hedgehog (HH) signalling

Activation of the HH signalling pathway in CAFs promoted tumorigenesis and metastasis in preclinical studies [300, 301]. In contrast, inhibition of the HH signalling pathway using the specific inhibitor sonidegib reduced the myCAF/iCAF ratio and impeded tumour growth [302]. Another HH inhibitor, vismodegib, also exhibited inhibitory effects on tumour growth in preclinical models [303]. These promising findings led to the evaluation of HH inhibitors in clinical trials. Sonidegib demonstrated robust efficacy in a phase II trial involving patients with basal cell carcinoma [304]. The combination of sonidegib and chemotherapy showed anti-tumour activity in triple-negative breast cancer (TNBC) patients [305]. Currently, the efficacy of combining sonidegib with pembrolizumab is under investigation for treating NSCLC [306]. Vismodegib, as a monotherapy, achieved a 71% ORR in basal cell carcinoma patients [307] and a DCR of 5.3% in gastric cancer patients [308]. In newly diagnosed metastatic pancreatic cancer patients, the combination of vismodegib and chemotherapy did not enhance efficacy of chemotherapy [309]. To facilitate future clinical trials for therapies targeting HH signalling, it will be beneficial to develop diagnostic approaches to evaluate HH signalling activation in cancer patients.

Other CAF targets

Other proteins highly expressed by CAFs have also been targeted in preclinical studies and clinical trials. For instance, niclosamide functioning as a FSP-1 transcriptional inhibitor, demonstrated potential in reducing liver metastasis of colon cancer and boosting efficacy of ICIs in preclinical models [310, 311]. Niclosamide exhibited favourable tolerability in patients and is being evaluated in a phase II trial for CRC [312]. Neutralising antibodies targeting PDPN showed inhibition of tumour growth and metastasis in xenograft models for osteosarcoma, oral cancer, and malignant pleural mesothelioma (MPM) [313,314,315], supporting clinical assessment of anti-PDPN antibodies in the future. The tumour-promoting characteristic of CAFs with high LRRC15 expression has inspired the development of LRRC15-targeted therapies. ABBV-085, a monomethyl auristatin-E (MMAE) antibody–drug conjugate targeting LRRC15, demonstrated anti-tumour efficacy in preclinical models [316]. The safety and tolerability of ABBV-085 in patients were assessed, with reported anti-tumour responses in sarcoma patients [317]. CAFs with elevated IL-1R expression were shown to promote tumour development and induce an immunosuppressive TME [241]. Anakinra, an FDA-approved IL-1R antagonist for treating rheumatoid arthritis, showed potential to reduce CAF-derived thymic stromal lymphopoietin, which correlates with poor survival rates in pancreatic cancer patients [318]. Encouragingly, combination of anakinra with 5-FU and bevacizumab has shown promise in treating patients with refractory CRC [319]. Currently, a phase Ib/II clinical trial is underway to explore the combination of anakinra with CAR-T therapy for the management of relapsed multiple myeloma [320]. As many of these targets may not be widely expressed in all cancer patients, a stringent selection of cancer patients for clinical trials is essential for future studies.

Targeting CAF-derived factors

Many tumour-promoting factors derived from CAFs have been identified in the past, positioning them as promising targets for therapeutic interventions. These factors can either directly interact with cancer cells to regulate tumour actions, or affect other stromal components like immune cells. Many clinical trials have been carried out to assess the efficacy of drugs targeting these CAF-derived factors (Fig. 4; Table 4).

Fig. 4
figure 4

Drugs targeting CAF-derived factors that promote tumour development. CAF: cancer-associated fibroblasts. The figure was generated using BioRender

Table 4 Clinical trials for therapies targeting CAF-derived factors and corresponding receptors

TGF-β

CAF-mediated TGF-β signalling pathway is involved in the crosstalk between CAFs and cancer cells. Activation of the TGF-β signalling pathway in cancer cells can increase proliferation, migration, invasion, immunosuppression, and therapy resistance. By inhibiting activation of latent TGF-β1, the agent SRK-181-mIgG1 can sensitise tumour response to anti-PD-1 treatment in preclinical models, without causing evident toxicities [323, 324]. In a phase I study, SRK-181 exhibited no dose-limiting toxicity when administered as a monotherapy or in combination with pembrolizumab [325], while the efficacy remains to be explored. Fresolimumab, a neutralizing monoclonal antibody for all TGF-β isoforms, exhibited good tolerance and anti-tumour activity in a phase I trial [326]. However, its immunoregulatory effects were found to be minimal in a subsequent phase II study [327]. An imaging study utilizing 89Zr radiolabelled fresolimumab demonstrated good penetration into recurrent high-grade gliomas, but the antibody did not yield clinical benefits, leading to discontinuation of further development for oncology indications [328]. Another anti-TGF-β monoclonal antibody SAR439459 demonstrated a synergistic effect with PD-1 blockade, enhancing anti-tumour immunity in a preclinical study [329]. Unfortunately, a recent study revealed a lack of efficacy and a notable risk of bleeding in cancer patients treated with this drug, resulting in termination of the trial [330].

An alternative strategy for targeting TGF-β involves designing ligand traps. AVID200, a receptor ectodomain trap computationally designed to target TGF-β1/3, increased T-cell-mediated cytotoxicity and enhanced the efficacy of ICIs in syngeneic preclinical models [331]. The safety prolife of AVID200 is currently under clinical evaluation [332]. Bifunctional molecules containing TGF-β traps have also been developed, and one notable example is M7824 that combines the TGF-βRII receptor (acting as a trap) with an anti-PD-L1 IgG1 [333]. Preclinical studies have demonstrated the tumour-targeting effect and anti-tumour efficacy of M7824 [333, 334]. However, a phase III clinical trial was terminated due to a lack of superior efficacy compared to pembrolizumab [335]. Another bifunctional TGF-β trap fused drug, anti-CTLA4-TGF-βRII, showed superior anti-tumour efficacy compared to an anti-CTLA4 antibody alone in preclinical models [336], but its efficacy in patients has not been investigated.

The cytoplasmic kinase activity of TGF-β receptors can also be targeted for cancer therapy. Several small molecule receptor kinase inhibitors have been developed for this purpose and are currently in clinical trials [337,338,339,340,341,342]. For example, vactosertib, an orally bioavailable TGF-β receptor kinase inhibitor, showed efficacy against multiple myeloma in preclinical models, either as a monotherapy or in combination with other treatments [343, 344], leading to the clinical assessment of vactosertib. Similar drugs such as galunisertib and LY3200882 are under clinical investigation.

IL-6

CAF-derived IL-6 contributes to cancer invasion, metastasis, angiogenesis, immune modulation, and drug resistance. Several drugs targeting IL-6 or the IL-6 receptor (IL-6R) received FDA approval for treating inflammatory diseases like rheumatoid arthritis [345]. Recently, their potential in cancer therapy has attracted attention, with observed anti-tumour efficacy in preclinical models [346, 347]. One such drug, siltuximab, a chimeric anti-IL-6 antagonistic antibody, received FDA approval for treating multicentric Castleman disease and is currently being investigated for treating cancers. In patients with castration-resistant prostate cancer (CRPC), elevated baseline IL-6 was correlated with poor survival, and siltuximab treatment resulted in a 23% stable disease (SD) rate [348]. Another anti-IL-6 antibody, clazakizumab, improved cancer cachexia in NSCLC patients, as shown by biomarker analysis [349]. In preclinical models resistant to anti-PD-L1 treatment, dual blockade of IL-6R and PD-L1 attenuated tumour growth and improved survival [350, 351], leading to clinical evaluation of this combination therapy. A combination of siltuximab and spartalizumab is currently in a phase Ib/II trial for metastatic pancreatic cancer [352]. Combination of siltuximab with chemotherapies achieved an impressive ORR of 90.9% in patients with untreated multiple myeloma [353]. Tocilizumab, an anti-IL-6R humanized monoclonal antibody, is also under clinical investigation in combination with ICIs.

Other interleukins

In addition to IL-6, CAFs can produce many other interleukins, including IL-10, IL-11, IL-22, IL-32, and inhibiting actions of these interleukins resulted in anti-tumour effects in some studies [22, 354,355,356]. For instance, neutralising IL-10 with an antibody potentiated anti-tumour immune reaction in a preclinical model mimicking human CRC liver metastases [357]. Interestingly, overexpression of IL-10 or administration of pegylated IL-10 in preclinical models also inhibited tumour growth [358]. Pegilodecakin, acting as an IL-10 receptor agonist, exhibited a notable 43% ORR in NSCLC patients when combined with nivolumab or pembrolizumab [359]. However, in another clinical study, addition of pegilodecakin failed to improve the efficacy of chemotherapy in advanced PDAC patients [360]. Therapeutics targeting IL-11/IL-11R signalling are recently developed, with a humanised anti-IL-11 antibody 9MW3811 currently in a phase I trial for treating solid tumours. Treatments targeting IL-22/IL-22R or IL-32/IL-32R signalling have not yet been developed.

CXC chemokines

CAFs secrete a range of C-X-C motif chemokine ligand (CXCL) family proteins that act on cancer cells and stromal cells, leading to increased tumour proliferation, metastasis, and immunosuppression. Preclinical studies have demonstrated great potential in targeting CXCL chemokines for cancer therapy. Inhibiting CAF-derived CXCL1 using antagonistic antibodies reversed radio-resistance in oesophageal squamous cell carcinoma xenograft models [361] and reduced growth of bladder cancer cells [362]. Another humanised monoclonal antibody NTC-001 neutralising CXCL1, is currently undergoing preclinical evaluation [363]. CAF-derived CXCL8 (also known as IL-8) can promote tumour resistance to cisplatin in gastric cancer [364]. An anti-CXCL8 neutralizing antibody BMS-986253, when combined with nivolumab, showed tolerable safety and resulted in partial response (PR) in cancer patients who had previously progressed after anti-PD-(L)1 or anti-CTLA-4 treatment [365]. The role of CXCL11 in tumour development is controversial. CAF-derived CXCL11 increased migration and metastasis of HCC [366], while cancer cell-secreted CXCL11 enhanced CD8+ T cell infiltration in a preclinical study [367]. Elevated levels of CXCL11 were associated with anti-tumour immune responses and improved prognosis in colon cancer [368]. CXCL12 secreted by CAFs contributes to tumour proliferation, invasion, metastasis, immunosuppression, and angiogenesis [369,370,371,372]. Combining a CXCL12 inhibitor, NOX-A12, with pembrolizumab induced immune response, resulting in SD in heavily pretreated cancer patients [373]. In addition, combining NOX-A12 with radiotherapy led to partial remission of target lesions in GBM patient [374]. While roles for CXCL2, CXCL3, CXCL5, and CXCL7 in cancers have been reported [46, 375, 376], specific treatments targeting these chemokines have not yet been developed. CAFs also produce CCL2 and CCL5, two other chemokine ligands promoting tumour growth and metastasis [49, 377]. BMS-813160, a dual antagonist targeting CCR2 and CCR5 (the receptors for CCL2 and CCL5), is currently under assessment for efficacy in combination with nivolumab [378].

Some treatments have been developed to target receptors of CXC ligands, considering the capacity of CXC receptors (CXCRs) in binding to multiple CXC ligands. For instance, CXCR2 is known to interact with seven CXCL proteins, including CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL7, and CXCL8 [379]. Several antagonists targeting CXCR2 are currently under clinical evaluation. AZD5069, a CXCR2 inhibitor, exhibited promising anti-tumour activity in patients with metastatic CRPC when combined with enzalutamide [380, 381]. Ongoing investigations are exploring the efficacy of AZD5069 in combination with ICIs. Additionally, some CXCR2 inhibitors, such as danirixin and elubrixin, which were initially developed for treating non-cancer diseases, are being repurposed for cancer treatment with encouraging prospects. CXCR1 as a receptor for CXCL6 and CXCL8, is also a promising target for cancer treatment. Reparixin, which was initially developed as a CXCR1/2 inhibitor to attenuate inflammatory responses in organ transplantation and tissue injury [382], demonstrated anti-tumour effects in preclinical models [383]. Unfortunately, a clinical trial assessing the efficacy of reparixin in treating TNBC was terminated due to lack of efficacy [384]. SX-682, another CXCR1/2 inhibitor, when combined with M7824 and CV301 (a vaccine for CEA and MUC1), resulted in disease controls in some patients but also caused grade 3 bleeding adverse effect [385]. Combination of SX-682 with other ICIs is currently assessed in phase I/II trials [386, 387]. CXCR4, the receptor for CXCL12, is also being targeted for cancer therapy in the clinic. AMD3100 as a CXCR4 antagonist was approved by FDA for autologous transplantation in patients with non-Hodgkin’s Lymphoma or multiple myeloma [388]. The combination of AMD3100 with bortezomib resulted in a clinical benefit rate of 60.6% and an ORR of 48.5% in pretreated multiple myeloma patients [389]. BL-8040, a cyclic peptide inhibitor for CXCR4, when combined with pembrolizumab and chemotherapy, demonstrated a DCR of 77% in pancreatic cancer patients [390]. However, another cyclic peptide inhibitor for CXCR4, LY2510924, did not improve the efficacy of sunitinib in patients with RCC [391], and was ineffective in SCLC patients [392]. Notably, an anti-CXCR4 antagonist antibody, ulocuplumab, resulted in a 55.2% ORR and a clinical benefit rate of 72.4% when combined with lenalidomide and dexamethasone [393].

In contrast to the tumour promoting CXCL proteins, some CXC chemokines exhibit anti-tumour activity. These chemokines are usually secreted by cancer cells or other stromal cells rather than CAFs. Notably, CXCL9 and CXCL10 inhibited tumour growth and enhanced the efficacy of ICIs in preclinical cancer models [394,395,396]. These findings has led to the development of NG-641, an oncolytic adenoviral vector engineered to encode four immunostimulatory transgenes, including CXCL9, CXCL10, IFNα, and a bispecific T cell activator antibody targeting both FAP and CD3 [397]. The safety profile of NG-641 is currently under phase I clinical assessment, with no result released at the current stage.

HGF

HGF produced by CAFs can activate the c-MET receptor tyrosine kinase on tumour cells, promoting tumour growth and metastasis [398]. The humanised anti-HGF antagonistic antibody ficlatuzumab did not yield clinical benefits as a monotherapy, but resulted in a 19% ORR in patients with HNSCC when combined with cetuximab [399]. In another study, combining ficlatuzumab with gefitinib showed no significant difference compared to gefitinib monotherapy [400]. Emibetuzumab, another anti-HGF antagonistic antibody, was well tolerated but achieved only 4.3% ORR in patients with MET-positive NSCLC [401]. Combining emibetuzumab with ramucirumab (an anti-VEGFR2 antibody) resulted in a 6.7% ORR and a 60% DCR in HCC patients [402].

The c-MET receptor tyrosine kinase was also targeted for treating different cancers. Unfortunately, rilotumumab, a c-MET targeting agent, failed to meet the primary endpoint and was associated with worse OS in a phase III study [403]. However, the combination of erlotinib and onartuzumab, another antagonistic antibody for c-MET, resulted in improvements in both progression-free survival and OS in MET-positive NSCLC patients [404]. The FDA has now approved capmatinib and tepotinib (two highly selective MET inhibitors) for treating metastatic NSCLC with MET exon 14 skipping [405]. Tepotinib monotherapy was also more effective than sorafenib (targeting VFGFR, PDGFR, c-Kit, RET) in treating HCC patients with MET-positive tumours [406]. The combination therapy of capmatinib and pembrolizumab was not well tolerated and did not enhance ICI efficacy in NSCLC patients [407]. These studies suggest that treatments targeting HGF-c-Met signalling may only be effective to a fraction of cancer patients that need to be carefully selected in future clinical trials.

FGF

FGF proteins secreted by tumour stromal cells interact with FGF receptors (FGFRs) on cancer cells, resulting in enhanced cancer cell growth [408]. Aberrant activation of FGFR in cancer has been observed and can occur through variants, gene fusion, and copy number amplification [409]. Considering the important roles of FGF/FGFR signalling in cancer, treatments targeting this signalling have been developed. An example is FP-1039, which serves as a FGF ligand trap consisting of a Fc region and extracellular domain of FGFR1. FP-1039 treatment showed a 36% ORR in MPM and a 47% ORR in NSCLC in a phase Ib study [410, 411]. Due to the versatility of FGFR in binding different FGFs, interventions have also been developed to inhibit actions of FGFR. Erdafitinib and pemigatinib, two TKIs targeting FGFR1-4 and FGFR1-3 respectively, obtained FDA approval for treating advanced urothelial cancer with FGFR2/3 genetic alterations and myeloid/lymphoid neoplasms with FGFR1 rearrangement [412]. In a phase II trial, erdafitinib demonstrated a 40% ORR in patients with advanced or metastatic urothelial cancer harbouring FGFR alterations [413]. Comparable results were reported in cholangiocarcinoma (CCA) patients with FGFR alterations, in which erdafitinib achieved a 40.9% ORR in a phase IIa study [414]. Pemigatinib, in comparation, resulted in a 35.5% ORR in CCA patients with FGFR2 fusions or rearrangements [415].

AZD4547, a selective inhibitor of FGFR1-3, showed a 10% ORR in patients with endocrine-resistant breast cancer [416], and a 5% ORR in solid tumours with aberrations in FGFR pathway [417]. Other selective inhibitors for FGFR have also been evaluated in the clinic, exhibiting variable efficacy [418,419,420,421,422,423]. Non-selective inhibitors targeting FGFR have also been explored in the clinic. For instance, dovitinib targeting FGFR1/3, VEGFR1/3, c-KIT, FLT, showed a 3.0% ORR and a 49.3% DCR in advanced and metastatic RCC [424]. Ponatinib, which targets FGFR1 and other tyrosine kinases, exhibited a 7% ORR in GIST with KIT mutations after the failure of TKI treatment [425]. Nintedanib, an FDA-approved drug targeting FGFR1-3, VEGFR1-3, PDGFRα/β, FLT3, could enhance the efficacy of docetaxel in NSCLC [426]. More non-selective FGFR inhibitors have been reported and summarised by others [409, 427].

Antagonistic antibodies targets FGFR have also been developed and evaluated. Bemarituzumab targeting FGFR2b achieved a 53% ORR in gastric cancer harbouring FGFR2 overexpression or amplification [428]. Another antibody targeting FGFR3 showed a 29.6% ORR when combined with pembrolizumab for treating metastatic urothelial cancer [429]. Recent advancements on FGFR targeted therapy also include two antibody–drug conjugates, BAY1187982 and LY3076226. The BAY1187982 targeting FGFR2 to deliver auristatin-based payloads, showed poor tolerability in a phase I trial, leading to termination of this study [430]. In contrast, LY3076226 targeting FGFR3 with a cleavable linker and the maytansine derivative DM4 payload, exhibited acceptable safety and tolerability, but no responses were observed [431]. In the future, the combination of these drugs with other treatments could be explored.

Hyaluronan (HA)

CAFs also produce high-molecular-mass polysaccharides like HA to regulate cancer behaviours [432, 433]. The HA forms substantial complexes with proteoglycans, contributing to increased tumour interstitial fluid pressure, which limits penetration of therapeutic treatments into tumours [434]. Enzymatic depletion of HA with a recombinant HA-degrading enzyme resulted in reduced tumour cell ECM, decreased interstitial fluid pressure, decompression of tumour vessels, increased tumour vascular area, inhibited tumour growth, and enhanced chemotherapy efficacy [435]. These findings promoted clinical investigation of a HA-degrading enzyme, PEGPH20, in combination with other anti-cancer therapies. Unfortunately, the combination of PEGPH20 with chemotherapy resulted in increased toxicity and decreased OS in general PDAC patients [436]. Another study involving PDAC patients with elevated HA levels showed that combining PEGPH20 with chemotherapy cannot improve OS and progression-free survival [437]. However, when combined with pembrolizumab, PEGPH20 improved OS in HA-high PDAC patients [438]. The combination of PEGPG20 with atezolizumab showed very limited activity in PDAC and no benefit in GC patients [439]. Interestingly, VCN-01, an oncolytic virus expressing hyaluronidase, showed encouraging clinical activity in PDAC, achieving an ORR of 50% in a phase I trial [440], implying that the delivery method for HA-degrading enzyme could make a difference in therapeutic outcomes. In contrast to the systemically delivery of PEGPH20, the VCN-01 has the unique capability to induce local tumour production of hyaluronidase, potentially resulting in a more targeted and effective distribution of the enzyme in tumours.

