
Cancer‑associated fibroblasts in human malignancies, with a particular emphasis on sarcomas (Review)
- Authors:
- Published online on: August 6, 2025 https://doi.org/10.3892/ijo.2025.5785
- Article Number: 79
-
Copyright: © Benesova et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Over the last few decades, the prognosis of patients with cancer has improved thanks to various treatment plans and therapies, including immunological and biological approaches (1,2). Despite these therapeutic advancements, however, the incidence of cancer has increased, and cancer remains one of the leading causes of mortality (3). Although numerous factors have been shown to drive the onset, biological behavior and spread of cancer, the specific composition of the tumor microenvironment (TME) has been demonstrated to have an important role in these processes, and this has therefore been increasingly studied (4).
The TME consists of acellular and cellular components. The acellular portion contains multiple biologically active molecules, including cytokines and growth factors, whereas the cellular portion consists of tumor cells, immune cells, fibroblasts and endothelial cells (5). In the complex interplay of cellular constituents within the TME, cancer-associated fibroblasts (CAFs) have emerged as major contributors to TME dynamics. CAFs represent a heterogenous subset of fibroblasts that are characterized by their activation state (6). The development of CAFs, however, remains largely unknown, although several putative explanatory mechanisms have been proposed (6).
CAFs have been extensively studied in various types of carcinomas, although their role in sarcomas remains largely unexplored. This lack of exploration is probably due to the fundamental differences in the origins of carcinomas and sarcomas, which thereby influence the roles that CAFs have in the pathophysiology and treatment of these diseases. Carcinomas originate from epithelial tissues, whereas sarcomas arise from mesenchymal tissues. Epithelial cells line the body's organs and participate in a number of different functions, including absorption, secretion, excretion, filtration and protection. Carcinomas are typically found in internal organs (for example, the lungs, breast and colon) and skin. On the other hand, mesenchymal tissues are supportive and connective tissues, including bones, tendons, cartilage, vascular tissue, muscle and fat tissues. A significant portion of connective tissues is composed of extracellular matrix (ECM) produced by fibroblasts, and this varies between each subtype. Although the precise function of each specialized tissue differs, fundamental functions they hold in common include transport, support, defense, repair and tissue connections. Owing to the multifunctionality of mesenchymal cells, sarcomas may occur in any part of the body, with the extremities (75%) and retroperitoneum and trunk walls (10%) being the most common sites (7).
The differences that exist in the origin of these cancers are especially important for understanding the distinct roles of CAFs. In carcinomas, CAFs are known to exert a significant influence both on tumor growth and progression and on the therapeutic responsiveness of tumors through their interactions with epithelial cancer cells. However, in sarcomas, which originate from mesenchymal tissues, CAFs may have different roles due to their shared lineage and gene expression profiles with the tumor cells. This may represent both a challenge and an opportunity in terms of studying their role in the context of sarcomas. For example, an overlap between certain sarcoma subtypes and fibroblasts and the expression of the same genes may render the analysis of CAFs in the context of sarcomas challenging; however, it may also offer advantages in terms of treatment possibilities.
The present review aimed to summarize the current knowledge regarding the role of CAFs in human malignancies, with a particular focus on sarcomas, and to integrate this knowledge with the already-established knowledge of CAFs in carcinomas wherever possible.
Developmental processes underlying the formation of CAFs, and their basic classification
CAFs may originate from various cells within the TME (8); however, CAFs are mostly derived from local tissue-resident fibroblasts. The tissue-resident fibroblasts initially surround the lesion and undergo a process known as stromagenesis (8). During this process, normal fibroblasts are transformed into pro-tumorigenic fibroblasts, which subsequently continue to surround the lesion (6).
Research studies have classified CAFs in numerous ways, and the majority of these have divided CAFs into three major types based on their functional properties: Myofibroblasts (myCAFs), inflammatory CAFs (iCAFs) and antigen-presenting CAFs (apCAFs) (9) (Fig. 1). MyCAFs and iCAFs are found in virtually all tumors (10). On the other hand, apCAFs are typically derived from mesothelial cells, and these have been identified more recently (11). They are considered to primarily modulate immunity through the expression of the major histocompatibility complex II (MHC II) (12). These three subtypes have evolved through various evolutionary pathways, thereby acquiring their diverse functions and clinical relevance across different tumor types (13).
Contributions made by CAFs to the dynamics of the TME
CAFs engage in crosstalk with cancer cells to modulate the tumor stroma, thereby altering its metabolism, inducing angiogenesis and promoting local inflammation, all processes which, when combined, lead to the promotion (14) or inhibition (15) of tumorigenesis.
Pro-tumorigenic activities of CAFs
There are multiple mechanisms through which CAFs elicit a range of pro-tumorigenic processes. One mechanism through which CAFs are able to facilitate tumorigenesis involves epithelial-mesenchymal transition (EMT). EMT is a physiological process that is implicated in embryonic development, wound healing and stem cell behavior (16). However, during tumor development, EMT also fosters fibrosis and promotes cancer progression (17). CAFs have also been demonstrated to produce multiple biologically active molecules, including cytokines and growth factors, such as transforming growth factor-β (TGF-β), platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), matrix metalloproteinase 9 (MMP9) and multiple interleukins (ILs) (8,18). All these molecules contribute to CAF-promoted tumorigenesis and its associated processes, such as angiogenesis (19,20).
Under physiological conditions, normal fibroblasts are at rest, and have little to no metabolic activity prior to their activation (21). However, this 'normality' is disrupted in the TME, where CAFs become permanently stimulated and undergo metabolic reprogramming. The reprogrammed fibroblasts subsequently initiate a support for tumor growth and cancer cell invasion (22). This support is materialized through the secretion of ECM proteins and the supply of energy and nutrients to the TME (6), the latter of which is mediated by catabolic processes in the activated CAFs, leading to the release of nutrients and metabolites into the TME, including lactate, ketone bodies, fatty acids and glutamine (23). The catabolic processes in CAFs are driven by autophagy and this process is initiated and driven by cancer cells, which undergo stress in the TME. This stress leads to the activation of multiple signaling pathways, resulting in the generation of reactive oxygen species, which, in turn, induce catabolic processes in CAFs (24). Specifically, the lactate that is released from CAFs causes acidification of the TME; this, in turn, activates MMP9, thereby contributing to the development of chemoresistance (25).
Further enhancement of the CAF-mediated support of cancer cells and tumor development and growth is mediated via pro-inflammatory cytokines. Cancer cells and CAFs both secrete pro-inflammatory cytokines such as IL-6 and IL-1, which promote further epigenetic modifications of CAFs (26). This process subsequently provides CAFs with the capacity to promote tumor development through enhancing actomyosin contractility and ECM remodeling, which thereby facilitates the migration of cancer cells (27). A previous study of Biffi et al (28) suggested that the secretion of multiple cytokines may even provide the main driving force that shapes the heterogeneity of CAFs in the TME.
An essential property of CAFs within the TME lies in their capacity to induce immunosuppression. This is achieved through the formation of fibrotic barriers, which have the effect of impeding the infiltration of immune cells, notably cytotoxic CD8+ T cells, into the TME (29,30). CAF- or cancer cell-produced collagen (31,32), which is the main composite of the fibrotic barriers (33), has also been shown to inhibit the effector (cytotoxic) functions of the tumor-infiltrating cytotoxic CD8+ T cells through the immune checkpoint molecule, leukocyte-associated immunoglobulin-like receptor 1 (34,35). Another important immunosuppressive feature is the capacity of CAFs to recruit or induce polarization of myeloid-derived suppressor cells (MDSCs), T cell-suppressing neutrophils and regulatory T cells (Tregs), subsequently inhibiting CD8+ T cell effector functions (10,36). In addition to this, as antigen presenters, CAFs have also been shown to prompt the transformation of naive CD4+ T cells into antigen-specific Tregs (12), thereby promoting the suppression of antigen-specific anti-tumor immunity.
Anti-tumorigenic activities of CAFs
Overall, the role of CAFs in cancer treatment has been considered to be mainly pro-tumorigenic. However, studies have indicated that, under certain conditions, CAFs are also able to exert anti-tumorigenic effects. For example, an inflammatory subpopulation of CAFs has been found to be associated with improved overall survival rates in patients with colorectal cancer (37), and their depletion was found to accelerate pancreatic cancer, thereby reducing survival (38). The mechanisms underlying their anti-tumorigenic activities may be that CAFs either limit tumor growth (39) or inhibit tumor cell proliferation (40).
The dynamic phenotypes of CAFs may also be interchanged (37). For instance, CAFs were shown to be modulated by the vitamin D analog calcipotriol into an anti-tumorigenic phenotype in pancreatic cancer (41). In the same study, CAFs under the influence of calcipotriol were shown to have a decreased production of several pro-tumorigenic factors, including prostaglandin E2, IL-6 and leukemia inhibitory factor (41). These findings provide further evidence of the little-known anti-tumorigenic activities of CAF populations, which may, in the future, force us to modify our views on their role in cancer and how they may be therapeutically targeted (15). This is especially important in the context of sarcomas, where their pro- or anti-tumorigenic activities are, at present, largely unexplored.
Identification and role of CAFs in different types of carcinomas
The identification of CAFs is often challenging due to high heterogeneity and lack of discernible differences in surface markers compared with normal fibroblasts (6). Nevertheless, CAFs have been traditionally described to exhibit increased expression levels of markers, including α-smooth muscle actin (α-SMA), fibroblast-specific protein 1, fibroblast activation protein (FAP), PDGF receptor (PDGFR) and vimentin (42). However, a novel single-cell sequencing study revealed a much higher complexity of distinct CAF subpopulations based on different gene programs (43); moreover, CAF subpopulation phenotypes may vary among different types of carcinomas.