CAF reprogramming

Reprogramming activated CAFs into quiescent CAFs is another strategy for cancer therapy targeting CAFs in TME (Fig. 5; Table 5). This approach could be promising for treating pancreatic cancer, where ablation of CAFs unexpectedly accelerated tumour growth in preclinical models. All-Trans Retinoic Acid (ATRA) as a standard treatment for patients with acute promyelocytic leukemia, could transform activated CAFs into quiescent CAFs. In pancreatic cancer, ATRA binds to retinoic acid receptor beta on pancreatic stellate cells, suppressing ECM remodelling and inhibiting tumour cell invasion [441]. Combining ATRA with gemcitabine led to enhanced anti-tumour effect in KPC mice [442]. The combination of ATRA with gemcitabine-nab-paclitaxel was safe and well tolerated in PDAC patients, resulting in a median OS longer than previously reported for chemotherapy-only treatments [443]. In addition, the combination of ATRA with pembrolizumab exhibited an ORR of 71% and a 50% complete response in patients with metastatic melanoma [444]. Additional studies are required to investigate whether the addition of ATRA can augment the efficacy of other therapies in different cancer types.

Fig. 5
figure 5

Drugs aiming to reprogram activated CAFs into quiescent CAFs. The figure was generated using BioRender

Table 5 Clinical trials for therapies aiming to deactivate CAFs

Vitamin D treatment also showed potential to deactivate CAFs and reduce the production of tumour-promoting factors [445, 446]. In patient with early stage lung adenocarcinoma and low vitamin D level, vitamin D treatment resulted in improved relapse-free survival and OS [447]. Nonetheless, the vitamin D analogue seocalcitol failed to demonstrate any objective anti-tumour activity in advanced pancreatic cancer [448]. A phase II study also reported no improvement with vitamin D supplementation in addition to chemotherapy in CRC [449]. Minnelide, a plant-derived compound, showed ability to deactivate CAFs and anti-tumour efficacy in preclinical models for pancreatic and liver cancers [450, 451]. Combining minnelide with chemotherapy led to a synergistic effect in pancreatic cancer models [452]. Clinical studies involving minnelide are ongoing, and no results have been reported. Angiotensin receptor blockers can also potentially reprogram CAFs into a quiescent state, and targeted delivery of angiotensin receptor blockers to tumours enhanced efficacy of immunotherapy in preclinical models [453]. While these therapies have shown promising results, clinical studies are so far limited.

Conclusion and prospects

Significant progress has been made in the discovery and characterization of CAFs in the past. It is now widely acknowledged that CAFs play a pivotal role in tumour development and at least partially contribute to the failures of current anti-cancer therapies. Treatments targeting CAFs have been developed, and promising results have been observed in many preclinical studies. However, the translation of these CAF-targeted therapies into clinical interventions has proven challenging and has not been as successful as anticipated. A key obstacle is the absence of clinically relevant animal models to assess efficacy of CAF-targeted therapies. Unlike therapies directly targeting tumour cells, the effectiveness of CAF-targeted therapies largely depends on the microenvironment and the composition of tumour stroma in patients. Unfortunately, due to the complexity and heterogeneity of TME, these factors cannot be fully recapitulated in most preclinical models, leading to inconsistent outcomes of CAF-targeted therapies in animal models and patients. Cell line xenografts and allografts remain the most used models for examining CAF-targeted therapies in preclinical studies. To establish stromal abundant tumours in animal models, CAFs are often co-injected with tumour cells. However, the spatial distribution and phenotypes of these introduced CAFs may differ significantly from those observed in patients. Recent CAF classifications in cancer patients have identified diverse CAF subtypes with distinct functions, another complexity that many preclinical models fail to represent.

CAFs are a large and heterogeneous cell population within the intricate TME, playing complex roles in regulating tumour growth. Molecularly, CAFs interact with cancer cells and other stromal cells through secreted signalling molecules and receptors. They secrete a range of growth factors, chemokines, and cytokines that can directly affect receptors on cancer cells or other stromal cells, such as immune cells in the TME. Spatially, CAFs influence tumour growth by remodelling the ECM and forming physical barriers that impact tumour cell expansion and the infiltration of cells and treatments. These behaviours endow CAFs with multifaceted roles in cancers. The contribution of each characteristic to tumour growth may vary depending on the cancer type. For instance, in PDAC, the growth-inhibitory effect of the physical barrier formed by CAFs may outweigh the tumour-promoting effects of CAF-secreted factors. However, such physical barriers might also create niches that contribute to treatment resistance. Given the significant roles and high abundance of CAFs in tumours, targeting CAFs could be a potent strategy for treating cancers, especially when combined with other therapies. Nonetheless, treatment approaches should be carefully evaluated for different cancer types, and more innovative strategies are needed to eliminate their pro-tumour roles while preserving their tumour-restricting functions.

Current therapeutic approaches targeting CAFs primarily rely on utilisation of small inhibitors and antibodies. Nonetheless, these treatments exhibit a relatively modest inhibitory effect on CAFs, and resistance to such therapies could emerge over time. In response to these challenges, there has been a growing interest in using radioligand therapy or radiopharmaceutical therapy to deplete CAFs. These therapies have shown remarkable results in preclinical models, prompting the evaluation of treatments like 177Lu-FAPi in clinical settings. One notable advantage of radioligand therapy lies in its prolonged therapeutic effect, attributed to the long half-life of the delivered radioisotope. Moreover, the beta particle range of 177Lu enables these drugs to simultaneously inhibit growth of adjacent tumour cells [454]. This innovative approach presents a potential breakthrough in targeting CAFs with greater efficacy and sustained effects. Inspired by the development of 177Lu-FAPi, other radiopharmaceutical therapies targeting CAFs can be developed by radiolabelling existing CAF-targeted treatments with 177Lu or other suitable radioisotopes. These drugs may have superior CAF-ablating efficacy, as both the vehicle and carried radioisotopes contribute to the inhibition and depletion of CAFs.

Treatment strategies focusing on tumour-promoting factors derived from CAFs are appealing in scenarios where stromal barriers restrict cancer cell growth and movement. It is important to note, however, that targeting a single factor may only be successful in specific preclinical models and a limited subgroup of cancer patients where the specific factor plays a predominant role in promoting tumour growth. Given that CAFs can produce multiple tumour-promoting factors, these strategies are less likely to have a significant impact across broad cancer patients. The inhibitory effects of such therapies may be counterbalanced by increased expression of other tumour-promoting factors.

Most CAF-targeted therapies directly inhibit or regulate growth and behaviours of CAFs rather than tumours. Although these therapies can alter the TME and thereby affect tumour growth, their efficacy could be further enhanced when combined with other cancer treatments including chemotherapy, targeted therapy, and immunotherapy. However, the dosage, tolerability, and safety profiles of combination therapies should be carefully investigated. To reduce systemic toxicity caused by combination therapies and enhance tumour-specific targeting of the stromal cells, bispecific antibodies can be considered to concurrently target CAFs and cancer cells.

An expeditious approach for advancing development of CAF-targeted drugs is to repurpose existing non-cancer drugs already in clinical trials or approved by FDA. As activated fibroblasts in inflammatory conditions share similarities with CAFs, drugs with anti-fibrotic properties originally developed for conditions like idiopathic pulmonary fibrosis could be repurposed for inhibiting CAFs [455, 456]. This repurposing approach offers an accelerated pathway for developing CAF-targeted drugs, benefited from their established safety profiles and tolerability in other conditions.

While many clinical trials for CAF-targeted therapies primarily focus on patients with advanced and metastatic cancers, it will be worthwhile to explore the potential of these therapies in preventing cancer metastasis and relapse, as CAFs play essential roles in cancer cell dissemination and dormancy. Furthermore, assessing the feasibility of using CAF-targeted therapies as neoadjuvant treatments could open new avenues for future cancer treatment.

The accurate selection of patients is fundamental to ensuring the reliability and success of clinical trials for CAF-targeted therapies. Given the inherent heterogeneity of CAFs and individual variations, it is anticipated that these therapies will be effective in only a subset of cancer patients. Therefore, patients should be carefully selected based on reliable criteria, such as stroma-tumour ratio and target expression level.

Availability of data and materials

Not applicable.

Abbreviations

TME:

Tumour microenvironment

CAF:

Cancer-associated fibroblast

TAM:

Tumour-associated macrophage

Treg:

Regulatory T

ECM:

Extracellular matrix

scRNA-seq:

Single-cell RNA sequencing

EMT:

Epithelial-mesenchymal transition

KPC:

LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre

SAA3:

Serum Amyloid A3

LRCC15:

Leucine rich repeat containing 15

FAP:

Fibroblast activation protein

IFN:

Interferon

TNF:

Tumour necrosis factor

α-SMA:

α-Smooth muscle actin

CSC:

Cancer stem cell

HGF:

Hepatocyte growth factor

SHH:

Sonic hedgehog

PDGFR:

Platelet-derived growth factor receptor

NF:

Normal fibroblast

PDPN:

Podoplanin

FSP-1:

Fibroblast-specific protein 1

TN-C:

Tenascin-C

POSTN:

Periostin

PD-1:

Programmed cell death protein 1

PD-L1:

Programmed cell death ligand 1

Gal-1:

Galectin-1

TKI:

Tyrosine kinase inhibitor

CAV1:

Caveolin 1

myCAF:

Myofibroblastic CAF

iCAF:

Inflammatory CAF

apCAF:

Antigen-presenting CAF

HH:

Hedgehog

HAS2:

Hyaluronan synthase 2

FGF:

Fibroblast growth factor

FAPI:

FAP inhibitor

ICI:

Immune checkpoint inhibitor.

CXCL:

C-X-C motif chemokine ligand

CXCR:

CXC receptor

ATRA:

All-trans retinoic acid

OSCC:

Oral squamous cell carcinoma

HCC:

Hepatocellular carcinoma

PDAC:

Pancreatic ductal adenocarcinoma

GI:

Gastrointestinal

GBM:

Glioblastoma

CRC:

Colorectal cancer

NSCLC:

Non-small cell lung cancer

SCLC:

Small cell lung cancer

HNSCC:

Head and neck squamous cell carcinoma

EAC:

Oesophageal cancer

CRPC:

Castration-resistant prostate cancer

SCC:

Squamous cell carcinoma

ccRCC:

Clear cell renal cell carcinoma

TNBC:

Triple-negative breast cancer

MPM:

Malignant pleural mesothelioma

BCC:

Basal cell carcinoma

MM:

Multiple myeloma

CCA:

Cholangiocarcinoma

UCC:

Urothelial carcinoma

GIST:

Gastrointestinal stromal tumour

OS:

Overall survival

DFS:

Disease free survival

DCR:

Disease control rate

ORR:

Objective response rate

SD:

Stable disease

PR:

Partial response

PFS:

Progression free survival

References

  1. Paget S. The distribution of secondary growths in cancer of the breast. Cancer Metastasis Rev. 1989;8(2):98–101.

    PubMed  CAS  Google Scholar 

  2. Lei X, Lei Y, Li J-K, Du W-X, Li R-G, Yang J, et al. Immune cells within the tumor microenvironment: biological functions and roles in cancer immunotherapy. Cancer Lett. 2020;470:126–33.

    Article  PubMed  CAS  Google Scholar 

  3. Lu C, Liu Y, Ali NM, Zhang B, Cui X. The role of innate immune cells in the tumor microenvironment and research progress in anti-tumor therapy. Front Immunol. 2023;13:1039260.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Galli F, Aguilera JV, Palermo B, Markovic SN, Nisticò P, Signore A. Relevance of immune cell and tumor microenvironment imaging in the new era of immunotherapy. J Exp Clin Cancer Res. 2020;39(1):89.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Anjali B, Nishka B, Sneha M, Gurbind S, Sandeep Kumar Y, Aloukick KS. Role of various immune cells in the tumor microenvironment. Dis Res. 2023;3(1):30–40.

    Article  Google Scholar 

  6. de Visser KE, Joyce JA. The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell. 2023;41(3):374–403.

    Article  PubMed  Google Scholar 

  7. Sullivan L, Pacheco RR, Kmeid M, Chen A, Lee H. Tumor stroma ratio and its significance in locally advanced colorectal cancer. Curr Oncol. 2022;29(5):3232–41.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Almangush A, Alabi RO, Troiano G, Coletta RD, Salo T, Pirinen M, et al. Clinical significance of tumor-stroma ratio in head and neck cancer: a systematic review and meta-analysis. BMC Cancer. 2021;21(1):480.

    Article  PubMed  PubMed Central  Google Scholar 

  9. van Pelt GW, Sandberg TP, Morreau H, Gelderblom H, van Krieken J, Tollenaar R, et al. The tumour-stroma ratio in colon cancer: the biological role and its prognostic impact. Histopathology. 2018;73(2):197–206.

    Article  PubMed  Google Scholar 

  10. Wang K, Ma W, Wang J, Yu L, Zhang X, Wang Z, et al. Tumor-stroma ratio is an independent predictor for survival in esophageal squamous cell carcinoma. J Thorac Oncol. 2012;7(9):1457–61.

    Article  PubMed  Google Scholar 

  11. Kemi N, Eskuri M, Herva A, Leppänen J, Huhta H, Helminen O, et al. Tumour-stroma ratio and prognosis in gastric adenocarcinoma. Br J Cancer. 2018;119(4):435–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Hosaka K, Yang Y, Seki T, Fischer C, Dubey O, Fredlund E, et al. Pericyte-fibroblast transition promotes tumor growth and metastasis. Proc Natl Acad Sci USA. 2016;113(38):E5618-5627.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Zhao Y, Shen M, Wu L, Yang H, Yao Y, Yang Q, et al. Stromal cells in the tumor microenvironment: accomplices of tumor progression? Cell Death Dis. 2023;14(9):587.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Xu M, Zhang T, Xia R, Wei Y, Wei X. Targeting the tumor stroma for cancer therapy. Mol Cancer. 2022;21(1):208.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Czekay RP, Cheon DJ, Samarakoon R, Kutz SM, Higgins PJ. Cancer-associated fibroblasts: mechanisms of tumor progression and novel therapeutic targets. Cancers. 2022;14(5):1231.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Belhabib I, Zaghdoudi S, Lac C, Bousquet C, Jean C. Extracellular matrices and cancer-associated fibroblasts: targets for cancer diagnosis and therapy? Cancers. 2021;13(14):3466.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. 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(3):174–86.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Erdogan B, Webb DJ. Cancer-associated fibroblasts modulate growth factor signaling and extracellular matrix remodeling to regulate tumor metastasis. Biochem Soc Trans. 2017;45(1):229–36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. 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(1):48.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Papanicolaou M, Parker AL, Yam M, Filipe EC, Wu SZ, Chitty JL, et al. Temporal profiling of the breast tumour microenvironment reveals collagen XII as a driver of metastasis. Nat Commun. 2022;13(1):4587.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Stouten I, van Montfoort N, Hawinkels LJAC. The tango between cancer-associated fibroblasts (CAFs) and immune cells in affecting immunotherapy efficacy in pancreatic cancer. Int J Mol Sci. 2023;24(10):8707.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Mao X, Xu J, Wang W, Liang C, Hua J, Liu J, et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer. 2021;20(1):131.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. 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(3):463-479.e410.

    Article  PubMed  CAS  Google Scholar 

  24. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;6(5):392–401.

    Article  PubMed  CAS  Google Scholar 

  25. Yang D, Liu J, Qian H, Zhuang Q. Cancer-associated fibroblasts: from basic science to anticancer therapy. Exp Mol Med. 2023;55(7):1322–32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Rettig WJ, Garin-Chesa P, Beresford HR, Oettgen HF, Melamed MR, Old LJ. Cell-surface glycoproteins of human sarcomas: differential expression in normal and malignant tissues and cultured cells. Proc Natl Acad Sci USA. 1988;85(9):3110–4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Park JE, Lenter MC, Zimmermann RN, Garin-Chesa P, Old LJ, Rettig WJ. Fibroblast activation protein, a dual specificity serine protease expressed in reactive human tumor stromal fibroblasts. J Biol Chem. 1999;274(51):36505–12.

    Article  PubMed  CAS  Google Scholar 

  28. Crane JN, Graham DS, Mona CE, Nelson SD, Samiei A, Dawson DW, et al. Fibroblast activation protein expression in sarcomas. Sarcoma. 2023;2023:2480493.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Fitzgerald AA, Weiner LM. The role of fibroblast activation protein in health and malignancy. Cancer Metastasis Rev. 2020;39(3):783–803.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Jacob M, Chang L, Puré E. Fibroblast activation protein in remodeling tissues. Curr Mol Med. 2012;12(10):1220–43.

    Article  PubMed  CAS  Google Scholar 

  31. Wang Z, Wang J, Lan T, Zhang L, Yan Z, Zhang N, et al. Role and mechanism of fibroblast-activated protein-α expression on the surface of fibroblast-like synoviocytes in rheumatoid arthritis. Front Immunol. 2023;14:1135384.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Lay AJ, Zhang HE, McCaughan GW, Gorrell MD. Fibroblast activation protein in liver fibrosis. Front Biosci (Landmark Ed). 2019;24(1):1–17.

    Article  PubMed  Google Scholar 

  33. Ebert LM, Yu W, Gargett T, Toubia J, Kollis PM, Tea MN, et al. Endothelial, pericyte and tumor cell expression in glioblastoma identifies fibroblast activation protein (FAP) as an excellent target for immunotherapy. Clin Transl Immunol. 2020;9(10): e1191.

    Article  CAS  Google Scholar 

  34. 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(4):895–905.

    Article  PubMed  CAS  Google Scholar 

  35. Lyu Z, Li Y, Zhu D, Wu S, Hu F, Zhang Y, et al. Fibroblast activation protein-alpha is a prognostic biomarker associated with ferroptosis in stomach adenocarcinoma. Fronti Cell Dev Biol. 2022;10: 859999.

    Article  Google Scholar 

  36. Muilwijk T, Akand M, Daelemans S, Marien K, Waumans Y, Kockx M, et al. Stromal marker fibroblast activation protein drives outcome in T1 non-muscle invasive bladder cancer. PLoS ONE. 2021;16(9): e0257195.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Kalaei Z, Manafi-Farid R, Rashidi B, Kiani FK, Zarei A, Fathi M, et al. The Prognostic and therapeutic value and clinical implications of fibroblast activation protein-α as a novel biomarker in colorectal cancer. Cell Commun Signal. 2023;21(1):139.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Yanagawa N, Sugai M, Shikanai S, Sugimoto R, Osakabe M, Uesugi N, et al. High expression of fibroblast-activating protein is a prognostic marker in non-small cell lung carcinoma. Thoracic Cancer. 2022;13(16):2377–84.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Park H, Lee Y, Lee H, Kim JW, Hwang JH, Kim J, et al. The prognostic significance of cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. Tumour Biol. 2017;39(10):1010428317718403.

    Article  PubMed  Google Scholar 

  40. Ariga N, Sato E, Ohuchi N, Nagura H, Ohtani H. Stromal expression of fibroblast activation protein/seprase, a cell membrane serine proteinase and gelatinase, is associated with longer survival in patients with invasive ductal carcinoma of breast. Int J Cancer. 2001;95(1):67–72.

    Article  PubMed  CAS  Google Scholar 

  41. Cheng JD, Dunbrack RL Jr, Valianou M, Rogatko A, Alpaugh RK, Weiner LM. Promotion of tumor growth by murine fibroblast activation protein, a serine protease, in an animal model. Cancer Res. 2002;62(16):4767–72.