Breast cancer
Breast cancer is the most frequently diagnosed type of cancer among women globally and is one of the leading causes of death in females (44). Over the past decade, notable advances in its treatment have been made, and these are attributable to the introduction of novel therapeutic agents, including anti-HER2 monoclonal antibodies and cell cycle inhibitors (45,46). Despite these improvements, however, advanced and metastatic breast cancer remains incurable and continues to be a major contributor to cancer-associated deaths.
CAFs exhibit different phenotypes in breast cancer (47), and these phenotypes are currently categorized according to the expression of surface markers into the following subtypes: CAF-S1, CAF-S2, CAF-S3 and CAF-S4 (48). Whereas CAF-S2 and CAF-S3 are also found in healthy tissues, CAF-S1 and CAF-S4 are only present in tumors and metastatic lymph nodes (48), and have been shown to have higher expression levels of α-SMA, a marker often associated with poorer prognosis (49). The CAF-S1 type is known for its utilization of C-X-C motif chemokine ligand 12 (CXCL12)/TGF-β-dependent signaling pathways, whereas CAF-S4 exerts its contractile abilities in promoting cancer cell motility through Notch-driven signaling pathways (50). Therefore, the presence of CAF-S1 and CAF-S4 in breast cancer has been assumed to be associated with a worse prognosis and the development of metastases (50). Other contributors to the CAF-mediated promotion of aggressive tumor behavior are the NOD-like receptor (NLR) family pyrin domain containing 3 (NLRP3)/IL-1β signaling pathway (51) and exosomal microRNAs released from CAFs, both of which promote cancer cell stemness and EMT (52-55).
Pancreatic cancer
Pancreatic cancer remains one of the most lethal malignancies, with a 5-year survival rate of ~10% (56). The majority of patients are diagnosed at an advanced stage due to the absence of early symptoms, or the presence of non-specific symptoms during the localized stage of the disease (56). Surgical resection remains the cornerstone of curative treatment for patients with small, localized pancreatic tumors, providing significant survival benefits. However, for patients with unresectable, metastatic or recurrent pancreatic cancer, surgical resection can only offer limited benefit. Management of metastatic or recurrent disease primarily involves chemotherapy, with or without targeted therapy. Given the limitations of current treatments in terms of achieving a cure, it is recommended to identify patients who may benefit from other potentially effective therapies, such as immunotherapy with immune checkpoint inhibitors (ICIs), or those who may be suitable candidates for clinical trials (57).
In pancreatic cancer, CAFs have also been shown to play a crucial role in shaping the TME and disease progression. Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer, and also one of the leading causes of cancer-associated mortality globally (58). Its common occurrence is mainly attributed to the delayed detection of tumors and limited therapy options. Pancreatic tumors are characterized by a dense stroma that is rich in CAFs (59). CAFs in pancreatic cancer can also be divided into subpopulations on the basis of their origin. CAFs infiltrating PDAC mostly originate from resident fibroblasts, pancreatic stellate cells, tumor-infiltrating mesenchymal stem cells (MSCs) or mesothelial cells. In the majority of cases, three subsets of CAFs in pancreatic cancer are recognized: iCAFs, myCAFs and apCAFs (60). MyCAFs express α-SMA molecules, whereas iCAFs are known rather for the secretion of pro-inflammatory cytokines, including IL-6. ApCAFs are characterized by their high expression levels of MHC II (61). The three identified subsets of CAFs within pancreatic tumors exhibit distinct functional profiles that are tailored to support various aspects of pancreatic cancer progression distributed across different spatial regions within the TME. MyCAFs are identified close to the tumor margin, whereas IL-6-positive iCAFs are predominantly observed in regions that were more distant from the tumor (62). IL-6 has been demonstrated to be secreted by iCAFs through paracrine signaling from the malignant epithelium, which is particularly relevant since elevated levels of IL-6 are generally indicative of a higher risk of developing extensive hepatic metastases in patients with PDAC (63,64). The balance between the myCAF and iCAF phenotypes is relatively dynamic and this balance is mainly regulated by differential engagement of the IL-1/JAK/STAT and TGF-β signaling pathways (28). The presence of iCAFs in PDAC has been shown to be associated with the activation of NF-κB signaling, as evidenced by reduced iCAF gene expression upon specific inhibition of NF-κB signaling. Activated NF-κB signaling in iCAFs has been shown to induce gene expression associated with tumor progression (65). Regarding apCAFs, these cells are highly potent antigen presenters, whose interaction with naive CD4+ T cells leads to the induction of antigen-specific Tregs (11). In PDAC, there is also evidence to suggest that CAF-induced regulation is mediated via extracellular vesicles (EVs) (66). Moreover, chemotherapy-exposed CAFs have also been shown to regulate the survival and proliferation of cancer cells, as they respond to the therapy through releasing increased numbers of EVs, with a concomitant increase in the level of the chemoresistance-inducing factor Snail, thereby promoting cancer cell survival and chemotherapy drug resistance (67).