    PubMed  CAS  Google Scholar 

  42. Liao D, Luo Y, Markowitz D, Xiang R, Reisfeld RA. Cancer associated fibroblasts promote tumor growth and metastasis by modulating the tumor immune microenvironment in a 4T1 murine breast cancer model. PLoS ONE. 2009;4(11): e7965.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Lo A, Li CP, Buza EL, Blomberg R, Govindaraju P, Avery D, et al. Fibroblast activation protein augments progression and metastasis of pancreatic ductal adenocarcinoma. JCI Insight. 2017;2(19): e92232.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Ji D, Jia J, Cui X, Li Z, Wu A. FAP promotes metastasis and chemoresistance via regulating YAP1 and macrophages in mucinous colorectal adenocarcinoma. iScience. 2023;26(6): 106600.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Lee H-O, Mullins SR, Franco-Barraza J, Valianou M, Cukierman E, Cheng JD. FAP-overexpressing fibroblasts produce an extracellular matrix that enhances invasive velocity and directionality of pancreatic cancer cells. BMC Cancer. 2011;11(1):245.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Barrett RL, Puré E. Cancer-associated fibroblasts and their influence on tumor immunity and immunotherapy. Elife. 2020;9: e57243.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Kraman M, Bambrough PJ, Arnold JN, Roberts EW, Magiera L, Jones JO, et al. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science. 2010;330(6005):827–30.

    Article  PubMed  CAS  Google Scholar 

  48. Fearon DT. The carcinoma-associated fibroblast expressing fibroblast activation protein and escape from immune surveillance. Cancer Immunol Res. 2014;2(3):187–93.

    Article  PubMed  CAS  Google Scholar 

  49. 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(14):4124–35.

    Article  CAS  Google Scholar 

  50. Santos AM, Jung J, Aziz N, Kissil JL, Puré E. Targeting fibroblast activation protein inhibits tumor stromagenesis and growth in mice. J Clin Invest. 2009;119(12):3613–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Teichgräber V, Monasterio C, Chaitanya K, Boger R, Gordon K, Dieterle T, et al. Specific inhibition of fibroblast activation protein (FAP)-alpha prevents tumor progression in vitro. Adv Med Sci. 2015;60(2):264–72.

    Article  PubMed  Google Scholar 

  52. Lin Y, Li B, Yang X, Cai Q, Liu W, Tian M, et al. Fibroblastic FAP promotes intrahepatic cholangiocarcinoma growth via MDSCs recruitment. Neoplasia. 2019;21(12):1133–42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Huang M, Fu M, Wang J, Xia C, Zhang H, Xiong Y, et al. TGF-β1-activated cancer-associated fibroblasts promote breast cancer invasion, metastasis and epithelial-mesenchymal transition by autophagy or overexpression of FAP-α. Biochem Pharmacol. 2021;188: 114527.

    Article  PubMed  CAS  Google Scholar 

  54. Wen X, He X, Jiao F, Wang C, Sun Y, Ren X, et al. Fibroblast activation protein-α-positive fibroblasts promote gastric cancer progression and resistance to immune checkpoint blockade. Oncol Res. 2017;25(4):629–40.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Wang H, Wu Q, Liu Z, Luo X, Fan Y, Liu Y, et al. Downregulation of FAP suppresses cell proliferation and metastasis through PTEN/PI3K/AKT and Ras-ERK signaling in oral squamous cell carcinoma. Cell Death Dis. 2014;5(4): e1155.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. An J, Hou D, Wang L, Wang L, Yang Y, Wang H. Fibroblast activation protein-alpha knockdown suppresses prostate cancer cell invasion and proliferation. Histol Histopathol. 2022;37(6):597–607.

    PubMed  CAS  Google Scholar 

  57. Muchlińska A, Nagel A, Popęda M, Szade J, Niemira M, Zieliński J, et al. Alpha-smooth muscle actin-positive cancer-associated fibroblasts secreting osteopontin promote growth of luminal breast cancer. Cell Mol Biol Lett. 2022;27(1):45.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Chen J, Yang P, Xiao Y, Zhang Y, Liu J, Xie D, et al. Overexpression of α-sma-positive fibroblasts (CAFs) in nasopharyngeal carcinoma predicts poor prognosis. J Cancer. 2017;8(18):3897–902.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Chuaysri C, Thuwajit P, Paupairoj A, Chau-In S, Suthiphongchai T, Thuwajit C. Alpha-smooth muscle actin-positive fibroblasts promote biliary cell proliferation and correlate with poor survival in cholangiocarcinoma. Oncol Rep. 2009;21(4):957–69.

    PubMed  CAS  Google Scholar 

  60. Patel AK, Vipparthi K, Thatikonda V, Arun I, Bhattacharjee S, Sharan R, et al. A subtype of cancer-associated fibroblasts with lower expression of alpha-smooth muscle actin suppresses stemness through BMP4 in oral carcinoma. Oncogenesis. 2018;7(10):78.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Huang M, Li Y, Zhang H, Nan F. Breast cancer stromal fibroblasts promote the generation of CD44+CD24- cells through SDF-1/CXCR4 interaction. J Exp Clin Cancer Res. 2010;29(1):80.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Lau EY, Lo J, Cheng BY, Ma MK, Lee JM, Ng JK, et al. Cancer-associated fibroblasts regulate tumor-initiating cell plasticity in hepatocellular carcinoma through c-Met/FRA1/HEY1 signaling. Cell Rep. 2016;15(6):1175–89.

    Article  PubMed  CAS  Google Scholar 

  63. Mace TA, Ameen Z, Collins A, Wojcik S, Mair M, Young GS, et al. Pancreatic cancer-associated stellate cells promote differentiation of myeloid-derived suppressor cells in a STAT3-dependent manner. Cancer Res. 2013;73(10):3007–18.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol. 2017;17(9):559–72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Zhang R, Qi F, Zhao F, Li G, Shao S, Zhang X, et al. Cancer-associated fibroblasts enhance tumor-associated macrophages enrichment and suppress NK cells function in colorectal cancer. Cell Death Dis. 2019;10(4):273.

    Article  PubMed  PubMed Central  Google Scholar 

  66. 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(4):422.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Özdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25(6):719–34.

    Article  PubMed  PubMed Central  Google Scholar 

  68. 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(6):735–47.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Heldin C-H. Targeting the PDGF signaling pathway in tumor treatment. Cell Commun Signal. 2013;11(1):97.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Guérit E, Arts F, Dachy G, Boulouadnine B, Demoulin J-B. PDGF receptor mutations in human diseases. Cell Mol Life Sci. 2021;78(8):3867–81.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Ying HZ, Chen Q, Zhang WY, Zhang HH, Ma Y, Zhang SZ, et al. PDGF signaling pathway in hepatic fibrosis pathogenesis and therapeutics (review). Mol Med Rep. 2017;16(6):7879–89.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Gouveia L, Kraut S, Hadzic S, Vazquéz-Liébanas E, Kojonazarov B, Wu C-Y, et al. Lung developmental arrest caused by PDGF-A deletion: consequences for the adult mouse lung. Am J Physiol Lung Cell Mol Physiol. 2020;318(4):L831–43.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Boström H, Gritli-Linde A, Betsholtz C. PDGF-A/PDGF alpha-receptor signaling is required for lung growth and the formation of alveoli but not for early lung branching morphogenesis. Dev Dyn. 2002;223(1):155–62.

    Article  PubMed  Google Scholar 

  74. Tian Y, Zhan Y, Jiang Q, Lu W, Li X. Expression and function of PDGF-C in development and stem cells. Open Biol. 2021;11(12): 210268.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Nordby Y, Richardsen E, Rakaee M, Ness N, Donnem T, Patel HRH, 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(1):43378.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Winkler EA, Bell RD, Zlokovic BV. Pericyte-specific expression of PDGF beta receptor in mouse models with normal and deficient PDGF beta receptor signaling. Mol Neurodegener. 2010;5:32.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Hägglöf C, Hammarsten P, Josefsson A, Stattin P, Paulsson J, Bergh A, et al. Stromal PDGFRbeta expression in prostate tumors and non-malignant prostate tissue predicts prostate cancer survival. PLoS ONE. 2010;5(5): e10747.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Strell C, Stenmark Tullberg A, Jetne Edelmann R, Akslen LA, Malmström P, Fernö M, et al. Prognostic and predictive impact of stroma cells defined by PDGFRb expression in early breast cancer: results from the randomized SweBCG91RT trial. Breast Cancer Res Treat. 2021;187(1):45–55.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Fujino S, Miyoshi N, Ohue M, Takahashi Y, Yasui M, Hata T, et al. Platelet-derived growth factor receptor-β gene expression relates to recurrence in colorectal cancer. Oncol Rep. 2018;39(5):2178–84.

    PubMed  CAS  Google Scholar 

  80. Madsen CV, Dahl Steffensen K, Waldstrøm M, Jakobsen A. Immunohistochemical expression of platelet-derived growth factor receptors in ovarian cancer patients with long-term follow-up. Patholog Res Int. 2012;2012: 851432.

    PubMed  PubMed Central  Google Scholar 

  81. Chang KK, Yoon C, Yi BC, Tap WD, Simon MC, Yoon SS. Platelet-derived growth factor receptor-α and -β promote cancer stem cell phenotypes in sarcomas. Oncogenesis. 2018;7(6):47.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Erdogan B, Ao M, White LM, Means AL, Brewer BM, Yang L, et al. Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin. J Cell Biol. 2017;216(11):3799–816.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Primac I, Maquoi E, Blacher S, Heljasvaara R, Van Deun J, Smeland HYH, et al. Stromal integrin α11 regulates PDGFRβ signaling and promotes breast cancer progression. J Clin Investig. 2019;129(11):4609–28.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Peña C, Céspedes MV, Lindh MB, Kiflemariam S, Mezheyeuski A, Edqvist PH, et al. STC1 expression by cancer-associated fibroblasts drives metastasis of colorectal cancer. Cancer Res. 2013;73(4):1287–97.

    Article  PubMed  Google Scholar 

  85. Aoto K, Ito K, Aoki S. Complex formation between platelet-derived growth factor receptor β and transforming growth factor β receptor regulates the differentiation of mesenchymal stem cells into cancer-associated fibroblasts. Oncotarget. 2018;9(75):34090–102.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Djurec M, Graña O, Lee A, Troulé K, Espinet E, Cabras L, et al. Saa3 is a key mediator of the protumorigenic properties of cancer-associated fibroblasts in pancreatic tumors. Proc Natl Acad Sci USA. 2018;115(6):E1147-e1156.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. 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(31):4457–68.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Lakins MA, Ghorani E, Munir H, Martins CP, Shields JD. Cancer-associated fibroblasts induce antigen-specific deletion of CD8 (+) T cells to protect tumour cells. Nat Commun. 2018;9(1):948.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Wu Z, Xu J, Tang R, Wang W, Zhang B, Yu X, et al. The role of PDGFRA in predicting oncological and immune characteristics in pancreatic ductal adenocarcinoma. J Oncol. 2022;2022:4148805.

    PubMed  PubMed Central  Google Scholar 

  90. Yoon H, Tang C-M, Banerjee S, Yebra M, Noh S, Burgoyne AM, et al. Cancer-associated fibroblast secretion of PDGFC promotes gastrointestinal stromal tumor growth and metastasis. Oncogene. 2021;40(11):1957–73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Hayashi Y, Bardsley MR, Toyomasu Y, Milosavljevic S, Gajdos GB, Choi KM, et al. Platelet-derived growth factor receptor-α regulates proliferation of gastrointestinal stromal tumor cells with mutations in KIT by stabilizing ETV1. Gastroenterology. 2015;149(2):420-432.e416.

    Article  PubMed  CAS  Google Scholar 

  92. Peng G, Wang Y, Ge P, Bailey C, Zhang P, Zhang D, et al. The HIF1α-PDGFD-PDGFRα axis controls glioblastoma growth at normoxia/mild-hypoxia and confers sensitivity to targeted therapy by echinomycin. J Exp Clin Cancer Res. 2021;40(1):278.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Bai F, Liu S, Liu X, Hollern DP, Scott A, Wang C, et al. PDGFRβ is an essential therapeutic target for BRCA1-deficient mammary tumors. Breast Cancer Res. 2021;23(1):10.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Li Y, Tao Y, Gao S, Li P, Zheng J, Zhang S, et al. Cancer-associated fibroblasts contribute to oral cancer cells proliferation and metastasis via exosome-mediated paracrine miR-34a-5p. EBioMedicine. 2018;36:209–20.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Maehira H, Miyake T, Iida H, Tokuda A, Mori H, Yasukawa D, et al. Vimentin expression in tumor microenvironment predicts survival in pancreatic ductal adenocarcinoma: heterogeneity in fibroblast population. Ann Surg Oncol. 2019;26(13):4791–804.

    Article  PubMed  Google Scholar 

  96. Ngan CY, Yamamoto H, Seshimo I, Tsujino T, Man-i M, Ikeda JI, et al. Quantitative evaluation of vimentin expression in tumour stroma of colorectal cancer. Br J Cancer. 2007;96(6):986–92.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Szubert S, Koper K, Dutsch-Wicherek MM, Jozwicki W. High tumor cell vimentin expression indicates prolonged survival in patients with ovarian malignant tumors. Ginekol Pol. 2019;90(1):11–9.

    Article  PubMed  Google Scholar 

  98. Strouhalova K, Přechová M, Gandalovičová A, Brábek J, Gregor M, Rosel D. Vimentin intermediate filaments as potential target for cancer treatment. Cancers. 2020;12(1):184.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Liu CY, Lin HH, Tang MJ, Wang YK. Vimentin contributes to epithelial-mesenchymal transition cancer cell mechanics by mediating cytoskeletal organization and focal adhesion maturation. Oncotarget. 2015;6(18):15966–83.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Richardson AM, Havel LS, Koyen AE, Konen JM, Shupe J, Wiles WG IV, et al. Vimentin is required for lung adenocarcinoma metastasis via heterotypic tumor cell–cancer-associated fibroblast interactions during collective invasion. Clin Cancer Res. 2018;24(2):420–32.

    Article  PubMed  CAS  Google Scholar 

  101. Berr AL, Wiese K, dos Santos G, Koch CM, Anekalla KR, Kidd M, et al. Vimentin is required for tumor progression and metastasis in a mouse model of non–small cell lung cancer. Oncogene. 2023;42(25):2074–87.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Dmello C, Sawant S, Alam H, Gangadaran P, Tiwari R, Dongre H, et al. Vimentin-mediated regulation of cell motility through modulation of beta4 integrin protein levels in oral tumor derived cells. Int J Biochem Cell Biol. 2016;70:161–72.

    Article  PubMed  CAS  Google Scholar 

  103. Xuan B, Ghosh D, Jiang J, Shao R, Dawson MR. Vimentin filaments drive migratory persistence in polyploidal cancer cells. Proc Natl Acad Sci. 2020;117(43):26756–65.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Schacht V, Ramirez MI, Hong Y-K, Hirakawa S, Feng D, Harvey N, et al. T1α/podoplanin deficiency disrupts normal lymphatic vasculature formation and causes lymphedema. EMBO J. 2003;22(14):3546–56.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Fu J, Gerhardt H, McDaniel JM, Xia B, Liu X, Ivanciu L, et al. Endothelial cell O-glycan deficiency causes blood/lymphatic misconnections and consequent fatty liver disease in mice. J Clin Investig. 2008;118(11):3725–37.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Kong LL, Yang NZ, Shi LH, Zhao GH, Zhou W, Ding Q, et al. The optimum marker for the detection of lymphatic vessels. Mol Clin Oncol. 2017;7(4):515–20.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Suzuki J, Aokage K, Neri S, Sakai T, Hashimoto H, Su Y, et al. Relationship between podoplanin-expressing cancer-associated fibroblasts and the immune microenvironment of early lung squamous cell carcinoma. Lung Cancer. 2021;153:1–10.

    Article  PubMed  CAS  Google Scholar 

  108. Pula B, Jethon A, Piotrowska A, Gomulkiewicz A, Owczarek T, Calik J, et al. Podoplanin expression by cancer-associated fibroblasts predicts poor outcome in invasive ductal breast carcinoma. Histopathology. 2011;59(6):1249–60.

    Article  PubMed  Google Scholar 

  109. Shindo K, Aishima S, Ohuchida K, Fujiwara K, Fujino M, Mizuuchi Y, et al. Podoplanin expression in cancer-associated fibroblasts enhances tumor progression of invasive ductal carcinoma of the pancreas. Mol Cancer. 2013;12:1–16.

    Article  Google Scholar 

  110. 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(4):784–96.

    Article  PubMed  CAS  Google Scholar 

  111. Suchanski J, Tejchman A, Zacharski M, Piotrowska A, Grzegrzolka J, Chodaczek G, et al. Podoplanin increases the migration of human fibroblasts and affects the endothelial cell network formation: A possible role for cancer-associated fibroblasts in breast cancer progression. PLoS ONE. 2017;12(9): e0184970.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Takahashi A, Ishii G, Neri S, Yoshida T, Hashimoto H, Suzuki S, et al. Podoplanin-expressing cancer-associated fibroblasts inhibit small cell lung cancer growth. Oncotarget. 2015;6(11):9531–41.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Choi SY, Sung R, Lee SJ, Lee TG, Kim N, Yoon SM, et al. Podoplanin, α-smooth muscle actin or S100A4 expressing cancer-associated fibroblasts are associated with different prognosis in colorectal cancers. J Korean Med Sci. 2013;28(9):1293–301.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Chihara N, Madi A, Kondo T, Zhang H, Acharya N, Singer M, et al. Induction and transcriptional regulation of the co-inhibitory gene module in T cells. Nature. 2018;558(7710):454–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Bieniasz-Krzywiec P, Martín-Pérez R, Ehling M, García-Caballero M, Pinioti S, Pretto S, et al. Podoplanin-expressing macrophages promote lymphangiogenesis and lymphoinvasion in breast cancer. Cell Metab. 2019;30(5):917-936.e910.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Hsu YB, Huang CF, Lin KT, Kuo YL, Lan MC, Lan MY. Podoplanin, a potential therapeutic target for nasopharyngeal carcinoma. Biomed Res Int. 2019;2019:7457013.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Sasano T, Gonzalez-Delgado R, Muñoz NM, Carlos-Alcade W, Cho MS, Sheth RA, et al. Podoplanin promotes tumor growth, platelet aggregation, and venous thrombosis in murine models of ovarian cancer. J Thromb Haemost. 2022;20(1):104–14.

    Article  PubMed  CAS  Google Scholar 

  118. Sikorska J, Gaweł D, Domek H, Rudzińska M, Czarnocka B. Podoplanin (PDPN) affects the invasiveness of thyroid carcinoma cells by inducing ezrin, radixin and moesin (E/R/M) phosphorylation in association with matrix metalloproteinases. BMC Cancer. 2019;19(1):85.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Lawson WE, Polosukhin VV, Zoia O, Stathopoulos GT, Han W, Plieth D, et al. Characterization of fibroblast-specific protein 1 in pulmonary fibrosis. Am J Respir Crit Care Med. 2005;171(8):899–907.

    Article  PubMed  Google Scholar 

  120. Schneider M, Kostin S, Strøm CC, Aplin M, Lyngbaek S, Theilade J, et al. S100A4 is upregulated in injured myocardium and promotes growth and survival of cardiac myocytes. Cardiovasc Res. 2007;75(1):40–50.

    Article  PubMed  CAS  Google Scholar 

  121. Ye F, Liang Y, Wang Y, Le Yang R, Luo D, Li Y, et al. Cancer-associated fibroblasts facilitate breast cancer progression through exosomal circTBPL1-mediated intercellular communication. Cell Death Dis. 2023;14(7):471.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  122. Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res. 2007;67(21):10123–8.