Intraductal papillary mucinous neoplasms (IPMNs) are pancreatic cystic tumors that are derived from cells lining the pancreatic ducts (68). The association between IPMNs and PDAC is both unclear and complex; however, it has been demonstrated that IPMN lesions may increase the risk of developing PDAC (69). It has also been shown that iCAFs are abundant in PDAC, and myCAFs are abundant in high-grade IPMNs. This may suggest that iCAFs could contribute to the transformation of IPMNs into PDAC (70).
Hepatocellular carcinoma
Primary liver cancer is the sixth most prevalent cancer worldwide, and the third leading cause of cancer-associated deaths. Hepatocellular carcinoma is the predominant type of primary liver cancer, comprising 75-86% of all cases of liver cancer. The incidence of hepatocellular carcinoma is two to three times higher in men compared with women, and these higher rates of incidence and mortality for men have been shown for most countries. At present, the treatment landscape for hepatocellular carcinoma is undergoing a major transformation, with combinations of different immunotherapies emerging as key components alongside targeted therapies (71-73).
Hepatocellular carcinoma typically arises from fibrosis and cirrhosis, and is characterized by an abundance of stromal myofibroblasts (74). Similarly to other types of tumor, CAFs are associated with poor prognosis in cases of hepatocellular carcinoma, and they have been shown to promote the migration and invasion of hepatocellular carcinoma cancer cells. Moreover, Liu et al (75) described an associated activation of the Hedgehog/TGF-β signaling pathway in hepatocellular carcinoma in response to several CAF-secreted chemokines. CAFs in hepatocellular carcinoma have been shown to promote cancer cell proliferation by secreting hepatocyte growth factor, thereby preventing cancer cells from undergoing necrosis (76). CAFs in hepatocellular carcinoma are also reported to enhance the stemness of cancer cells through producing hepatocyte growth factor and IL-6 (77,78). There is also emerging evidence to suggest that CAFs may also provide an immunosuppressive TME in hepatocellular carcinoma, as they attract MDSCs (79) and neutrophils that are subsequently primed by CAFs to impair T cell function via programmed cell death protein 1 (PD-1)-mediated inhibition (36).
CAFs have also been shown to make an important contribution to therapy resistance in hepatocellular carcinoma. For example, CAF-secreted phosphoprotein 1 was identified as the most important factor responsible for the resistance of hepatocellular carcinoma cells to treatment with sorafenib and lenvatinib, both of which are offered as first-line systemic therapy for advanced hepatocellular carcinoma (80,81). Moreover, in an animal model of hepatocellular carcinoma, a higher abundance of CAFs was also demonstrated to be associated with low sensitivity of tumors to anti-PD-1 immunotherapy (82).
Colorectal carcinoma
Colorectal cancer ranks as the third most frequently diagnosed cancer and a trend of increasing incidence has been reported among young adults in the 21st century (83). Colorectal cancer ranks among the top five leading causes of cancer-associated mortality in both men and women globally (84,85). Of patients with colorectal cancer, ~50% develop liver metastases, which markedly contribute to the lethality of the disease. For metastatic colorectal cancer, doublet or triplet chemotherapy should be offered to patients with initially unresectable metastatic colorectal cancer, with specific therapies based on tumor characteristics such as microsatellite instability and the RAS mutation status. Cytoreductive surgery in combination with chemotherapy is often considered for colorectal peritoneal metastases. For patients with potentially curable liver metastases, however, perioperative chemotherapy or surgery alone is recommended. In all cases, multidisciplinary team management and shared decision-making are crucial (86,87).
CAFs are abundantly present in colorectal cancer, and various CAF markers (for example, a-SMA, PDGFR-B, FAP and ferroptosis suppressor protein-1) are often found to be expressed in different stages of the disease (88). A study by Wikberg et al (89) demonstrates the importance of the localization of the expression of these markers: Patients with colorectal cancer exhibiting high FAP expression in the tumor center, but not in the tumor periphery, were shown to have poorer prognosis, as FAP was expressed more highly in tumors with positive microsatellite instability, suggesting FAP to be a potential negative prognostic factor.
CAF-derived EVs also have a role in colorectal cancer. These EVs are capable of increasing cancer cell stemness and EMT and, therefore, of promoting metastasis and resistance to therapy (90). A study of Aizawa et al (91) described the secretion of Wnt family member 2 (Wnt2) by CAFs as an important cancer-promoting mechanism. The high expression of Wnt2 in CAFs was also found to correlate with the depth of the tumor, lymph node metastasis, TNM stage, venous invasion and disease recurrence.
Lung and bronchus carcinoma
As one of the most commonly diagnosed cancers, lung cancer is also the major cause of cancer-associated deaths globally (92). Of all lung cancer cases, ~85% are of the non-small-cell lung cancer (NSCLC) type. Due to the lack of effective screening programs and the late appearance of symptoms, however, the majority of patients are diagnosed at an advanced stage, which results in a very poor prognosis (93). Over the last 20 years, major progress has been achieved in the treatment of NSCLC, which has, at the same time, deepened our understanding of its biology and tumor progression. Small-molecule tyrosine kinase inhibitors and immunotherapy have provided significant survival benefits for specific patients. Nevertheless, the overall cure and survival rates for NSCLC, especially for the cancer in its metastatic stages, remain low (92,94).