    Article  PubMed  CAS  Google Scholar 

  123. Österreicher CH, Penz-Österreicher M, Grivennikov SI, Guma M, Koltsova EK, Datz C, et al. Fibroblast-specific protein 1 identifies an inflammatory subpopulation of macrophages in the liver. Proc Natl Acad Sci. 2011;108(1):308–13.

    Article  PubMed  Google Scholar 

  124. Im SB, Cho JM, Kim HB, Shin DH, Kwon MS, Lee IY, et al. FSP-1 expression in cancer cells is relevant to long-term oncological outcomes in nonmetastatic colorectal cancer. Korean J Clin Oncol. 2022;18(2):66–77.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Park CK, Jung WH, Koo JS. Expression of cancer-associated fibroblast-related proteins differs between invasive lobular carcinoma and invasive ductal carcinoma. Breast Cancer Res Treat. 2016;159(1):55–69.

    Article  PubMed  CAS  Google Scholar 

  126. O’Connell JT, Sugimoto H, Cooke VG, MacDonald BA, Mehta AI, LeBleu VS, et al. VEGF-A and Tenascin-C produced by S100A4+ stromal cells are important for metastatic colonization. Proc Natl Acad Sci U S A. 2011;108(38):16002–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Zhang J, Chen L, Xiao M, Wang C, Qin Z. FSP1+ fibroblasts promote skin carcinogenesis by maintaining MCP-1-mediated macrophage infiltration and chronic inflammation. Am J Pathol. 2011;178(1):382–90.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Grum-Schwensen B, Klingelhofer J, Berg CH, El-Naaman C, Grigorian M, Lukanidin E, et al. Suppression of tumor development and metastasis formation in mice lacking the S100A4(mts1) gene. Cancer Res. 2005;65(9):3772–80.

    Article  PubMed  CAS  Google Scholar 

  129. Jiao J, González Á, Stevenson HL, Gagea M, Sugimoto H, Kalluri R, et al. Depletion of S100A4+ stromal cells does not prevent HCC development but reduces the stem cell-like phenotype of the tumors. Exp Mol Med. 2018;50(1):e422–e422.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Rasanen K, Sriswasdi S, Valiga A, Tang HY, Zhang G, Perego M, et al. Comparative secretome analysis of epithelial and mesenchymal subpopulations of head and neck squamous cell carcinoma identifies S100A4 as a potential therapeutic target. Mol Cell Proteomics. 2013;12(12):3778–92.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575(7784):693–8.

    Article  PubMed  CAS  Google Scholar 

  132. Midwood KS, Orend G. The role of tenascin-C in tissue injury and tumorigenesis. J Cell Commun Signal. 2009;3(3–4):287–310.

    Article  PubMed  PubMed Central  Google Scholar 

  133. De Wever O, Nguyen QD, Van Hoorde L, Bracke M, Bruyneel E, Gespach C, et al. Tenascin-C and SF/HGF produced by myofibroblasts in vitro provide convergent pro-invasive signals to human colon cancer cells through RhoA and Rac. Faseb j. 2004;18(9):1016–8.

    Article  PubMed  Google Scholar 

  134. Jang I, Beningo KA. Integrins, CAFs and mechanical forces in the progression of cancer. Cancers. 2019;11(5):721.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Ni WD, Yang ZT, Cui CA, Cui Y, Fang LY, Xuan YH. Tenascin-C is a potential cancer-associated fibroblasts marker and predicts poor prognosis in prostate cancer. Biochem Biophys Res Commun. 2017;486(3):607–12.

    Article  PubMed  CAS  Google Scholar 

  136. Furuhashi S, Morita Y, Matsumoto A, Ida S, Muraki R, Kitajima R, et al. Tenascin C in pancreatic cancer-associated fibroblasts enhances epithelial mesenchymal transition and is associated with resistance to immune checkpoint inhibitor. Am J Cancer Res. 2023;13(11):5641–55.

    PubMed  PubMed Central  CAS  Google Scholar 

  137. Shen C, Wang C, Yin Y, Chen H, Yin X, Cai Z, et al. Tenascin-C expression is significantly associated with the progression and prognosis in gastric GISTs. Medicine. 2019;98(2): e14045.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  138. Ming X, Qiu S, Liu X, Li S, Wang Y, Zhu M, et al. Prognostic role of tenascin-c for cancer outcome: a meta-analysis. Technol Cancer Res Treat. 2019;18:1533033818821106.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  139. Yang Z-T, Yeo S-Y, Yin Y-X, Lin Z-H, Lee H-M, Xuan Y-H, et al. Tenascin-C, a prognostic determinant of esophageal squamous cell carcinoma. PLoS ONE. 2016;11(1): e0145807.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Yang Z, Zhang C, Feng Y, Quan M, Cui Y, Xuan Y. Tenascin-C predicts poor outcomes for patients with colorectal cancer and drives cancer stemness via Hedgehog signaling pathway. Cancer Cell Int. 2020;20(1):122.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Sun Z, Schwenzer A, Rupp T, Murdamoothoo D, Vegliante R, Lefebvre O, et al. Tenascin-C promotes tumor cell migration and metastasis through integrin α9β1–mediated YAP inhibition. Can Res. 2018;78(4):950–61.

    Article  CAS  Google Scholar 

  142. Murdamoothoo D, Sun Z, Yilmaz A, Riegel G, Abou-Faycal C, Deligne C, et al. Tenascin-C immobilizes infiltrating T lymphocytes through CXCL12 promoting breast cancer progression. EMBO Mol Med. 2021;13(6): e13270.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  143. Spenlé C, Loustau T, Murdamoothoo D, Erne W, Beghelli-de la Forest Divonne S, Veber R, et al. Tenascin-C orchestrates an immune-suppressive tumor microenvironment in oral squamous cell carcinoma. Cancer Immunol Res. 2020;8(9):1122–38.

    Article  PubMed  Google Scholar 

  144. Sun Z, Velázquez-Quesada I, Murdamoothoo D, Ahowesso C, Yilmaz A, Spenlé C, et al. Tenascin-C increases lung metastasis by impacting blood vessel invasions. Matrix Biol. 2019;83:26–47.

    Article  PubMed  CAS  Google Scholar 

  145. Rupp T, Langlois B, Koczorowska MM, Radwanska A, Sun Z, Hussenet T, et al. Tenascin-C orchestrates glioblastoma angiogenesis by modulation of pro- and anti-angiogenic signaling. Cell Rep. 2016;17(10):2607–19.

    Article  PubMed  CAS  Google Scholar 

  146. Liu Y, Yang L-Y, Chen D-X, Chang C, Yuan Q, Zhang Y, et al. Tenascin-C as a potential biomarker and therapeutic target for esophageal squamous cell carcinoma. Transl Oncol. 2024;42: 101888.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  147. Yilmaz A, Loustau T, Salomé N, Poilil Surendran S, Li C, Tucker RP, et al. Advances on the roles of tenascin-C in cancer. J Cell Sci. 2022;135(18): jcs260244.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  148. Dhaouadi S, Bouhaouala-Zahar B, Orend G. Tenascin-C targeting strategies in cancer. Matrix Biol. 2024;130:1–19.

    Article  PubMed  CAS  Google Scholar 

  149. Ratajczak-Wielgomas K, Grzegrzolka J, Piotrowska A, Gomulkiewicz A, Witkiewicz W, Dziegiel P. Periostin expression in cancer-associated fibroblasts of invasive ductal breast carcinoma. Oncol Rep. 2016;36(5):2745–54.

    Article  PubMed  CAS  Google Scholar 

  150. Wei W-F, Chen X-J, Liang L-J, Yu L, Wu X-G, Zhou C-F, et al. Periostin+cancer-associated fibroblasts promote lymph node metastasis by impairing the lymphatic endothelial barriers in cervical squamous cell carcinoma. Mol Oncol. 2021;15(1):210–27.

    Article  PubMed  CAS  Google Scholar 

  151. Kikuchi Y, Kashima TG, Nishiyama T, Shimazu K, Morishita Y, Shimazaki M, et al. Periostin is expressed in pericryptal fibroblasts and cancer-associated fibroblasts in the colon. J Histochem Cytochem. 2008;56(8):753–64.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  152. Deng X, Ao S, Hou J, Li Z, Lei Y, Lyu G. Prognostic significance of periostin in colorectal cancer. Chin J Cancer Res. 2019;31(3):547–56.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  153. Underwood TJ, Hayden AL, Derouet M, Garcia E, Noble F, White MJ, et al. Cancer-associated fibroblasts predict poor outcome and promote periostin-dependent invasion in oesophageal adenocarcinoma. J Pathol. 2015;235(3):466–77.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  154. Neuzillet C, Nicolle R, Raffenne J, Tijeras-Raballand A, Brunel A, Astorgues-Xerri L, et al. Periostin- and podoplanin-positive cancer-associated fibroblast subtypes cooperate to shape the inflamed tumor microenvironment in aggressive pancreatic adenocarcinoma. J Pathol. 2022;258(4):408–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  155. Akinjiyan FA, Dave RM, Alpert E, Longmore GD, Fuh KC. DDR2 expression in cancer-associated fibroblasts promotes ovarian cancer tumor invasion and metastasis through periostin-ITGB1. Cancers. 2022;14(14):3482.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  156. Yue H, Li W, Chen R, Wang J, Lu X, Li J. Stromal POSTN induced by TGF-β1 facilitates the migration and invasion of ovarian cancer. Gynecol Oncol. 2021;160(2):530–8.

    Article  PubMed  CAS  Google Scholar 

  157. Yoshikawa M, Takatsu F, Suzawa K, Habu T, Masayoshi O, Iwata K, et al. Abstract 5845: Periostin secreted by cancer-associated fibroblasts promotes cancer progression and drug resistance in non-small cell lung cancer. Cancer Res. 2023;83(7_Supplement):5845–5845.

    Article  Google Scholar 

  158. Kikuchi Y, Kunita A, Iwata C, Komura D, Nishiyama T, Shimazu K, et al. The niche component periostin is produced by cancer-associated fibroblasts, supporting growth of gastric cancer through ERK activation. Am J Pathol. 2014;184(3):859–70.

    Article  PubMed  CAS  Google Scholar 

  159. Liu Y, Li F, Gao F, Xing L, Qin P, Liang X, et al. Role of microenvironmental periostin in pancreatic cancer progression. Oncotarget. 2016;8(52):89552.

    Article  PubMed  PubMed Central  Google Scholar 

  160. Yu B, Wu K, Wang X, Zhang J, Wang L, Jiang Y, et al. Periostin secreted by cancer-associated fibroblasts promotes cancer stemness in head and neck cancer by activating protein tyrosine kinase 7. Cell Death Dis. 2018;9(11):1082.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Lin S-C, Liao Y-C, Chen P-M, Yang Y-Y, Wang Y-H, Tung S-L, et al. Periostin promotes ovarian cancer metastasis by enhancing M2 macrophages and cancer-associated fibroblasts via integrin-mediated NF-κB and TGF-β2 signaling. J Biomed Sci. 2022;29(1):109.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  162. Wei T, Wang K, Liu S, Fang Y, Hong Z, Liu Y, et al. Periostin deficiency reduces PD-1(+) tumor-associated macrophage infiltration and enhances anti-PD-1 efficacy in colorectal cancer. Cell Rep. 2023;42(2): 112090.

    Article  PubMed  CAS  Google Scholar 

  163. Ma H, Wang J, Zhao X, Wu T, Huang Z, Chen D, et al. Periostin promotes colorectal tumorigenesis through integrin-FAK-Src pathway-mediated YAP/TAZ activation. Cell Rep. 2020;30(3):793-806.e796.

    Article  PubMed  CAS  Google Scholar 

  164. Okazaki T, Tamai K, Shibuya R, Nakamura M, Mochizuki M, Yamaguchi K, et al. Periostin is a negative prognostic factor and promotes cancer cell proliferation in non-small cell lung cancer. Oncotarget. 2018;9(58):31187–99.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Shimazaki M, Kudo A. Impaired capsule formation of tumors in periostin-null mice. Biochem Biophys Res Commun. 2008;367(4):736–42.

    Article  PubMed  CAS  Google Scholar 

  166. Hu W-W, Chen P-C, Chen J-M, Wu Y-M, Liu P-Y, Lu C-H, et al. Periostin promotes epithelial-mesenchymal transition via the MAPK/miR-381 axis in lung cancer. Oncotarget. 2017;8(37):62248.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Camby I, Le Mercier M, Lefranc F, Kiss R. Galectin-1: a small protein with major functions. Glycobiology. 2006;16(11):137R-157R.

    Article  PubMed  CAS  Google Scholar 

  168. Chong Y, Tang D, Xiong Q, Jiang X, Xu C, Huang Y, et al. Galectin-1 from cancer-associated fibroblasts induces epithelial–mesenchymal transition through β1 integrin-mediated upregulation of Gli1 in gastric cancer. J Exp Clin Cancer Res. 2016;35(1):175.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Tang D, Gao J, Wang S, Ye N, Chong Y, Huang Y, et al. Cancer-associated fibroblasts promote angiogenesis in gastric cancer through galectin-1 expression. Tumor Biol. 2016;37(2):1889–99.

    Article  CAS  Google Scholar 

  170. Zheng L, Xu C, Guan Z, Su X, Xu Z, Cao J, et al. Galectin-1 mediates TGF-β-induced transformation from normal fibroblasts into carcinoma-associated fibroblasts and promotes tumor progression in gastric cancer. Am J Transl Res. 2016;8(4):1641–58.

    PubMed  PubMed Central  CAS  Google Scholar 

  171. Rizzolio S, Orrù C, Volante M, Bellomo SE, Migliore C, Giordano S, et al. CAF-released galectin 1 mediates non-cell-autonomous resistance to ceritinib in NSCLC. Research Square, 2023.

  172. Wu M-H, Hong H-C, Hong T-M, Chiang W-F, Jin Y-T, Chen Y-L. Targeting galectin-1 in carcinoma-associated fibroblasts inhibits oral squamous cell carcinoma metastasis by downregulating MCP-1/CCL2 expression. Clin Cancer Res. 2011;17(6):1306–16.

    Article  PubMed  CAS  Google Scholar 

  173. Zhu X, Wang K, Zhang K, Xu F, Yin Y, Zhu L, et al. Galectin-1 knockdown in carcinoma-associated fibroblasts inhibits migration and invasion of human MDA-MB-231 breast cancer cells by modulating MMP-9 expression. Acta Biochim Biophys Sin. 2016;48(5):462–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  174. Dalotto-Moreno T, Croci DO, Cerliani JP, Martinez-Allo VC, Dergan-Dylon S, Méndez-Huergo SP, et al. Targeting galectin-1 overcomes breast cancer-associated immunosuppression and prevents metastatic disease. Cancer Res. 2013;73(3):1107–17.

    Article  PubMed  CAS  Google Scholar 

  175. Chung LY, Tang SJ, Sun GH, Chou TY, Yeh TS, Yu SL, et al. Galectin-1 promotes lung cancer progression and chemoresistance by upregulating p38 MAPK, ERK, and cyclooxygenase-2. Clin Cancer Res. 2012;18(15):4037–47.

    Article  PubMed  CAS  Google Scholar 

  176. Miao J-H, Wang S-Q, Zhang M-H, Yu FB, Zhang L, Yu Z-X, et al. Knockdown of galectin-1 suppresses the growth and invasion of osteosarcoma cells through inhibition of the MAPK/ERK pathway. Oncol Rep. 2014;32(4):1497–504.

    Article  PubMed  CAS  Google Scholar 

  177. Thijssen VL, Postel R, Brandwijk RJ, Dings RP, Nesmelova I, Satijn S, et al. Galectin-1 is essential in tumor angiogenesis and is a target for antiangiogenesis therapy. Proc Natl Acad Sci U S A. 2006;103(43):15975–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  178. Cagnoni AJ, Giribaldi ML, Blidner AG, Cutine AM, Gatto SG, Morales RM, et al. Galectin-1 fosters an immunosuppressive microenvironment in colorectal cancer by reprogramming CD8(+) regulatory T cells. Proc Natl Acad Sci USA. 2021;118(21): e2102950118.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  179. Parton RG. Caveolae: structure, function, and relationship to disease. Annu Rev Cell Dev Biol. 2018;34:111–36.

    Article  PubMed  CAS  Google Scholar 

  180. Simón L, Campos A, Leyton L, Quest AFG. Caveolin-1 function at the plasma membrane and in intracellular compartments in cancer. Cancer Metastasis Rev. 2020;39(2):435–53.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Mercier I, Casimiro MC, Wang C, Rosenberg AL, Quong J, Minkeu A, et al. Human breast cancer-associated fibroblasts (CAFs) show caveolin-1 downregulation and RB tumor suppressor functional inactivation: implications for the response to hormonal therapy. Cancer Biol Ther. 2008;7(8):1212–25.

    Article  PubMed  CAS  Google Scholar 

  182. Zhao X, He Y, Gao J, Fan L, Li Z, Yang G, et al. Caveolin-1 expression level in cancer associated fibroblasts predicts outcome in gastric cancer. PLoS ONE. 2013;8(3): e59102.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  183. Meng Q, Fang Z, Mao X, Tang R, Liang C, Hua J, et al. Metabolic reprogramming of cancer-associated fibroblasts in pancreatic cancer contributes to the intratumor heterogeneity of PET-CT. Comput Struct Biotechnol J. 2023;21:2631–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  184. Kamposioras K, Tsimplouli C, Verbeke C, Anthoney A, Daoukopoulou A, Papandreou CN, et al. Silencing of caveolin-1 in fibroblasts as opposed to epithelial tumor cells results in increased tumor growth rate and chemoresistance in a human pancreatic cancer model. Int J Oncol. 2019;54(2):537–49.

    PubMed  CAS  Google Scholar 

  185. Guan H, Liu Y, Li M. Loss of caveolin-1 in cancer associated fibroblasts promotes hepatocellular carcinoma development. Int J Clin Exp Med. 2018;11(6):5648–56.

    Google Scholar 

  186. Shen X-J, Zhang H, Tang G-S, Wang X-D, Zheng R, Wang Y, et al. Caveolin-1 is a modulator of fibroblast activation and a potential biomarker for gastric cancer. Int J Biol Sci. 2015;11(4):370–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  187. Lin MI, Yu J, Murata T, Sessa WC. Caveolin-1-deficient mice have increased tumor microvascular permeability, angiogenesis, and growth. Cancer Res. 2007;67(6):2849–56.

    Article  PubMed  CAS  Google Scholar 

  188. Mercier I, Camacho J, Titchen K, Gonzales DM, Quann K, Bryant KG, et al. Caveolin-1 and accelerated host aging in the breast tumor microenvironment: chemoprevention with rapamycin, an mTOR inhibitor and anti-aging drug. Am J Pathol. 2012;181(1):278–93.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  189. Wang R, Li Z, Guo H, Shi W, Xin Y, Chang W, et al. Caveolin 1 knockdown inhibits the proliferation, migration and invasion of human breast cancer BT474 cells. Mol Med Rep. 2014;9(5):1723–8.

    Article  PubMed  CAS  Google Scholar 

  190. Díaz MI, Díaz P, Bennett JC, Urra H, Ortiz R, Orellana PC, et al. Caveolin-1 suppresses tumor formation through the inhibition of the unfolded protein response. Cell Death Dis. 2020;11(8):648.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Pasquale EB. Eph receptors and ephrins in cancer progression. Nat Rev Cancer. 2024;24(1):5–27.

    Article  PubMed  CAS  Google Scholar 

  192. Janes PW, Vail ME, Ernst M, Scott AM. Eph receptors in the immunosuppressive tumor microenvironment. Cancer Res. 2021;81(4):801–5.

    Article  PubMed  CAS  Google Scholar 

  193. Kikuchi S, Kaibe N, Morimoto K, Fukui H, Niwa H, Maeyama Y, et al. Overexpression of Ephrin A2 receptors in cancer stromal cells is a prognostic factor for the relapse of gastric cancer. Gastric Cancer. 2015;18(3):485–94.