During the disease progression, CAFs fulfill a crucial role in the constitution of a supportive niche for cancer stemness, EMT and cancer cell growth (95,96), suggesting that CAFs act as feeder cells in lung cancer (97). EMT induced by CAFs has been shown to enhance the capability of cancer cells to migrate and form the secondary tumor foci via several molecular mechanisms. Secretion of IL-6 by CAFs has been described as driving the EMT via the subsequent activation of JAK2/STAT3 signaling (98,99). CAFs, as described previously, have been shown to regulate cancer cells via exosomal transfer. In lung cancer, CAFs have been shown to promote cell migration and invasion by triggering the PTEN/PI3K/Akt pathway with EVs via miR-210 (100). CAFs were also found to perform a crucial role in TME remodeling (101). CAFs were reported to promote the recruitment of CCR2+ monocytes via the production of chemokine (C-C motif) ligand 2. The recruited monocytes were subsequently polarized by CAFs into cells with the MDSC phenotype, causing the robust suppression of adaptive anti-tumor immunity, specifically CD8+ T-cell proliferation and interferon γ (IFN-γ) production.
In recent years, the use of ICIs has elicited significant results in several cancer diagnoses. In lung cancer, however, only a small portion of patients were found to respond to this type of therapy (102). This low responsiveness has been attributed to CAFs, as they have been shown to prevent immune cell infiltration, specifically of the cytotoxic CD8+ T cells, resulting in remodeling of the ECM, thereby promoting immune escape (103,104). Therefore, several recent studies have suggested targeting CAFs as a potential path to overcome ICI therapy resistance in lung cancer (105-107).
Prostate carcinoma
Prostate cancer is one of the most common malignancies in men, predominantly affecting those over the age of 65 (92,108). The majority of prostate cancers are adenocarcinomas, which typically originate in the peripheral part of the gland, and often progress slowly (109). For clinically localized cases, standard treatments include radical prostatectomy and radiation therapy. Androgen ablation, which involves either bilateral orchiectomy or the use of luteinizing hormone-releasing hormone agonists or anti-androgens, has been demonstrated to reduce tumor burden, thereby extending median progression-free survival (110). Although the treatment is initially effective, the majority of patients on hormonal therapy eventually develop castration-resistant prostate cancer, with ~70% of patients progressing to metastatic disease, primarily in the bones (80-90% of advanced and metastatic cases) (111,112).
Prostate CAFs have a crucial role in driving prostate carcinoma progression, influencing the supportive microenvironment through age-associated stromal changes and cancer-specific alterations (113,114). Multiple sources contribute to the heterogeneity of CAFs in prostate cancer, including resident fibroblasts undergoing EMT (115,116), bone marrow-derived MSCs recruited through chemotaxis (117), senescent stromal cells that promote tumor progression (118) and pericytes that potentially resemble CAFs (116). CAFs exhibit diverse functions within the prostate cancer TME, categorized into myCAFs, apCAFs and immune-regulatory CAFs (119). In particular, myCAFs occupy a central role in ECM remodeling, promoting collagen deposition and cancer cell growth, upon which they induce ECM stiffness and morphological alterations (120-122).
CAFs are sensitive to androgen levels and contribute to resistance to anti-androgen therapies, thereby promoting the development of castration-resistant prostate cancer (123-125). CAFs have been shown to mediate resistance to anti-androgen therapies through modulating androgen receptor signaling and enhancing cytokine secretion (126,127). Moreover, CAFs were found to decrease sensitivity to anti-androgens and chemotherapies through various signaling pathways, including the PI3K/Akt pathway (128,129).
Understanding the involvement of CAFs in human sarcomas
Sarcomas are a rare and heterogeneous group of tumors, currently divided into at least 100 different types, and the number of types continues to increase as further new markers are introduced (130,131). However, sarcomas are traditionally divided into two main groups: Soft tissue sarcoma (STS) and bone sarcoma (Fig. 2). Among the STSs, the most common types in adults are liposarcoma (LPS), leiomyosarcoma (LMS) and undifferentiated pleomorphic sarcoma (UPS), whereas the most common sarcoma types in children and adolescents are rhabdomyosarcoma (RMS) and osteosarcoma. Bone sarcomas are represented by osteosarcoma, Ewing sarcoma (EWS) and chondrosarcoma (Fig. 2).
The genetic expression and developmental processes of sarcomas closely resemble those of fibroblasts/CAFs, reflecting their shared mesenchymal tissue origin. This shared origin suggests that CAFs in sarcomas may exhibit distinct behaviors and interactions compared with those in carcinomas. Given that fibroblasts are crucial components of adipose tissue and bone, their presence and function in the TME of sarcomas may be markedly more complex, with substantial variations existing between different types of sarcomas.