    Article  PubMed  CAS  Google Scholar 

  194. Wu X, Zahari MS, Renuse S, Sahasrabuddhe NA, Chaerkady R, Kim MS, et al. Quantitative phosphoproteomic analysis reveals reciprocal activation of receptor tyrosine kinases between cancer epithelial cells and stromal fibroblasts. Clin Proteomics. 2018;15:21.

    Article  PubMed  PubMed Central  Google Scholar 

  195. Curtis M, Kenny HA, Ashcroft B, Mukherjee A, Johnson A, Zhang Y, et al. Fibroblasts mobilize tumor cell glycogen to promote proliferation and metastasis. Cell Metab. 2019;29(1):141-155.e149.

    Article  PubMed  CAS  Google Scholar 

  196. Astin JW, Batson J, Kadir S, Charlet J, Persad RA, Gillatt D, et al. Competition amongst Eph receptors regulates contact inhibition of locomotion and invasiveness in prostate cancer cells. Nat Cell Biol. 2010;12(12):1194–204.

    Article  PubMed  CAS  Google Scholar 

  197. Vail ME, Murone C, Tan A, Hii L, Abebe D, Janes PW, et al. Targeting EphA3 inhibits cancer growth by disrupting the tumor stromal microenvironment. Cancer Res. 2014;74(16):4470–81.

    Article  PubMed  CAS  Google Scholar 

  198. Vail ME, Farnsworth RH, Hii L, Allen S, Arora S, Anderson RL, et al. Inhibition of EphA3 expression in tumour stromal cells suppresses tumour growth and progression. Cancers. 2023;15(18):4646.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  199. Talia M, Cirillo F, Spinelli A, Zicarelli A, Scordamaglia D, Muglia L, et al. The Ephrin tyrosine kinase a3 (EphA3) is a novel mediator of RAGE-prompted motility of breast cancer cells. J Exp Clin Cancer Res. 2023;42(1):164.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  200. Nakajima K, Ino Y, Naito C, Nara S, Shimasaki M, Ishimoto U, et al. Neoadjuvant therapy alters the collagen architecture of pancreatic cancer tissue via Ephrin-A5. Br J Cancer. 2022;126(4):628–39.

    Article  PubMed  CAS  Google Scholar 

  201. Lagares D, Ghassemi-Kakroodi P, Tremblay C, Santos A, Probst CK, Franklin A, et al. ADAM10-mediated ephrin-B2 shedding promotes myofibroblast activation and organ fibrosis. Nat Med. 2017;23(12):1405–15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  202. Mueller AC, Piper M, Goodspeed A, Bhuvane S, Williams JS, Bhatia S, et al. Induction of ADAM10 by radiation therapy drives fibrosis, resistance, and epithelial-to-mesenchyal transition in pancreatic cancer. Cancer Res. 2021;81(12):3255–69.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  203. Kakarla M, ChallaSivaKanaka S, Dufficy MF, Gil V, Filipovich Y, Vickman R, et al. Ephrin B activate Src family kinases in fibroblasts inducing stromal remodeling in prostate cancer. Cancers. 2022;14(9):2336.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  204. Cremasco V, Astarita JL, Grauel AL, Keerthivasan S, MacIsaac K, Woodruff MC, et al. FAP delineates heterogeneous and functionally divergent stromal cells in immune-excluded breast tumors. Cancer Immunol Res. 2018;6(12):1472–85.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  205. Higashino N, Koma YI, Hosono M, Takase N, Okamoto M, Kodaira H, et al. Fibroblast activation protein-positive fibroblasts promote tumor progression through secretion of CCL2 and interleukin-6 in esophageal squamous cell carcinoma. Lab Invest. 2019;99(6):777–92.

    Article  PubMed  CAS  Google Scholar 

  206. Ziani L, Chouaib S, Thiery J. Alteration of the antitumor immune response by cancer-associated fibroblasts. Front Immunol. 2018;9:414.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Li T, Yi S, Liu W, Jia C, Wang G, Hua X, et al. Colorectal carcinoma-derived fibroblasts modulate natural killer cell phenotype and antitumor cytotoxicity. Med Oncol. 2013;30(3):663.

    Article  PubMed  Google Scholar 

  208. Lee HW, Park YM, Lee SJ, Cho HJ, Kim D-H, Lee J-I, et al. Alpha-Smooth Muscle Actin (ACTA2) is required for metastatic potential of human lung adenocarcinoma. Clin Cancer Res. 2013;19(21):5879–89.

    Article  PubMed  CAS  Google Scholar 

  209. Ding Y, Lv C, Zhou Y, Zhang H, Zhao L, Xu Y, et al. Vimentin loss promotes cancer proliferation through up-regulating Rictor/AKT/β-catenin signaling pathway. Exp Cell Res. 2021;405(1): 112666.

    Article  PubMed  CAS  Google Scholar 

  210. Kondo R, Sakamoto N, Harada K, Hashimoto H, Morisue R, Yanagihara K, et al. Cancer-associated fibroblast-dependent and -independent invasion of gastric cancer cells. J Cancer Res Clin Oncol. 2023;149(8):5309–19.

    Article  PubMed  CAS  Google Scholar 

  211. Zhang J, Chen L, Liu X, Kammertoens T, Blankenstein T, Qin Z. Fibroblast-specific protein 1/S100A4-positive cells prevent carcinoma through collagen production and encapsulation of carcinogens. Can Res. 2013;73(9):2770–81.

    Article  CAS  Google Scholar 

  212. Sugimoto H, Mundel TM, Kieran MW, Kalluri R. Identification of fibroblast heterogeneity in the tumor microenvironment. Cancer Biol Ther. 2006;5(12):1640–6.

    Article  PubMed  CAS  Google Scholar 

  213. 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(3):579–96.

    Article  PubMed  PubMed Central  Google Scholar 

  214. Biffi G, Oni TE, Spielman B, Hao Y, Elyada E, Park Y, et al. IL1-induced JAK/STAT signaling is antagonized by TGFβ to shape CAF heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov. 2019;9(2):282–301.

    Article  PubMed  Google Scholar 

  215. Peiffer R, Boumahd Y, Gullo C, Crake R, Letellier E, Bellahcène A, et al. Cancer-associated fibroblast diversity shapes tumor metabolism in pancreatic cancer. Cancers. 2022;15(1):61.

    Article  PubMed  PubMed Central  Google Scholar 

  216. 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(7934):148–54.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  217. Wu SZ, Roden DL, Wang C, Holliday H, Harvey K, Cazet AS, et al. Stromal cell diversity associated with immune evasion in human triple-negative breast cancer. EMBO J. 2020;39(19): e104063.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  218. 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(5):1307.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  219. Kieffer Y, Hocine HR, Gentric G, Pelon F, Bernard C, Bourachot B, et al. Single-cell analysis reveals fibroblast clusters linked to immunotherapy resistance in cancer. Cancer Discov. 2020;10(9):1330–51.

    Article  PubMed  CAS  Google Scholar 

  220. Houthuijzen JM, de Bruijn R, van der Burg E, Drenth AP, Wientjens E, Filipovic T, et al. CD26-negative and CD26-positive tissue-resident fibroblasts contribute to functionally distinct CAF subpopulations in breast cancer. Nat Commun. 2023;14(1):183.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  221. Chung HC, Cho EJ, Lee H, Kim WK, Oh JH, Kim SH, et al. Integrated single-cell RNA sequencing analyses suggest developmental paths of cancer-associated fibroblasts with gene expression dynamics. Clin Transl Med. 2021;11(7): e487.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  222. Lippert AL, Johnson KA, Pasch CA, Kraus SG, Emmerich PB, Clipson L, et al. Abstract 3198: validation and analysis of cancer associated fibroblast subtype markers in metastatic colorectal cancer. Cancer Res. 2022;82(12_Supplement):3198–3198.

    Article  Google Scholar 

  223. Khaliq AM, Erdogan C, Kurt Z, Turgut SS, Grunvald MW, Rand T, et al. Refining colorectal cancer classification and clinical stratification through a single-cell atlas. Genome Biol. 2022;23(1):113.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  224. Liu W, Wang M, Wang M, Liu M. Single-cell and bulk RNA sequencing reveal cancer-associated fibroblast heterogeneity and a prognostic signature in prostate cancer. Medicine. 2023;102(32): e34611.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  225. Grout JA, Sirven P, Leader AM, Maskey S, Hector E, Puisieux I, et al. Spatial positioning and matrix programs of cancer-associated fibroblasts promote T-cell exclusion in human lung tumors. Cancer Discov. 2022;12(11):2606–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  226. Wang H, Li N, Liu Q, Guo J, Pan Q, Cheng B, et al. Antiandrogen treatment induces stromal cell reprogramming to promote castration resistance in prostate cancer. Cancer Cell. 2023;41(7):1345-1362.e1349.

    Article  PubMed  CAS  Google Scholar 

  227. Hong Z, Xie W, Zhuo H, Wei X, Wang K, Cheng J, et al. Crosstalk between cancer cells and cancer-associated fibroblasts mediated by TGF-β1–IGFBP7 signaling promotes the progression of infiltrative gastric cancer. Cancers. 2023;15(15):3965.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  228. Schütz S, Solé-Boldo L, Lucena-Porcel C, Hoffmann J, Brobeil A, Lonsdorf AS, et al. Functionally distinct cancer-associated fibroblast subpopulations establish a tumor promoting environment in squamous cell carcinoma. Nat Commun. 2023;14(1):5413.

    Article  PubMed  PubMed Central  Google Scholar 

  229. Davidson G, Helleux A, Vano YA, Lindner V, Fattori A, Cerciat M, et al. Mesenchymal-like tumor cells and myofibroblastic cancer-associated fibroblasts are associated with progression and immunotherapy response of clear cell renal cell carcinoma. Can Res. 2023;83(17):2952–69.

    Article  CAS  Google Scholar 

  230. Affo S, Nair A, Brundu F, Ravichandra A, Bhattacharjee S, Matsuda M, et al. Promotion of cholangiocarcinoma growth by diverse cancer-associated fibroblast subpopulations. Cancer Cell. 2021;39(6):866-882.e811.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  231. Bhattacharjee S, Hamberger F, Ravichandra A, Miller M, Nair A, Affo S, et al. Tumor restriction by type I collagen opposes tumor-promoting effects of cancer-associated fibroblasts. J Clin Investig. 2021;131(11): e146987.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  232. Schwörer S, Cimino FV, Ros M, Tsanov KM, Ng C, Lowe SW, et al. Hypoxia potentiates the inflammatory fibroblast phenotype promoted by pancreatic cancer cell-derived cytokines. Cancer Res. 2023;83(10):1596–610.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Mello AM, Ngodup T, Lee Y, Donahue KL, Li J, Rao A, et al. Hypoxia promotes an inflammatory phenotype of fibroblasts in pancreatic cancer. Oncogenesis. 2022;11(1):56.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  234. Picard FSR, Lutz V, Brichkina A, Neuhaus F, Ruckenbrod T, Hupfer A, et al. IL-17A-producing CD8(+) T cells promote PDAC via induction of inflammatory cancer-associated fibroblasts. Gut. 2023;72(8):1510–22.

    Article  PubMed  CAS  Google Scholar 

  235. Cui Zhou D, Jayasinghe RG, Chen S, Herndon JM, Iglesia MD, Navale P, et al. Spatially restricted drivers and transitional cell populations cooperate with the microenvironment in untreated and chemo-resistant pancreatic cancer. Nat Genet. 2022;54(9):1390–405.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  236. Zhang X, Zheng S, Hu C, Li G, Lin H, Xia R, et al. Cancer-associated fibroblast-induced lncRNA UPK1A-AS1 confers platinum resistance in pancreatic cancer via efficient double-strand break repair. Oncogene. 2022;41(16):2372–89.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  237. Dings MPG, Manoukian P, Waasdorp C, Quik JSE, Strijker M, Lodestijn SC, et al. Serum levels of iCAF-derived osteoglycin predict favorable outcome in pancreatic cancer. Int J Cancer. 2023;152(3):511–23.

    Article  PubMed  CAS  Google Scholar 

  238. Kim J, Park C, Kim KH, Kim EH, Kim H, Woo JK, et al. Single-cell analysis of gastric pre-cancerous and cancer lesions reveals cell lineage diversity and intratumoral heterogeneity. NPJ Precis Oncol. 2022;6(1):9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  239. Li X, Sun Z, Peng G, Xiao Y, Guo J, Wu B, et al. Single-cell RNA sequencing reveals a pro-invasive cancer-associated fibroblast subgroup associated with poor clinical outcomes in patients with gastric cancer. Theranostics. 2022;12(2):620–38.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  240. Li C, Chen T, Liu J, Wang Y, Zhang C, Guo L, et al. FGF19-induced inflammatory CAF promoted neutrophil extracellular trap formation in the liver metastasis of colorectal cancer. Adv Sci. 2023;10(24):2302613.

    Article  CAS  Google Scholar 

  241. Koncina E, Nurmik M, Pozdeev VI, Gilson C, Tsenkova M, Begaj R, et al. IL1R1+ cancer-associated fibroblasts drive tumor development and immunosuppression in colorectal cancer. Nat Commun. 2023;14(1):4251.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  242. Chen Z, Zhou L, Liu L, Hou Y, Xiong M, Yang Y, et al. Single-cell RNA sequencing highlights the role of inflammatory cancer-associated fibroblasts in bladder urothelial carcinoma. Nat Commun. 2020;11(1):5077.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  243. Lo A, Wang LS, Scholler J, Monslow J, Avery D, Newick K, et al. Tumor-promoting desmoplasia is disrupted by depleting FAP-expressing stromal cells. Cancer Res. 2015;75(14):2800–10.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  244. 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(8):1102–23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  245. 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(6):656-673.e657.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  246. Kerdidani D, Aerakis E, Verrou K-M, Angelidis I, Douka K, Maniou M-A, et al. Lung tumor MHCII immunity depends on in situ antigen presentation by fibroblasts. J Exp Med. 2022;219(2): e20210815.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  247. Iida T, Mizutani Y, Esaki N, Ponik SM, Burkel BM, Weng L, et al. Pharmacologic conversion of cancer-associated fibroblasts from a protumor phenotype to an antitumor phenotype improves the sensitivity of pancreatic cancer to chemotherapeutics. Oncogene. 2022;41(19):2764–77.

    Article  PubMed  CAS  Google Scholar 

  248. Cords L, Tietscher S, Anzeneder T, Langwieder C, Rees M, de Souza N, et al. Cancer-associated fibroblast classification in single-cell and spatial proteomics data. Nat Commun. 2023;14(1):4294.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  249. Wong HY, Sheng Q, Hesterberg AB, Croessmann S, Rios BL, Giri K, et al. Single cell analysis of cribriform prostate cancer reveals cell intrinsic and tumor microenvironmental pathways of aggressive disease. Nat Commun. 2022;13(1):6036.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  250. Ni C, Lou X, Yao X, Wang L, Wan J, Duan X, et al. ZIP1+ fibroblasts protect lung cancer against chemotherapy via connexin-43 mediated intercellular Zn2+ transfer. Nat Commun. 2022;13(1):5919.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  251. Brichkina A, Polo P, Sharma SD, Visestamkul N, Lauth M. A quick guide to CAF subtypes in pancreatic cancer. Cancers. 2023;15(9):2614.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  252. Han C, Liu T, Yin R. Biomarkers for cancer-associated fibroblasts. Biomarker Res. 2020;8(1):64.

    Article  Google Scholar 

  253. Menezes S, Okail MH, Jalil SMA, Kocher HM, Cameron AJM. Cancer-associated fibroblasts in pancreatic cancer: new subtypes, new markers, new targets. J Pathol. 2022;257(4):526–44.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  254. DeLeon-Pennell KY, Barker TH, Lindsey ML. Fibroblasts: the arbiters of extracellular matrix remodeling. Matrix Biol. 2020;91–92:1–7.

    Article  PubMed  PubMed Central  Google Scholar 

  255. Smolgovsky S, Theall B, Wagner N, Alcaide P. Fibroblasts and immune cells: at the crossroad of organ inflammation and fibrosis. Am J Physiol Heart Circ Physiol. 2024;326(2):H303-h316.

    Article  PubMed  CAS  Google Scholar 

  256. 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(8):617–38.

    Article  PubMed  CAS  Google Scholar 

  257. Gauthier V, Kyriazi M, Nefla M, Pucino V, Raza K, Buckley CD, et al. Fibroblast heterogeneity: keystone of tissue homeostasis and pathology in inflammation and ageing. Front Immunol. 2023;14:1137659.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  258. Gomes RN, Manuel F, Nascimento DS. The bright side of fibroblasts: molecular signature and regenerative cues in major organs. NPJ Regen Med. 2021;6(1):43.

    Article  PubMed  PubMed Central  Google Scholar 

  259. Łuszczyński K, Soszyńska M, Komorowski M, Lewandowska P, Zdanowski R, Sobiepanek A, et al. Markers of dermal fibroblast subpopulations for viable cell isolation via cell sorting: a comprehensive review. Cells. 2024;13(14):1206.

    Article  PubMed  PubMed Central  Google Scholar 

  260. Plikus MV, Wang X, Sinha S, Forte E, Thompson SM, Herzog EL, et al. Fibroblasts: origins, definitions, and functions in health and disease. Cell. 2021;184(15):3852–72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  261. Lendahl U, Muhl L, Betsholtz C. Identification, discrimination and heterogeneity of fibroblasts. Nat Commun. 2022;13(1):3409.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  262. Smyth LCD, Highet B, Jansson D, Wu J, Rustenhoven J, Aalderink M, et al. Characterisation of PDGF-BB:PDGFRβ signalling pathways in human brain pericytes: evidence of disruption in Alzheimer’s disease. Commun Biol. 2022;5(1):235.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  263. Zhang Y, Ertl HC. Depletion of FAP+ cells reduces immunosuppressive cells and improves metabolism and functions CD8+T cells within tumors. Oncotarget. 2016;7(17):23282–99.

    Article  PubMed  PubMed Central  Google Scholar 

  264. Adams S, Miller GT, Jesson MI, Watanabe T, Jones B, Wallner BP. PT-100, a small molecule dipeptidyl peptidase inhibitor, has potent antitumor effects and augments antibody-mediated cytotoxicity via a novel immune mechanism. Cancer Res. 2004;64(15):5471–80.

    Article  PubMed  CAS  Google Scholar 

  265. Kakarla S, Chow KK, Mata M, Shaffer DR, Song XT, Wu MF, et al. Antitumor effects of chimeric receptor engineered human T cells directed to tumor stroma. Mol Ther. 2013;21(8):1611–20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  266. Wang LC, 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(2):154–66.

    Article  PubMed  CAS  Google Scholar 

  267. Schuberth PC, Hagedorn C, Jensen SM, Gulati P, van den Broek M, Mischo A, et al. Treatment of malignant pleural mesothelioma by fibroblast activation protein-specific re-directed T cells. J Transl Med. 2013;11:187.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  268. Fischer E, Chaitanya K, Wüest T, Wadle A, Scott AM, van den Broek M, et al. Radioimmunotherapy of fibroblast activation protein positive tumors by rapidly internalizing antibodies. Clin Cancer Res. 2012;18(22):6208–18.

    Article  PubMed  CAS  Google Scholar 

  269. Ostermann E, Garin-Chesa P, Heider KH, Kalat M, Lamche H, Puri C, et al. Effective immunoconjugate therapy in cancer models targeting a serine protease of tumor fibroblasts. Clin Cancer Res. 2008;14(14):4584–92.

    Article  PubMed  CAS  Google Scholar 

  270. Fabre M, Ferrer C, Domínguez-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(13):3420–30.

    Article  PubMed  CAS  Google Scholar 

  271. Tanswell P, Garin-Chesa P, Rettig WJ, Welt S, Divgi CR, Casper ES, et al. Population pharmacokinetics of antifibroblast activation protein monoclonal antibody F19 in cancer patients. Br J Clin Pharmacol. 2001;51(2):177–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  272. Scott AM, Wiseman G, Welt S, Adjei A, Lee FT, 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(5):1639–47.