STS
STS accounts for ~1% of all malignancies in adults (36). STS can be formed anywhere in the body, although the extremities are the most common sites (7). The most prevalent subtypes of STS are LPS, LMS and UPS. Among other highly prevalent subtypes of STS are myxofibrosarcoma, synovial sarcoma and RMS in children (7). The TME varies extensively within each histological subtype, and also according to its anatomic location and the tumor stage (132); the same holds true for fibroblasts. Since fibroblasts can acquire a wide variety of phenotypes, and given their specific functions at distinct locations of the body (133), they should be evaluated individually within each histological subset of STS.
LPS
LPS arises from adipose tissues and represents one of the most common types of STSs in adults, representing ~20% of all cases of adult STS (134). LPS predominantly manifests in individuals aged 50-70, with an equal incidence in both sexes. The anatomical distribution of LPS is wide, with each subtype characterized by distinct morphological features, distinctive natural history, and specific genetic alterations crucial for diagnosis (134). According to the latest World Health Organization classification, LPS is divided into five subtypes: Well-differentiated LPS, dedifferentiated LPS (DDLPS), myxoid LPS (MLPS), pleomorphic LPS (PLPS) and myxoid pleomorphic LPS (MPLPS) (134).
The primary approach for treating all LPS subtypes is surgical resection. The degree of resection required depends on the location of the tumor and its histopathological characteristics. Achieving a complete gross resection is paramount for all sarcomas. The conventional first-line therapeutic approach for unresectable or metastatic disease involves systemic chemotherapy that is based on anthracyclines. Any decision to use adjuvant therapy for patients with high-risk LPS should be made on an individual basis; nevertheless, the chemotherapeutic sensitivity of LPS remains grossly low (134).
Fibroblasts are an important component of adipose tissue. Adipocytes regulate their size through expansion or shrinkage, and therefore there is an urgent requirement for continual tissue remodeling. ECM produced by fibroblasts provides structural support and has the function of storing growth factors and cytokines and also has the function of protecting adipocytes from mechanical stress (135). Moreover, adipocytes appear to be able to generate CAFs (42,136). Specifically, it has been shown that adipocytes, in the presence of cancer cells, are able to differentiate into cancer-associated adipocytes (CAAs) that express matrix remodeling proteins, leading to the overproduction of ECM. These CAAs subsequently give rise to adipocyte-derived fibroblasts, which fuel migratory properties in various types of cancer (137,138).
Our knowledge regarding the role of CAFs in LPS is relatively poor. Harati et al (139) isolated CAFs from one surgically removed high-grade PLPS and one intermediate MLPS. An in vitro co-culture with the LPS SW872 cell line induced cell proliferation and an increased viability of cancer cells compared with co-culture with dermal fibroblasts (139). Moreover, the co-cultivation of CAFs originating from high-grade PLPS led to a further enhanced proliferative capability of the cancer cells. In agreement with these results, Xu et al (140) observed increased proliferative and migratory capabilities of various LPS cell lines when co-cultured with CAFs isolated from retroperitoneal DDLPS. Notably, the secretion of thrombospondin-2 by LPS cells was shown to induce the transformation of fibroblasts into CAFs in vitro. Therefore, thrombospondin-2 may be an attractive therapeutic target for LPS (140). CAFs were also found to be able to influence clinicopathological features of retroperitoneal LPS. It was shown that the expression of α-SMA was correlated with tumor size, grade and subtype; however, it had no effect on overall survival, disease-free survival or recurrence-free survival (140). CAFs were also shown to influence the efficacy of chemotherapeutics (141) and it appears that their presence could decrease the sensitivity of LPS to the chemotherapeutic drug doxorubicin (139).
RMS
RMS is the most common STS in children and adolescents, classified as a high-grade neoplasm of skeletal myoblast-like cells (142). RMS comprises two primary subtypes, alveolar and embryonal, initially identified by their light-microscope characteristics, each of which are driven by different molecular mechanisms and presenting unique clinical challenges. Effective treatment requires control of the primary tumor, which can develop in various anatomical locations, as well as managing disseminated disease, assumed to be present in all cases.
RMS are typically skeletal muscle tumors. However, RMS may develop on sites that normally lack skeletal muscles as well. This raises a question about RMS origins; one of the proposed hypotheses points to mesenchymal progenitor cells. These cells may circulate between organs, which could explain RMS occurrence at sites without skeletal muscles. Moreover, these cells may give rise to fibroblasts as well (143).
Skeletal muscle fibroblasts are mainly located in the space between muscle fibers. Although they represent a small fraction of cells within the muscle, they have an irreplaceable role in terms of supporting force transduction, maintaining muscle structure and responding to muscle injuries. Recently, fibroblasts were also detected at the border of the perimysium and endomysium (133,144).