    PubMed  CAS  Google Scholar 

  273. Hofheinz RD, Al-Batran SE, 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. Onkologie. 2003;26(1):44–8.

    PubMed  CAS  Google Scholar 

  274. Narra K, Mullins SR, Lee HO, 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(11):1691–9.

    Article  PubMed  CAS  Google Scholar 

  275. Eager RM, Cunningham CC, Senzer NN, Stephenson J, Anthony SP, O’Day SJ, et al. Phase II assessment of talabostat and cisplatin in second-line stage IV melanoma. BMC Cancer. 2009;9(1):263.

    Article  PubMed  PubMed Central  Google Scholar 

  276. Aggarwal RR, Costin D, O’Neill VJ, Corsi-Travali S, Adurthi S, Adedoyin A, et al. Phase 1b study of BXCL701, a novel small molecule inhibitor of dipeptidyl peptidases (DPP), combined with pembrolizumab (pembro), in men with metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol. 2020;38(15):e17581–e17581.

    Article  Google Scholar 

  277. Weinberg BA, Gutierrez M, Tesfaye AA, Tan MT, Noel MS, He AR, et al. Phase II trial of BXCL701 and pembrolizumab in patients with metastatic pancreatic ductal adenocarcinoma (EXPEL-PANC). J Clin Oncol. 2023;41(16):TPS4194.

    Article  Google Scholar 

  278. Soerensen MM, Ros W, Rodriguez-Ruiz ME, Robbrecht D, Rohrberg KS, Martin-Liberal J, et al. Safety, PK/PD, and anti-tumor activity of RO6874281, an engineered variant of interleukin-2 (IL-2v) targeted to tumor-associated fibroblasts via binding to fibroblast activation protein (FAP). J Clin Oncol. 2018;36(15):e15155–e15155.

    Article  Google Scholar 

  279. Brünker P, Wartha K, Friess T, Grau-Richards S, Waldhauer I, Koller CF, et al. RG7386, a novel tetravalent FAP-DR5 antibody, effectively triggers FAP-dependent, avidity-driven DR5 hyperclustering and tumor cell apoptosis. Mol Cancer Ther. 2016;15(5):946–57.

    Article  PubMed  Google Scholar 

  280. Bendell J, Blay J-Y, Cassier P, Bauer T, Terret C, Mueller C, et al. Abstract A092: phase 1 trial of RO6874813, a novel bispecific FAP-DR5 antibody, in patients with solid tumors. Mol Cancer Ther. 2018;17(1_Supplement):A092–A092.

    Article  Google Scholar 

  281. Melero I, Tanos T, Bustamante M, Sanmamed MF, Calvo E, Moreno I, et al. A first-in-human study of the fibroblast activation protein-targeted, 4–1BB agonist RO7122290 in patients with advanced solid tumors. Sci Transl Med. 2023;15(695): eabp9229.

    Article  PubMed  CAS  Google Scholar 

  282. Hiltbrunner S, Britschgi C, Schuberth P, Bankel L, Nguyen-Kim TDL, Gulati P, et al. Local delivery of CAR T cells targeting fibroblast activation protein is safe in patients with pleural mesothelioma: first report of FAPME, a phase I clinical trial. Ann Oncol. 2021;32(1):120–1.

    Article  PubMed  CAS  Google Scholar 

  283. Kratochwil C, Flechsig P, Lindner T, Abderrahim L, Altmann A, Mier W, et al. (68)Ga-FAPI PET/CT: tracer uptake in 28 different kinds of cancer. J Nucl Med. 2019;60(6):801–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  284. Baum RP, Schuchardt C, Singh A, Chantadisai M, Robiller FC, Zhang J, et al. Feasibility, biodistribution, and preliminary dosimetry in peptide-targeted radionuclide therapy of diverse adenocarcinomas using (177)Lu-FAP-2286: first-in-humans results. J Nucl Med. 2022;63(3):415–23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  285. Liu Y, Watabe T, Kaneda-Nakashima K, Shirakami Y, Naka S, Ooe K, et al. Fibroblast activation protein targeted therapy using [(177)Lu]FAPI-46 compared with [(225)Ac]FAPI-46 in a pancreatic cancer model. Eur J Nucl Med Mol Imaging. 2022;49(3):871–80.

    Article  PubMed  CAS  Google Scholar 

  286. Zboralski D, Hoehne A, Bredenbeck A, Schumann A, Nguyen M, Schneider E, et al. Preclinical evaluation of FAP-2286 for fibroblast activation protein targeted radionuclide imaging and therapy. Eur J Nucl Med Mol Imaging. 2022;49(11):3651–67.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  287. Capaccione KM, Doubrovin M, Braumuller B, Leibowitz D, Bhatt N, Momen-Heravi F, et al. Evaluating the combined anticancer response of checkpoint inhibitor immunotherapy and FAP-targeted molecular radiotherapy in murine models of melanoma and lung cancer. Cancers. 2022;14(19):4575.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  288. Fu H, Huang J, Sun L, Wu H, Chen H. FAP-targeted radionuclide therapy of advanced radioiodine-refractory differentiated thyroid cancer with multiple cycles of 177Lu-FAPI-46. Clin Nucl Med. 2022;47(10):906–7.

    Article  PubMed  Google Scholar 

  289. Fu H, Huang J, Zhao T, Wang H, Chen Y, Xu W, et al. Fibroblast activation protein-targeted radioligand therapy with 177Lu-EB-FAPI for metastatic radioiodine refractory thyroid cancer: first-in-human, dose-escalation study. Clin Cancer Res. 2023;29:4740–50.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  290. Pandey P, Khan F, Upadhyay TK, Seungjoon M, Park MN, Kim B. New insights about the PDGF/PDGFR signaling pathway as a promising target to develop cancer therapeutic strategies. Biomed Pharmacother. 2023;161: 114491.

    Article  PubMed  CAS  Google Scholar 

  291. Doleschel D, Hoff S, Koletnik S, Rix A, Zopf D, Kiessling F, et al. Regorafenib enhances anti-PD1 immunotherapy efficacy in murine colorectal cancers and their combination prevents tumor regrowth. J Exp Clin Cancer Res. 2021;40(1):288.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  292. Balachandran VP, Cavnar MJ, Zeng S, Bamboat ZM, Ocuin LM, Obaid H, et al. Imatinib potentiates antitumor T cell responses in gastrointestinal stromal tumor through the inhibition of Ido. Nat Med. 2011;17(9):1094–100.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  293. Reilley MJ, Bailey A, Subbiah V, Janku F, Naing A, Falchook G, et al. Phase I clinical trial of combination imatinib and ipilimumab in patients with advanced malignancies. J Immunother Cancer. 2017;5(1):35.

    Article  PubMed  PubMed Central  Google Scholar 

  294. Hirai I, Tanese K, Fukuda K, Fusumae T, Nakamura Y, Sato Y, et al. Imatinib mesylate in combination with pembrolizumab in patients with advanced KIT-mutant melanoma following progression on standard therapy: a phase I/II trial and study protocol. Medicine. 2021;100(49): e27832.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  295. Bauer S, George S, von Mehren M, Heinrich MC. Early and NEXT-GENERATION KIT/PDGFRA kinase inhibitors and the future of treatment for advanced gastrointestinal stromal tumor. Front Oncol. 2021;11: 672500.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  296. Tap WD, Wagner AJ, Schöffski P, Martin-Broto J, Krarup-Hansen A, Ganjoo KN, et al. Effect of doxorubicin plus olaratumab vs doxorubicin plus placebo on survival in patients with advanced soft tissue sarcomas: the ANNOUNCE randomized clinical trial. JAMA. 2020;323(13):1266–76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  297. Schöffski P, Bahleda R, Wagner AJ, Burgess MA, Junker N, Chisamore M, et al. Results of an open-label, phase Ia/b study of pembrolizumab plus olaratumab in patients with unresectable, locally advanced, or metastatic soft-tissue sarcoma. Clin Cancer Res. 2023;29(17):3320–8.

    Article  PubMed  PubMed Central  Google Scholar 

  298. Mega A, Mebrahtu A, Aniander G, Ryer E, Sköld A, Sandegren A, et al. A PDGFRB- and CD40-targeting bispecific AffiMab induces stroma-targeted immune cell activation. MAbs. 2023;15(1):2223750.

    Article  PubMed  PubMed Central  Google Scholar 

  299. Mabry R, Gilbertson DG, Frank A, Vu T, Ardourel D, Ostrander C, et al. A dual-targeting PDGFRbeta/VEGF-A molecule assembled from stable antibody fragments demonstrates anti-angiogenic activity in vitro and in vivo. MAbs. 2010;2(1):20–34.

    Article  PubMed  PubMed Central  Google Scholar 

  300. Bailey JM, Mohr AM, Hollingsworth MA. Sonic hedgehog paracrine signaling regulates metastasis and lymphangiogenesis in pancreatic cancer. Oncogene. 2009;28(40):3513–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  301. Fendrich V, Oh E, Bang S, Karikari C, Ottenhof N, Bisht S, et al. Ectopic overexpression of Sonic Hedgehog (Shh) induces stromal expansion and metaplasia in the adult murine pancreas. Neoplasia. 2011;13(10):923–30.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  302. 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(7):2023–37.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  303. Wong H, Alicke B, West KA, Pacheco P, La H, Januario T, et al. Pharmacokinetic-pharmacodynamic analysis of vismodegib in preclinical models of mutational and ligand-dependent hedgehog pathway activation. Clin Cancer Res. 2011;17(14):4682–92.

    Article  PubMed  CAS  Google Scholar 

  304. Lewis K, Dummer R, Farberg AS, Guminski A, Squittieri N, Migden M. Effects of sonidegib following dose reduction and treatment interruption in patients with advanced basal cell carcinoma during 42-month BOLT trial. Dermatol Ther. 2021;11(6):2225–34.

    Article  Google Scholar 

  305. Ruiz-Borrego M, Jimenez B, Antolín S, García-Saenz JA, Corral J, Jerez Y, et al. A phase Ib study of sonidegib (LDE225), an oral small molecule inhibitor of smoothened or Hedgehog pathway, in combination with docetaxel in triple negative advanced breast cancer patients: GEICAM/2012–12 (EDALINE) study. Invest New Drugs. 2019;37(1):98–108.

    Article  PubMed  CAS  Google Scholar 

  306. Shanshal M, Foster NR, Lou Y, Zhao Y, Seetharam M, Mansfield AS, et al. A phase I trial of sequential dosing of sonidegib and pembrolizumab in advanced solid tumors (aST) and non–small-cell lung cancer (NSCLC). J Clin Oncol. 2023;41(16_suppl):9093–9093.

    Article  Google Scholar 

  307. Bertrand N, Guerreschi P, Basset-Seguin N, Saiag P, Dupuy A, Dalac-Rat S, et al. Vismodegib in neoadjuvant treatment of locally advanced basal cell carcinoma: first results of a multicenter, open-label, phase 2 trial (VISMONEO study): neoadjuvant vismodegib in locally advanced basal cell carcinoma. EClinicalMedicine. 2021;35: 100844.

    Article  PubMed  PubMed Central  Google Scholar 

  308. Kim R, Ji JH, Kim JH, Hong JY, Lim HY, Kang WK, et al. Safety and anti-tumor effects of vismodegib in patients with refractory advanced gastric cancer: a single-arm, phase-II trial. J Cancer. 2022;13(4):1097–102.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  309. De Jesus-Acosta A, Sugar EA, O’Dwyer PJ, Ramanathan RK, Von Hoff DD, Rasheed Z, et al. Phase 2 study of vismodegib, a hedgehog inhibitor, combined with gemcitabine and nab-paclitaxel in patients with untreated metastatic pancreatic adenocarcinoma. Br J Cancer. 2020;122(4):498–505.

    Article  PubMed  Google Scholar 

  310. Sack U, Walther W, Scudiero D, Selby M, Kobelt D, Lemm M, et al. Novel effect of antihelminthic niclosamide on S100A4-mediated metastatic progression in colon cancer. J Natl Cancer Inst. 2011;103(13):1018–36.

    Article  PubMed  CAS  Google Scholar 

  311. Luo F, Luo M, Rong Q-X, Zhang H, Chen Z, Wang F, et al. Niclosamide, an antihelmintic drug, enhances efficacy of PD-1/PD-L1 immune checkpoint blockade in non-small cell lung cancer. J Immunother Cancer. 2019;7(1):245.

    Article  PubMed  PubMed Central  Google Scholar 

  312. Burock S, Daum S, Tröger H, Kim TD, Krüger S, Rieke DT, et al. Niclosamide a new chemotherapy agent? Pharmacokinetics of the potential anticancer drug in a patient cohort of the NIKOLO trial. J Clin Oncol. 2018;36(15_suppl):e14536–e14536.

    Article  Google Scholar 

  313. Takemoto A, Takagi S, Ukaji T, Gyobu N, Kakino M, Takami M, et al. Targeting podoplanin for the treatment of osteosarcoma. Clin Cancer Res. 2022;28(12):2633–45.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  314. Kaneko MK, Nakamura T, Kunita A, Fukayama M, Abe S, Nishioka Y, et al. ChLpMab-23: cancer-specific human-mouse chimeric anti-podoplanin antibody exhibits antitumor activity via antibody-dependent cellular cytotoxicity. Monoclon Antib Immunodiagn Immunother. 2017;36(3):104–12.

    Article  PubMed  CAS  Google Scholar 

  315. Abe S, Kaneko MK, Tsuchihashi Y, Izumi T, Ogasawara S, Okada N, et al. Antitumor effect of novel anti-podoplanin antibody NZ-12 against malignant pleural mesothelioma in an orthotopic xenograft model. Cancer Sci. 2016;107(9):1198–205.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  316. Slemmons KK, Mukherjee S, Meltzer P, Purcell JW, Helman LJ. LRRC15 antibody-drug conjugates show promise as osteosarcoma therapeutics in preclinical studies. Pediatr Blood Cancer. 2021;68(2): e28771.

    Article  PubMed  CAS  Google Scholar 

  317. Demetri GD, Luke JJ, Hollebecque A, Powderly JD 2nd, Spira AI, Subbiah V, et al. First-in-human phase I Study of ABBV-085, an antibody-drug conjugate targeting LRRC15, in sarcomas and other advanced solid tumors. Clin Cancer Res. 2021;27(13):3556–66.

    Article  PubMed  CAS  Google Scholar 

  318. Brunetto E, De Monte L, Balzano G, Camisa B, Laino V, Riba M, et al. The IL-1/IL-1 receptor axis and tumor cell released inflammasome adaptor ASC are key regulators of TSLP secretion by cancer associated fibroblasts in pancreatic cancer. J Immunother Cancer. 2019;7(1):45.

    Article  PubMed  PubMed Central  Google Scholar 

  319. Isambert N, Hervieu A, Hennequin A, Borg C, Rebe C, Derangere V, et al. 5-fluorouracil plus bevacizumab plus anakinra for patients with metastatic colorectal cancer refractory to standard therapies (IRAFU): an investigator-initiated, open-label, single-arm, multicentre, phase 2 study. J Clin Oncol. 2018;36(15_suppl):e15540–e15540.

    Article  Google Scholar 

  320. Costa LJ, Mailankody S, Shaughnessy P, Hari P, Kaufman JL, Larson SM, et al. Anakinra (AKR) prophylaxis (ppx) in patients (pts) with relapsed/refractory multiple myeloma (RRMM) receiving orvacabtagene autoleucel (orva-cel). J Clin Oncol. 2021;39(15_suppl):2537–2537.

    Article  Google Scholar 

  321. Yang K, Han L, Wu S, Qu X, Li Q, Zhao C, et al. Real-world outcomes of regorafenib combined with immune checkpoint inhibitors in patients with advanced or metastatic microsatellite stable colorectal cancer: a multicenter study. Cancer Immunol Immunother. 2022;71(6):1443–51.

    Article  PubMed  CAS  Google Scholar 

  322. Martin-Broto J, Hindi N, Grignani G, Martinez-Trufero J, Redondo A, Valverde C, et al. Nivolumab and sunitinib combination in advanced soft tissue sarcomas: a multicenter, single-arm, phase Ib/II trial. J Immunother Cancer. 2020;8(2): e001561.

    Article  PubMed  PubMed Central  Google Scholar 

  323. Martin CJ, Datta A, Littlefield C, Kalra A, Chapron C, Wawersik S, et al. Selective inhibition of TGFβ1 activation overcomes primary resistance to checkpoint blockade therapy by altering tumor immune landscape. Sci Transl Med. 2020;12(536): eaay8456.

    Article  PubMed  CAS  Google Scholar 

  324. Welsh BT, Faucette R, Bilic S, Martin CJ, Schürpf T, Chen D, et al. Nonclinical development of SRK-181: an anti-latent TGFβ1 monoclonal antibody for the treatment of locally advanced or metastatic solid tumors. Int J Toxicol. 2021;40(3):226–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  325. Yap T, Barve M, Gainor J, Bockorny B, Ju Y, Cote S, et al. 532 First-in-human phase 1 trial of SRK-181: a latent TGFβ1 inhibitor, alone or in combination with anti-PD-(L)1 treatment in patients with advanced solid tumors (DRAGON trial). J Immunother Cancer. 2021;9(Suppl 2):A563–A563.

    Google Scholar 

  326. Morris JC, Tan AR, Olencki TE, Shapiro GI, Dezube BJ, Reiss M, et al. Phase I Study of GC1008 (Fresolimumab): a human anti-transforming growth factor-beta (TGFβ) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PLoS ONE. 2014;9(3): e90353.

    Article  PubMed  PubMed Central  Google Scholar 

  327. Stevenson JP, Kindler HL, Papasavvas E, Sun J, Jacobs-Small M, Hull J, et al. Immunological effects of the TGFβ-blocking antibody GC1008 in malignant pleural mesothelioma patients. Oncoimmunology. 2013;2(8): e26218.

    Article  PubMed  PubMed Central  Google Scholar 

  328. den Hollander MW, Bensch F, Glaudemans AW, Oude Munnink TH, Enting RH, den Dunnen WF, et al. TGF-β antibody uptake in recurrent high-grade glioma imaged with 89Zr-fresolimumab PET. J Nucl Med. 2015;56(9):1310–4.

    Article  Google Scholar 

  329. Greco R, Qu H, Qu H, Theilhaber J, Shapiro G, Gregory R, et al. Pan-TGFβ inhibition by SAR439459 relieves immunosuppression and improves antitumor efficacy of PD-1 blockade. Oncoimmunology. 2020;9(1):1811605.

    Article  PubMed  PubMed Central  Google Scholar 

  330. Robbrecht D, Doger B, Grob J-J, Bechter OE, Miguel MJ, Vieito M, et al. Safety and efficacy results from the expansion phase of the first-in-human study evaluating TGFβ inhibitor SAR439459 alone and combined with cemiplimab in adults with advanced solid tumors. J Clin Oncol. 2022;40(16_suppl):2524–2524.

    Article  Google Scholar 

  331. Tremblay G, Gruosso T, Denis J-F, Figueredo R, Koropatnick J, O’Connor-McCourt M. Abstract 6710: AVID200, a first-in-class selective TGF-beta 1 and -beta 3 inhibitor, sensitizes tumors to immune checkpoint blockade therapies. Cancer Res. 2020;80(16_Supplement):6710–6710.

    Article  Google Scholar 

  332. Yap TA, Lakhani NJ, Araujo DV, Ahnert JR, Chandana SR, Sharma M, et al. AVID200, first-in-class TGF-beta 1 and 3 selective and potent inhibitor: Safety and biomarker results of a phase I monotherapy dose-escalation study in patients with advanced solid tumors. J Clin Oncol. 2020;38(15_suppl):3587–3587.

    Article  Google Scholar 

  333. Lind H, Gameiro SR, Jochems C, Donahue RN, Strauss J, Gulley JM, et al. Dual targeting of TGF-β and PD-L1 via a bifunctional anti-PD-L1/TGF-βRII agent: status of preclinical and clinical advances. J Immunother Cancer. 2020;8(1): e000433.

    Article  PubMed  PubMed Central  Google Scholar 

  334. Burvenich IJG, Goh YW, Guo N, Gan HK, Rigopoulos A, Cao D, et al. Radiolabelling and preclinical characterization of 89Zr-Df-radiolabelled bispecific anti-PD-L1/TGF-βRII fusion protein bintrafusp alfa. Eur J Nucl Med Mol Imaging. 2021;48(10):3075–88.

    Article  PubMed  CAS  Google Scholar 

  335. Cheng B, Ding K, Chen P, Ji J, Luo T, Guo X, et al. Anti-PD-L1/TGF-βR fusion protein (SHR-1701) overcomes disrupted lymphocyte recovery-induced resistance to PD-1/PD-L1 inhibitors in lung cancer. Cancer Commun. 2022;42(1):17–36.

    Article  Google Scholar 

  336. Ravi R, Noonan KA, Pham V, Bedi R, Zhavoronkov A, Ozerov IV, et al. Bifunctional immune checkpoint-targeted antibody-ligand traps that simultaneously disable TGFβ enhance the efficacy of cancer immunotherapy. Nat Commun. 2018;9(1):741.

    Article  PubMed  PubMed Central  Google Scholar 

  337. Keedy VL, Bauer TM, Clarke JM, Hurwitz H, Baek I, Ha I, et al. Association of TGF-β responsive signature with anti-tumor effect of vactosertib, a potent, oral TGF-β receptor type I (TGFBRI) inhibitor in patients with advanced solid tumors. J Clin Oncol. 2018;36(15_suppl):3031–3031.

    Article  Google Scholar 

  338. Kim HS, Ahn J-H, Kim JE, Hong JY, Lee J, Kim SH, et al. A phase I study of TGF-β inhibitor, vactosertib in combination with imatinib in patients with advanced desmoid tumor (aggressive fibromatosis). J Clin Oncol. 2020;38(15):11557–11557.

    Article  Google Scholar 

  339. Kelley RK, Gane E, Assenat E, Siebler J, Galle PR, Merle P, et al. A phase 2 study of galunisertib (TGF-β1 receptor type I inhibitor) and sorafenib in patients with advanced hepatocellular carcinoma. Clin Transl Gastroenterol. 2019;10(7): e00056.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  340. Melisi D, Garcia-Carbonero R, Macarulla T, Pezet D, Deplanque G, Fuchs M, et al. Galunisertib plus gemcitabine vs. gemcitabine for first-line treatment of patients with unresectable pancreatic cancer. Br J Cancer. 2018;119(10):1208–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  341. 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, TGFβ receptor 1 inhibitor, LY3200882, in patients with advanced cancer. Clin Cancer Res. 2021;27(24):6666–76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  342. Malek E, Hwang S, Caimi PF, Metheny LL, Tomlinson BK, Cooper BW, et al. Phase Ib trial of vactosertib in combination with pomalidomide in relapsed multiple myeloma: a corticosteroid-free approach by targeting TGF-β signaling pathway. J Clin Oncol. 2021;39(15):8039–8039.

    Article  Google Scholar 

  343. Kim B-G, Choi SH, Luo G, Sergeeva O, Lee Z, Driscoll J, et al. Vactosertib, a TGF-ß receptor I kinase/ALK5 inhibitor, diminishes tumor progression and bone disease in a mouse model of multiple myeloma and overcomes resistance to proteasome inhibitors. Blood. 2018;132:1918.

    Article  Google Scholar 

  344. Malek E, Kim B-G, Valent J, Driscoll J, Caimi P, Kim S-J, et al. Preclinical studies and a phase I trial of the TGF-β receptor inhibitor, vactosertib (TEW-7197), in combination with pomalidomide in patients with multiple myeloma refractory to bortezomib or lenalidomide. Blood. 2018;132:1962.

    Article  Google Scholar 

  345. Nada H, Sivaraman A, Lu Q, Min K, Kim S, Goo JI, et al. Perspective for discovery of small molecule IL-6 inhibitors through study of structure-activity relationships and molecular docking. J Med Chem. 2023;66(7):4417–33.

    Article  PubMed  CAS  Google Scholar 

  346. Song L, Smith MA, Doshi P, Sasser K, Fulp W, Altiok S, et al. Antitumor efficacy of the anti-interleukin-6 (IL-6) antibody siltuximab in mouse xenograft models of lung cancer. J Thorac Oncol. 2014;9(7):974–82.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  347. Fulciniti M, Hideshima T, Vermot-Desroches C, Pozzi S, Nanjappa P, Shen Z, et al. A high-affinity fully human anti-IL-6 mAb, 1339, for the treatment of multiple myeloma. Clin Cancer Res. 2009;15(23):7144–52.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  348. Dorff TB, Goldman B, Pinski JK, Mack PC, Lara PN Jr, Van Veldhuizen PJ, et al. Clinical and correlative results of SWOG S0354: a phase II Trial of CNTO328 (siltuximab), a monoclonal antibody against interleukin-6, in chemotherapy-pretreated patients with castration-resistant prostate cancer. Clin Cancer Res. 2010;16(11):3028–34.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  349. Schuster M, Rigas JR, Orlov SV, Milovanovic B, Prabhash K, Smith JT, et al. ALD518, a humanized anti-IL-6 antibody, treats anemia in patients with advanced non-small cell lung cancer (NSCLC): results of a phase II, randomized, double-blind, placebo-controlled trial. J Clin Oncol. 2010;28(15):7631–7631.

    Article  Google Scholar 

  350. Huseni MA, Wang L, Klementowicz JE, Yuen K, Breart B, Orr C, et al. CD8(+) T cell-intrinsic IL-6 signaling promotes resistance to anti-PD-L1 immunotherapy. Cell Rep Med. 2023;4(1): 100878.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  351. Li J, Xu J, Yan X, Jin K, Li W, Zhang R. Targeting interleukin-6 (IL-6) sensitizes Anti-PD-L1 treatment in a colorectal cancer preclinical model. Med Sci Monit. 2018;24:5501–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  352. Akce M, Shaib WL, Diab M, Alese OB, Wu C, Thomas S, et al. Phase Ib/II trial of siltuximab and spartalizumab in patients in metastatic pancreatic cancer. J Clin Oncol. 2022;40(4):TPS626.

    Article  Google Scholar 

  353. Shah JJ, Feng L, Thomas SK, Berkova Z, Weber DM, Wang M, et al. Siltuximab (CNTO 328) with lenalidomide, bortezomib and dexamethasone in newly-diagnosed, previously untreated multiple myeloma: an open-label phase I trial. Blood Cancer J. 2016;6(2):e396–e396.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  354. Wang X, Che X, Liu C, Fan Y, Bai M, Hou K, et al. Cancer-associated fibroblasts-stimulated interleukin-11 promotes metastasis of gastric cancer cells mediated by upregulation of MUC1. Exp Cell Res. 2018;368(2):184–93.

    Article  PubMed  CAS  Google Scholar 

  355. Li H, Zhang Q, Wu Q, Cui Y, Zhu H, Fang M, et al. Interleukin-22 secreted by cancer-associated fibroblasts regulates the proliferation and metastasis of lung cancer cells via the PI3K-Akt-mTOR signaling pathway. Am J Transl Res. 2019;11(7):4077–88.

    PubMed  PubMed Central  CAS  Google Scholar 

  356. Wen S, Hou Y, Fu L, Xi L, Yang D, Zhao M, et al. Cancer-associated fibroblast (CAF)-derived IL32 promotes breast cancer cell invasion and metastasis via integrin β3-p38 MAPK signalling. Cancer Lett. 2019;442:320–32.

    Article  PubMed  CAS  Google Scholar 

  357. Sullivan KM, Jiang X, Guha P, Lausted C, Carter JA, Hsu C, et al. Blockade of interleukin 10 potentiates antitumour immune function in human colorectal cancer liver metastases. Gut. 2023;72(2):325–37.

    Article  PubMed  CAS  Google Scholar 

  358. Oft M. IL-10: master switch from tumor-promoting inflammation to antitumor immunity. Cancer Immunol Res. 2014;2(3):194–9.

    Article  PubMed  CAS  Google Scholar 

  359. Naing A, Wong DJ, Infante JR, Korn WM, Aljumaily R, Papadopoulos KP, et al. Pegilodecakin combined with pembrolizumab or nivolumab for patients with advanced solid tumours (IVY): a multicentre, multicohort, open-label, phase 1b trial. Lancet Oncol. 2019;20(11):1544–55.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  360. Hecht JR, Lonardi S, Bendell JC, Sim H-W, Macarulla T, Lopez CD, et al. Randomized Phase III study of FOLFOX alone and with pegilodecakin as second-line therapy in patients with metastatic pancreatic cancer (SEQUOIA). J Clin Oncol. 2020;38(4_suppl):637–637.

    Article  Google Scholar 

  361. Zhang H, Yue J, Jiang Z, Zhou R, Xie R, Xu Y, et al. CAF-secreted CXCL1 conferred radioresistance by regulating DNA damage response in a ROS-dependent manner in esophageal squamous cell carcinoma. Cell Death Dis. 2017;8(5):e2790–e2790.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  362. Miyake M, Hori S, Morizawa Y, Tatsumi Y, Nakai Y, Anai S, et al. CXCL1-mediated interaction of cancer cells with tumor-associated macrophages and cancer-associated fibroblasts promotes tumor progression in human bladder cancer. Neoplasia. 2016;18(10):636–46.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  363. Murakami K, Sasaki Y, Furuya H, Rosser C. Abstract 2858: development of a first-in-class humanized antibody targeting CXCL1 in bladder cancer. Cancer Res. 2022;82(12_Supplement):2858–2858.

    Article  Google Scholar 

  364. Zhai J, Shen J, Xie G, Wu J, He M, Gao L, et al. Cancer-associated fibroblasts-derived IL-8 mediates resistance to cisplatin in human gastric cancer. Cancer Lett. 2019;454:37–43.

    Article  PubMed  CAS  Google Scholar 

  365. Davar D, Simonelli M, Gutierrez M, Calvo E, Melear J, Piha-Paul S, et al. 394 Interleukin-8–neutralizing monoclonal antibody BMS-986253 plus nivolumab (NIVO) in biomarker-enriched, primarily anti–PD-(L)1–experienced patients with advanced cancer: initial phase 1 results. J Immunother Cancer. 2020;8(Suppl 3):A239–40.

    Google Scholar 

  366. Liu G, Sun J, Yang ZF, Zhou C, Zhou PY, Guan RY, et al. Cancer-associated fibroblast-derived CXCL11 modulates hepatocellular carcinoma cell migration and tumor metastasis through the circUBAP2/miR-4756/IFIT1/3 axis. Cell Death Dis. 2021;12(3):260.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  367. Gao Q, Wang S, Chen X, Cheng S, Zhang Z, Li F, et al. Cancer-cell-secreted CXCL11 promoted CD8<sup>+</sup> T cells infiltration through docetaxel-induced-release of HMGB1 in NSCLC. J Immunother Cancer. 2019;7(1):42.

    Article  PubMed  PubMed Central  Google Scholar 

  368. Cao Y, Jiao N, Sun T, Ma Y, Zhang X, Chen H, et al. CXCL11 correlates with antitumor immunity and an improved prognosis in colon cancer. Front Cell Dev Biol. 2021;9: 646252.

    Article  PubMed  PubMed Central  Google Scholar 

  369. 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(32):4428–42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  370. Labernadie A, Kato T, Brugués A, Serra-Picamal X, Derzsi S, Arwert E, et al. A mechanically active heterotypic E-cadherin/N-cadherin adhesion enables fibroblasts to drive cancer cell invasion. Nat Cell Biol. 2017;19(3):224–37.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  371. Jiang H, Ge H, Shi Y, Yuan F, Yue H. CAFs secrete CXCL12 to accelerate the progression and cisplatin resistance of colorectal cancer through promoting M2 polarization of macrophages. Med Oncol. 2023;40(3):90.

    Article  PubMed  CAS  Google Scholar 

  372. Holter JC, Chang CW, Avendano A, Garg AA, Verma AK, Charan M, et al. Fibroblast-derived CXCL12 increases vascular permeability in a 3-D microfluidic model independent of extracellular matrix contractility. Front Bioeng Biotechnol. 2022;10: 888431.

    Article  PubMed  PubMed Central  Google Scholar 

  373. Halama N, Williams A, Prüfer U, Frömming A, Beyer D, Eulberg D, et al. Abstract CT117: Phase 1/2 study with CXCL12 inhibitor NOX-A12 and pembrolizumab in patients with microsatellite-stable, metastatic colorectal or pancreatic cancer. Cancer Res. 2020;80(16_Supplement):CT117.

    Article  Google Scholar 

  374. Giordano FA, Layer JP, Leonardelli S, Friker LL, Seidel C, Schaub C, et al. Radiotherapy and olaptesed pegol (NOX-A12) in partially resected or biopsy-only MGMT-unmethylated glioblastoma: interim data from the German multicenter phase 1/2 GLORIA trial. J Clin Oncol. 2022;40(16_suppl):2050–2050.

    Article  Google Scholar 

  375. 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(1):129–47.

    Article  PubMed  CAS  Google Scholar 

  376. Li Z, Zhou J, Zhang J, Li S, Wang H, Du J. Cancer-associated fibroblasts promote PD-L1 expression in mice cancer cells via secreting CXCL5. Int J Cancer. 2019;145(7):1946–57.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  377. Xu H, Zhao J, Li J, Zhu Z, Cui Z, Liu R, et al. Cancer associated fibroblast-derived CCL5 promotes hepatocellular carcinoma metastasis through activating HIF1α/ZEB1 axis. Cell Death Dis. 2022;13(5):478.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  378. Christenson E, Lim SJ, Wang H, Ferguson A, Parkinson R, Cetasaan Y, et al. Nivolumab and a CCR2/CCR5 dual antagonist (BMS-813160) with or without GVAX for locally advanced pancreatic ductal adenocarcinomas: results of phase I study. J Clin Oncol. 2023;41(4_suppl):730–730.

    Article  Google Scholar 

  379. Korbecki J, Kupnicka P, Chlubek M, Gorący J, Gutowska I, Baranowska-Bosiacka I. CXCR2 receptor: regulation of expression, signal transduction, and involvement in cancer. Int J Mol Sci. 2022;23(4):2168.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  380. Guo C, Sharp A, Vogl U, Colombo I, Stathis A, Jain S, et al. 454O A phase (Ph) I/II trial of the CXCR2 antagonist AZD5069 in combination with enzalutamide (ENZA) in patients (pts) with metastatic castration resistant prostate cancer (mCRPC). Ann Oncol. 2022;33:S745.

    Article  Google Scholar 

  381. Guo C, Sharp A, Gurel B, Crespo M, Figueiredo I, Jain S, et al. Targeting myeloid chemotaxis to reverse prostate cancer therapy resistance. Nature. 2023;623(7989):1053–61.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  382. Pawlick RL, Wink J, Pepper AR, Bruni A, Abualhassen N, Rafiei Y, et al. Reparixin, a CXCR1/2 inhibitor in islet allotransplantation. Islets. 2016;8(5):115–24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  383. Liotti F, De Pizzol M, Allegretti M, Prevete N, Melillo RM. Multiple anti-tumor effects of Reparixin on thyroid cancer. Oncotarget. 2017;8(22):35946–61.

    Article  PubMed  PubMed Central  Google Scholar 

  384. Goldstein LJ, Mansutti M, Levy C, Chang JC, Henry S, Fernandez-Perez I, et al. A randomized, placebo-controlled phase 2 study of paclitaxel in combination with reparixin compared to paclitaxel alone as front-line therapy for metastatic triple-negative breast cancer (fRida). Breast Cancer Res Treat. 2021;190(2):265–75.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  385. Redman J, Spira A, Javle M, Kelly K, Pavlakis N, Jehl G, et al. P-133 Clinical responses in pancreaticobiliary cancer patients who received bintrafusp alfa (BA) or BA plus CXCR1/2 inhibitor (SX-682) plus CEA/MUC1-targted vaccine (CV301). Ann Oncol. 2022;33:S297.

    Article  Google Scholar 

  386. Dunne RF, Ullman NA, Belt BA, Ruffolo LI, Burchard P, Hezel AF, et al. A phase I study to evaluate the safety and tolerability of SX-682 in combination with PD-1 inhibitor as maintenance therapy for unresectable pancreatic adenocarcinoma. J Clin Oncol. 2022;40(4_suppl):TPS631.

    Article  Google Scholar 

  387. Johnson B, Haymaker C, Morris VK, Dasari A, Higbie VS, Shen JP, et al. Abstract CT118: A phase I/II trial of a CXCR1/2 inhibitor in combination with anti-PD-1 for circulating tumor DNA (ctDNA) positive & refractoryRAS-mutated microsatellite stable (MSS) colorectal cancer. Cancer Res. 2023;83(8_supplement):CT118.

    Article  Google Scholar 

  388. De Clercq E. Mozobil® (Plerixafor, AMD3100), 10 years after its approval by the US Food and Drug Administration. Antivir Chem Chemother. 2019;27:2040206619829382.

    Article  PubMed  PubMed Central  Google Scholar 

  389. Ghobrial IM, Liu CJ, Zavidij O, Azab AK, Baz R, Laubach JP, et al. Phase I/II trial of the CXCR4 inhibitor plerixafor in combination with bortezomib as a chemosensitization strategy in relapsed/refractory multiple myeloma. Am J Hematol. 2019;94(11):1244–53.

    Article  PubMed  CAS  Google Scholar 

  390. Bockorny B, Semenisty V, Macarulla T, Borazanci E, Wolpin BM, Stemmer SM, et al. BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: the COMBAT trial. Nat Med. 2020;26(6):878–85.

    Article  PubMed  CAS  Google Scholar 

  391. Hainsworth JD, Reeves JA, Mace JR, Crane EJ, Hamid O, Stille JR, et al. A randomized, open-label phase 2 study of the CXCR4 inhibitor LY2510924 in combination with sunitinib versus sunitinib alone in patients with metastatic renal cell carcinoma (RCC). Target Oncol. 2016;11(5):643–53.

    Article  PubMed  Google Scholar 

  392. Salgia R, Stille JR, Weaver RW, McCleod M, Hamid O, Polzer J, et al. A randomized phase II study of LY2510924 and carboplatin/etoposide versus carboplatin/etoposide in extensive-disease small cell lung cancer. Lung Cancer. 2017;105:7–13.

    Article  PubMed  Google Scholar 

  393. Ghobrial IM, Liu C-J, Redd RA, Perez RP, Baz R, Zavidij O, et al. A phase Ib/II trial of the first-in-class anti-CXCR4 antibody ulocuplumab in combination with lenalidomide or bortezomib plus dexamethasone in relapsed multiple myeloma. Clin Cancer Res. 2020;26(2):344–53.

    Article  PubMed  CAS  Google Scholar 

  394. Seitz S, Dreyer TF, Stange C, Steiger K, Bräuer R, Scheutz L, et al. CXCL9 inhibits tumour growth and drives anti-PD-L1 therapy in ovarian cancer. Br J Cancer. 2022;126(10):1470–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  395. Xiao W, Huang H, Zheng P, Liu Y, Chen Y, Chen J, et al. The CXCL10/CXCR3 pathway contributes to the synergy of thermal ablation and PD-1 blockade therapy against tumors. Cancers. 2023;15(5):1427.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  396. House IG, Savas P, Lai J, Chen AXY, Oliver AJ, Teo ZL, et al. Macrophage-derived CXCL9 and CXCL10 are required for antitumor immune responses following immune checkpoint blockade. Clin Cancer Res. 2020;26(2):487–504.