It has been shown that there is a limited crosstalk of CAFs and RMS cells. Cancer cells typically remodel the ECM on their own, and express functional and structural components, including metalloproteinases, fibronectin and laminin. By contrast, RMS cells secrete macrophage migration inhibitory factor (MIF), which induces activation of MAPK, enhances cell adhesion and increases tumor vascularization, but decreases the recruitment of CAFs. Moreover, MIF was shown to impede the migration of cells toward the IL-8 gradient (145). RMS has also been found to secrete IL-8 under hypoxic conditions (146). Since its receptor C-X-C motif chemokine receptor 2 (CXCR2) is expressed by fibroblasts (147,148), this MIF-IL-8 axis could explain the decreased presence of CAFs within the TME of RMS. These findings diverge from the typical behavior observed in a number of types of cancer, where tumor progression and metastasis are often facilitated by the presence and activity of CAFs (141).
On the other hand, EVs derived from fusion-positive or -negative RMS have been shown to stimulate fibroblast proliferation and invasiveness (149). Detailed analysis has revealed that the presence of transmembrane glycoprotein CD147 on RMS-derived EVs was critical for the induction of these properties in BJ fibroblasts (150). Moreover, CD147 was identified to be one of the key molecules required for EV uptake by BJ fibroblasts (150).
LMS
LMS is a common type of STS, and ~50% of these tumors are formed in the retroperitoneum or intra-abdominal regions (151). Managing retroperitoneal LMS is challenging due to the tendency of this sarcoma subtype to metastasize rather than to recur locally. The lungs and liver are primary sites for metastasis, and once metastasis occurs, survival rates decrease markedly, with a median survival rate of only 13 months (152).
Surgery is the main treatment for LMS, and complete removal of the tumor is essential for improving survival. However, retroperitoneal tumors frequently recur, even after a number of years post-diagnosis (153). Tumor grade and size are crucial factors affecting overall survival (154). Therefore, addressing the risk of distant metastasis is critical in treating retroperitoneal LMS. Moreover, the prognosis of patients with retroperitoneal LMS remains poor, mainly since these tumors are typically already large at the time of initial detection (155).
LMSs are classically divided into extra-uterine and uterine subtypes (156). Myofibroblasts represent smooth muscle fibroblasts, and these are morphologically enlarged, interconnected through gap junctions and facilitate contractile force and tissue repair (157). To date, only a limited number of studies have investigated the role of CAFs within LMS. It was shown by Nagao et al (158) that uterine LMSs secrete specific miRNA in EVs, which has the effect of inducing the transformation of fibroblasts into CAFs. Further studies, however, are warranted to clarify the role of CAFs within the LMS microenvironment.
UPS
UPS is characterized as a high-grade and aggressive STS, and is one of the three most common histotypes of STSs that are associated with the poorest prognosis (159). This malignancy may be manifested in various locations, including soft tissues, bones and the retroperitoneum, and it has the potential to metastasize to multiple organs (160). The primary treatment strategy for UPS involves surgical excision, with the goal of achieving microscopically negative margins. In certain cases, the use of neoadjuvant or adjuvant radiotherapy and chemotherapy may be considered to enhance treatment outcomes (160).
The origin of UPS has been controversial for a long time. Formerly classified as malignant fibrous histiocytoma, the cells were thought to originate from histiocytoma (161,162). However, later studies have challenged this hypothesis (163,164), showing that the cells have uncertain differentiation due to the absence of differentiation markers, and that they originate from often highly transformed cells at early stages of their development from MSCs (165). The cells of UPS are extremely pleiomorphic, acquiring numerous shapes and sizes, thereby making it difficult to visually distinguish them from other cell types, including fibroblasts or CAFs (32,166-168). Based on this perspective, the identification of CAFs in UPS represents an even greater challenge, and at present, hardly any studies have been published that were directly dedicated to exploring the role of CAFs in UPS.
Bone sarcoma
Bone sarcoma is a group of rare tumors with distinct incidence, which most commonly appear in children and adolescents (133). It consists of osteosarcoma, EWS and chondrosarcoma (133). Fibroblasts are directly involved in cartilage degradation through matrix-degrading enzymes (133,169,170). They affect bone erosion via cathepsin K, and regulate the osteoclast/osteoblast axis; moreover, they are able to give rise to bone-forming osteoblasts (133,169,170).
EWS
EWS is an aggressive sarcoma standing at the 'crossroads' between bone and soft tissue cancer. The treatment for EWS typically involves starting with neoadjuvant chemotherapy, followed by surgical resection, which is subsequently followed by adjuvant chemotherapy. For patients with metastatic disease, the control of metastatic sites is usually postponed until after the adjuvant chemotherapy has been administered. However, although there are clear treatment plans for newly diagnosed cases, the approach to managing recurrent EWS requires individualized strategies (171).