    Article  PubMed  CAS  Google Scholar 

  397. Simon G, Subbiah V, Rosen L, Lenz H-J, Park H, Patel M, et al. 762 First-in-human phase 1a study of NG-641, a tumour-selective vector expressing a FAP-TAc bispecific antibody and immune enhancer module, in patients with metastatic/advanced epithelial tumours (STAR). J Immunother Cancer. 2022;10(Suppl 2):A794–A794.

    Google Scholar 

  398. Ding X, Ji J, Jiang J, Cai Q, Wang C, Shi M, et al. HGF-mediated crosstalk between cancer-associated fibroblasts and MET-unamplified gastric cancer cells activates coordinated tumorigenesis and metastasis. Cell Death Dis. 2018;9(9):867.

    Article  PubMed  PubMed Central  Google Scholar 

  399. Bauman JE, Saba NF, Roe D, Bauman JR, Kaczmar J, Bhatia A, et al. Randomized Phase II trial of ficlatuzumab with or without cetuximab in pan-refractory, recurrent/metastatic head and neck cancer. J Clin Oncol. 2023;41(22):3851–62.

    Article  PubMed  CAS  Google Scholar 

  400. Mok TSK, Geater SL, Su W-C, Tan E-H, Yang JC-H, Chang G-C, et al. A randomized Phase 2 study comparing the combination of ficlatuzumab and gefitinib with gefitinib alone in asian patients with advanced stage pulmonary adenocarcinoma. J Thorac Oncol. 2016;11(10):1736–44.

    Article  PubMed  Google Scholar 

  401. Camidge DR, Moran T, Demedts I, Grosch H, Mercurio J-PD, Mileham KF, et al. A randomized, open-label, phase 2 study of emibetuzumab plus erlotinib (LY+E) and emibetuzumab monotherapy (LY) in patients with acquired resistance to erlotinib and MET diagnostic positive (MET Dx+) metastatic NSCLC. J Clin Oncol. 2016;34(15):9070–9070.

    Article  Google Scholar 

  402. Harding JJ, Zhu AX, Bauer TM, Choueiri TK, Drilon A, Voss MH, et al. A phase Ib/II study of ramucirumab in combination with emibetuzumab in patients with advanced cancer. Clin Cancer Res. 2019;25(17):5202–11.

    Article  PubMed  CAS  Google Scholar 

  403. Cunningham D, Tebbutt NC, Davidenko I, Murad AM, Al-Batran S-E, Ilson DH, et al. Phase III, randomized, double-blind, multicenter, placebo (P)-controlled trial of rilotumumab (R) plus epirubicin, cisplatin and capecitabine (ECX) as first-line therapy in patients (pts) with advanced MET-positive (pos) gastric or gastroesophageal junction (G/GEJ) cancer: RILOMET-1 study. J Clin Oncol. 2015;33(15):4000–4000.

    Article  Google Scholar 

  404. Spigel DR, Ervin TJ, Ramlau RA, Daniel DB, Goldschmidt JH Jr, Blumenschein GR Jr, et al. Randomized phase II trial of Onartuzumab in combination with erlotinib in patients with advanced non-small-cell lung cancer. J Clin Oncol. 2013;31(32):4105–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  405. Mathieu LN, Larkins E, Akinboro O, Roy P, Amatya AK, Fiero MH, et al. FDA approval summary: capmatinib and tepotinib for the treatment of metastatic NSCLC harboring MET Exon 14 skipping mutations or alterations. Clin Cancer Res. 2022;28(2):249–54.

    Article  PubMed  CAS  Google Scholar 

  406. Ryoo B-Y, Ren Z, Kim T-Y, Pan H, Rau K-M, Choi H, et al. Phase II trial of tepotinib vs sorafenib in Asian patients (pts) with advanced hepatocellular carcinoma (HCC). Ann Oncol. 2018;29:viii207.

    Article  Google Scholar 

  407. Mok TSK, Cortinovis DL, Majem M, Johnson ML, Mardjuadi FI, Zhao X, et al. Efficacy and safety of capmatinib plus pembrolizumab in treatment (tx)-naïve patients with advanced non–small cell lung cancer (NSCLC) with high tumor PD-L1 expression: results of a randomized, open-label, multicenter, phase 2 study. J Clin Oncol. 2022;40(16_suppl):9118–9118.

    Article  Google Scholar 

  408. Babina IS, Turner NC. Advances and challenges in targeting FGFR signalling in cancer. Nat Rev Cancer. 2017;17(5):318–32.

    Article  PubMed  CAS  Google Scholar 

  409. Krook MA, Reeser JW, Ernst G, Barker H, Wilberding M, Li G, et al. Fibroblast growth factor receptors in cancer: genetic alterations, diagnostics, therapeutic targets and mechanisms of resistance. Br J Cancer. 2021;124(5):880–92.

    Article  PubMed  CAS  Google Scholar 

  410. van Brummelen EMJ, Levchenko E, Dómine M, Fennell DA, Kindler HL, Viteri S, et al. A phase Ib study of GSK3052230, an FGF ligand trap in combination with pemetrexed and cisplatin in patients with malignant pleural mesothelioma. Invest New Drugs. 2020;38(2):457–67.

    Article  PubMed  Google Scholar 

  411. Morgensztern D, Karaseva N, Felip E, Delgado I, Burdaeva O, Dómine M, et al. An open-label phase IB study to evaluate GSK3052230 in combination with paclitaxel and carboplatin, or docetaxel, in FGFR1-amplified non-small cell lung cancer. Lung Cancer. 2019;136:74–9.

    Article  PubMed  Google Scholar 

  412. Wekking D, Pretta A, Martella S, D’Agata AP, Joeun Choe J, Denaro N, et al. Fibroblast growth factor receptors as targets for anticancer therapy in cholangiocarcinomas and urothelial carcinomas. Heliyon. 2023;9(9): e19541.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  413. Siefker-Radtke AO, Necchi A, Park SH, García-Donas J, Huddart RA, Burgess EF, et al. Efficacy and safety of erdafitinib in patients with locally advanced or metastatic urothelial carcinoma: long-term follow-up of a phase 2 study. Lancet Oncol. 2022;23(2):248–58.

    Article  PubMed  CAS  Google Scholar 

  414. Feng Y-H, Su W-C, Oh D-Y, Shen L, Kim K-P, Liu X, et al. Updated analysis with longer follow up of a phase 2a study evaluating erdafitinib in Asian patients (pts) with advanced cholangiocarcinoma (CCA) and fibroblast growth factor receptor (FGFR) alterations. J Clin Oncol. 2022;40(4_suppl):430–430.

    Article  Google Scholar 

  415. Abou-Alfa GK, Sahai V, Hollebecque A, Vaccaro G, Melisi D, Al-Rajabi R, et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: a multicentre, open-label, phase 2 study. Lancet Oncol. 2020;21(5):671–84.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  416. Coombes RC, Badman PD, Lozano-Kuehne JP, Liu X, Macpherson IR, Zubairi I, et al. Results of the phase IIa RADICAL trial of the FGFR inhibitor AZD4547 in endocrine resistant breast cancer. Nat Commun. 2022;13(1):3246.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  417. Chae YK, Vaklavas C, Cheng HH, Hong F, Harris L, Mitchell EP, et al. Molecular analysis for therapy choice (MATCH) arm W: Phase II study of AZD4547 in patients with tumors with aberrations in the FGFR pathway. J Clin Oncol. 2018;36(15_suppl):2503–2503.

    Article  Google Scholar 

  418. Javle M, Roychowdhury S, Kelley RK, Sadeghi S, Macarulla T, Weiss KH, et al. Infigratinib (BGJ398) in previously treated patients with advanced or metastatic cholangiocarcinoma with FGFR2 fusions or rearrangements: mature results from a multicentre, open-label, single-arm, phase 2 study. Lancet Gastroenterol Hepatol. 2021;6(10):803–15.

    Article  PubMed  Google Scholar 

  419. Lassman AB, Sepúlveda-Sánchez JM, Cloughesy TF, Gil-Gil MJ, Puduvalli VK, Raizer JJ, et al. Infigratinib in patients with recurrent gliomas and FGFR alterations: a multicenter phase II study. Clin Cancer Res. 2022;28(11):2270–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  420. Sternberg CN, Petrylak DP, Bellmunt J, Nishiyama H, Necchi A, Gurney H, et al. FORT-1: Phase II/III study of rogaratinib versus chemotherapy in patients with locally advanced or metastatic urothelial carcinoma selected based on FGFR1/3 mRNA expression. J Clin Oncol. 2023;41(3):629–39.

    Article  PubMed  CAS  Google Scholar 

  421. Doi T, Shitara K, Kojima T, Kuboki Y, Matsubara N, Bando H, et al. Phase I study of the irreversible fibroblast growth factor receptor 1–4 inhibitor futibatinib in Japanese patients with advanced solid tumors. Cancer Sci. 2023;114(2):574–85.

    Article  PubMed  CAS  Google Scholar 

  422. Cleary JM, Iyer G, Oh D-Y, Mellinghoff IK, Goyal L, Ng MCH, et al. Final results from the phase I study expansion cohort of the selective FGFR inhibitor Debio 1,347 in patients with solid tumors harboring an FGFR gene fusion. J Clin Oncol. 2020;38(15):3603–3603.

    Article  Google Scholar 

  423. Michael M, Bang Y-J, Park YS, Kang Y-K, Kim TM, Hamid O, et al. A Phase 1 study of LY2874455, an oral selective pan-FGFR inhibitor, in patients with advanced cancer. Target Oncol. 2017;12(4):463–74.

    Article  PubMed  Google Scholar 

  424. Escudier B, Grünwald V, Ravaud A, Ou Y-C, Castellano D, Lin C-C, et al. Phase II results of dovitinib (TKI258) in patients with metastatic renal cell cancer. Clin Cancer Res. 2014;20(11):3012–22.

    Article  PubMed  CAS  Google Scholar 

  425. Heinrich MC, von Mehren M, Demetri GD, Fletcher JA, Sun JG, Kerstein D, et al. Ponatinib efficacy and safety in patients (pts) with advanced gastrointestinal stromal tumors (GIST) after tyrosine kinase inhibitor (TKI) failure: results from a phase 2 study. J Clin Oncol. 2015;33(15):10535–10535.

    Article  Google Scholar 

  426. Reck M, Kaiser R, Mellemgaard A, Douillard J-Y, Orlov S, Krzakowski M, et al. Docetaxel plus nintedanib versus docetaxel plus placebo in patients with previously treated non-small-cell lung cancer (LUME-Lung 1): a phase 3, double-blind, randomised controlled trial. Lancet Oncol. 2014;15(2):143–55.

    Article  PubMed  CAS  Google Scholar 

  427. Ellinghaus P, Neureiter D, Nogai H, Stintzing S, Ocker M. Patient selection approaches in FGFR inhibitor trials-many paths to the same end? Cells. 2022;11(19):3180.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  428. Wainberg ZA, Enzinger PC, Kang Y-K, Qin S, Yamaguchi K, Kim I-H, et al. Bemarituzumab in patients with FGFR2b-selected gastric or gastro-oesophageal junction adenocarcinoma (FIGHT): a randomised, double-blind, placebo-controlled, phase 2 study. Lancet Oncol. 2022;23(11):1430–40.

    Article  PubMed  CAS  Google Scholar 

  429. Siefker-Radtke AO, Lugowska I, Tupikowski K, Andric ZG, Rezazadeh Kalebasty A, Curigliano G, et al. 917P - Clinical activity of vofatamab (V), an FGFR3 selective antibody in combination with pembrolizumab (P) in metastatic urothelial carcinoma (mUC), updated interim analysis of FIERCE-22. Ann Oncol. 2019;30: v365.

    Article  Google Scholar 

  430. Kim SB, Meric-Bernstam F, Kalyan A, Babich A, Liu R, Tanigawa T, et al. First-in-human phase I study of aprutumab ixadotin, a fibroblast growth factor receptor 2 antibody-drug conjugate (BAY 1187982) in patients with advanced cancer. Target Oncol. 2019;14(5):591–601.

    Article  PubMed  PubMed Central  Google Scholar 

  431. Kollmannsberger C, Britten CD, Olszanski AJ, Walker JA, Zang W, Willard MD, et al. A phase 1 study of LY3076226, a fibroblast growth factor receptor 3 (FGFR3) antibody-drug conjugate, in patients with advanced or metastatic cancer. Invest New Drugs. 2021;39(6):1613–23.

    Article  PubMed  CAS  Google Scholar 

  432. McCarthy JB, El-Ashry D, Turley EA. Hyaluronan, cancer-associated fibroblasts and the tumor microenvironment in malignant progression. Front Cell Dev Biol. 2018;6:48.

    Article  PubMed  PubMed Central  Google Scholar 

  433. Zhang Z, Tao D, Zhang P, Liu X, Zhang Y, Cheng J, et al. Hyaluronan synthase 2 expressed by cancer-associated fibroblasts promotes oral cancer invasion. J Exp Clin Cancer Res. 2016;35(1):181.

    Article  PubMed  PubMed Central  Google Scholar 

  434. Provenzano PP, Hingorani SR. Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer. Br J Cancer. 2013;108(1):1–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  435. Thompson CB, Shepard HM, O’Connor PM, Kadhim S, Jiang P, Osgood RJ, et al. Enzymatic depletion of tumor hyaluronan induces antitumor responses in preclinical animal models. Mol Cancer Ther. 2010;9(11):3052–64.

    Article  PubMed  CAS  Google Scholar 

  436. 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(13):1062–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  437. Van Cutsem E, Tempero MA, Sigal D, Oh DY, 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(27):3185–94.

    Article  PubMed  PubMed Central  Google Scholar 

  438. Zhen DB, Whittle M, Ritch PS, Hochster HS, Coveler AL, George B, et al. Phase II study of PEGPH20 plus pembrolizumab for patients (pts) with hyaluronan (HA)-high refractory metastatic pancreatic adenocarcinoma (mPC): PCRT16–001. J Clin Oncol. 2022;40(4_suppl):576–576.

    Article  Google Scholar 

  439. Ko AH, Kim KP, Siveke JT, Lopez CD, Lacy J, O’Reilly EM, et al. Atezolizumab plus PEGPH20 versus chemotherapy in advanced pancreatic ductal adenocarcinoma and gastric cancer: MORPHEUS Phase Ib/II umbrella randomized study platform. Oncologist. 2023;28(6):553-e472.

    Article  PubMed  PubMed Central  Google Scholar 

  440. Garcia-Carbonero R, Bazan-Peregrino M, Gil-Martín M, Álvarez R, Macarulla T, Riesco-Martinez MC, et al. Phase I, multicenter, open-label study of intravenous VCN-01 oncolytic adenovirus with or without nab-paclitaxel plus gemcitabine in patients with advanced solid tumors. J Immunother Cancer. 2022;10(3): e003255.

    Article  PubMed  PubMed Central  Google Scholar 

  441. Chronopoulos A, Robinson B, Sarper M, Cortes E, Auernheimer V, Lachowski D, et al. ATRA mechanically reprograms pancreatic stellate cells to suppress matrix remodelling and inhibit cancer cell invasion. Nat Commun. 2016;7:12630.

    Article  PubMed  PubMed Central  Google Scholar 

  442. 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(3):286–96.

    Article  PubMed  PubMed Central  Google Scholar 

  443. Kocher HM, Basu B, Froeling FEM, Sarker D, Slater S, Carlin D, et al. Phase I clinical trial repurposing all-trans retinoic acid as a stromal targeting agent for pancreatic cancer. Nat Commun. 2020;11(1):4841.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  444. Tobin RP, Cogswell DT, Cates VM, Davis DM, Borgers JSW, Van Gulick RJ, et al. Targeting MDSC differentiation using ATRA: a phase I/II clinical trial combining pembrolizumab and all-trans retinoic acid for metastatic melanoma. Clin Cancer Res. 2023;29(7):1209–19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  445. Zhao ZX, Zhang YQ, Sun H, Chen ZQ, Chang JJ, Wang X, et al. Calcipotriol abrogates cancer-associated fibroblast-derived IL-8-mediated oxaliplatin resistance in gastric cancer cells via blocking PI3K/Akt signaling. Acta Pharmacol Sin. 2023;44(1):178–88.

    Article  PubMed  CAS  Google Scholar 

  446. Gorchs L, Ahmed S, Mayer C, Knauf A, Fernández Moro C, Svensson M, et al. The vitamin D analogue calcipotriol promotes an anti-tumorigenic phenotype of human pancreatic CAFs but reduces T cell mediated immunity. Sci Rep. 2020;10(1):17444.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  447. Akiba T, Morikawa T, Odaka M, Nakada T, Kamiya N, Yamashita M, et al. Vitamin D supplementation and survival of patients with non–small cell lung cancer: a randomized, double-blind, placebo-controlled trial. Clin Cancer Res. 2018;24(17):4089–97.

    Article  PubMed  CAS  Google Scholar 

  448. Evans TRJ, Colston KW, Lofts FJ, Cunningham D, Anthoney DA, Gogas H, et al. A phase II trial of the vitamin D analogue Seocalcitol (EB1089) in patients with inoperable pancreatic cancer. Br J Cancer. 2002;86(5):680–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  449. Ng K, Nimeiri HS, McCleary NJ, Abrams TA, Yurgelun MB, Cleary JM, et al. Effect of high-dose vs standard-dose vitamin D3 supplementation on progression-free survival among patients with advanced or metastatic colorectal cancer: the SUNSHINE randomized clinical trial. JAMA. 2019;321(14):1370–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  450. 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. Cancer Res. 2018;78(5):1321–33.

    Article  PubMed  CAS  Google Scholar 

  451. Banerjee S, Saluja A. Minnelide, a novel drug for pancreatic and liver cancer. Pancreatology. 2015;15(4 Suppl):S39-43.

    Article  PubMed  CAS  Google Scholar 

  452. Modi S, Giri B, Gupta VK, Lavania S, Sethi V, Sharma NS, et al. Minnelide synergizes with conventional chemotherapy by targeting both cancer and associated stroma components in pancreatic cancer. Cancer Lett. 2022;537: 215591.

    Article  PubMed  CAS  Google Scholar 

  453. Chauhan VP, Chen IX, Tong R, Ng MR, Martin JD, Naxerova K, et al. Reprogramming the microenvironment with tumor-selective angiotensin blockers enhances cancer immunotherapy. Proc Natl Acad Sci USA. 2019;116(22):10674–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  454. Tranel J, Palm S, Graves SA, Feng FY, Hope TA. Impact of radiopharmaceutical therapy (177Lu, 225Ac) microdistribution in a cancer-associated fibroblasts model. EJNMMI Phys. 2022;9(1):67.

    Article  PubMed  PubMed Central  Google Scholar 

  455. Mediavilla-Varela M, Boateng K, Noyes D, Antonia SJ. The anti-fibrotic agent pirfenidone synergizes with cisplatin in killing tumor cells and cancer-associated fibroblasts. BMC Cancer. 2016;16:176.

    Article  PubMed  PubMed Central  Google Scholar 

  456. Hauge A, Rofstad EK. Antifibrotic therapy to normalize the tumor microenvironment. J Transl Med. 2020;18(1):207.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

AMS was supported by NHRMC Investigator Grant (No. 1177837). IJGB is a National Imaging Facility Fellow.

Author information

Authors and Affiliations

Authors

Contributions

ZC and AMS designed the review. ZC, SQ, and SA collected the literature and wrote the manuscript. LDO, IJB, PWJ, and AMS revised the manuscript. All authors read, reviewed, and approved the manuscript.

Corresponding authors

Correspondence to Zhipeng Cao or Andrew M. Scott.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no conflict of interest.

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 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cao, Z., Quazi, S., Arora, S. et al. Cancer-associated fibroblasts as therapeutic targets for cancer: advances, challenges, and future prospects. J Biomed Sci 32, 7 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12929-024-01099-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12929-024-01099-2

Keywords