As aforementioned, EWS usually occurs in children and young adults, with peak incidence in adolescents (172). Differences in gene pathways with prognostic effect have been determined only in EWS with stromal content (173). This underlines the need to study the tumor stroma and associated underlying mechanisms that lead to tumor progression. Notably, not only stromal cells may have the capability to produce the ECM; CD73+ CAF-like tumor cells have been described in EWS. These cells deposit pro-tumorigenic ECM proteins, thereby contributing to tissue remodeling. These are termed EWS/FLI1-low cells, which are predominantly present along tumor borders and invasive fronts (174).
Osteosarcoma
Osteosarcoma is a bone cancer, with a peak occurrence during the second decade of life. The current treatment for osteosarcoma primarily involves surgery in conjunction with adjuvant chemotherapy. However, due to the rapid progression of the disease and the development of chemotherapy resistance, the effectiveness of chemotherapy is often limited, particularly in recurrent and certain primary cases (175,176).
Osteosarcoma mostly occurs in the extremities (177). These tumors are surrounded by ECM, which creates a physical barrier for infiltrating cells (178). However, there is an ongoing direct and indirect crosstalk between osteosarcoma cells and CAFs. The miRNA 1228 has been shown to be upregulated in CAFs from patients with osteosarcoma. It downregulates the protein, suppressor of cancer cell invasion, in osteosarcoma cells, which thereby promotes their proliferation and migration (179). On the other hand, osteosarcoma-derived EVs have been found to drive the differentiation of lung fibroblasts into CAFs, which subsequently exhibit both an increased expression of α-SMA and metalloproteinase MMP2 and MMP9, and an increased production of fibronectin (180). This may serve as a possible mechanism of metastatic niche formation. Moreover, TGF-β1 was also identified as a crucial molecule for fibroblast transdifferentiation (180). Osteosarcoma cells can secrete EVs with collagen type VI alpha 1 (COL6A1), which transforms normal fibroblasts into CAFs that secrete IL-6 and IL-8 (181). In addition, COL6A1 has been shown to induce TFG-β expression in CAFs, which subsequently triggers the invasiveness of osteosarcoma cells (181). The process of cell-projection pumping, the exchange of cellular and cytoplasmic molecules, has been reported to occur between osteosarcoma and human gingival fibroblasts. This interaction surprisingly led to a decreased production of IL-6, granulocyte-colony stimulating factor and granulocyte-macrophage colony-stimulating factor (182).
It is becoming clear that a variety of CAFs with diverse functions and prognostic impact are involved in osteosarcoma. In an in silico study performed by Xu et al (183), six subclusters of fibroblasts were identified. Specifically, TOP2A+ CAFs showed the highest association with carcinogenic pathways (p53 and cell cycle) and crosstalk with osteosarcoma cells. Only these TOP2A+ CAFs were found to serve as a prognostic factor (183). In a different study (184), similarly, high levels of two more CAF subsets that either resembled iCAFs or apCAFs were found to negatively influence survival probability in patients with osteosarcoma.
CAF identity in sarcomas
CAFs and sarcomas share not only the same mesenchymal origin, but also an enormous level of heterogeneity. Therefore, anticipating combinations of the inherent heterogeneity of CAFs, known from carcinomas, with the heterogeneity of sarcomas could potentially constitute countless combinations in which the identity of CAFs, whether pro- or anti-tumorigenic, may be almost impossible to unravel.
CAFs origins in sarcomas
Fibroblasts and sarcomas have a common mesenchymal origin. Putting aside the heterogeneous origins of CAFs that have been described previously, fibroblasts and connective/muscle tissues share developmental lineage. They both arise from the true mesenchyme during embryonic development. Moreover, certain fibroblasts, similarly to other cells of the connective/muscle tissues, have been shown to arise from MSCs in the post-natal period (185,186). These complicated developmental associations are further emphasized in RMS, which can arise in human body parts lacking skeletal muscles. One of the possible hypotheses underlying this phenomenon could be its development from mesenchymal progenitor cells that failed to develop into myocytes (143). Due to these tight developmental links, numerous sarcoma cancer cells share features with fibroblasts/CAFs with respect to the shared gene expression involved in cell adhesion, motility and ECM production. The most closely related sarcoma cells are histological subtypes of sarcomas that directly arise from fibroblasts (either fibrosarcoma or myxofibrosarcoma). At present, no information is available on whether CAFs are present in these tumors, or, if they are present, what the nature of their origin is; namely, neighboring tissue-resident fibroblasts, endothelial to mesenchymal transition, MSCs/tumor-associated MSCs, EMT, transdifferentiated fibroblasts or fibrocytes from the bone marrow (4). The absence of this information is primarily due to the absence of markers that could distinguish CAFs from sarcoma cells. This limitation is probably the main reason contributing to the scarcity of studies on sarcomas and CAFs in comparison with the abundance of studies on CAFs in carcinomas. Therefore, sarcomas and CAFs currently represent largely uncharted territory in terms of both clinical and academic research.
CAFs, sarcoma cells and lineage plasticity in the TME
ECM within the TME is a complex milieu, which is responsible for orchestrating numerous processes, including cell migration and immunomodulation. Although no comparative studies between sarcomas and carcinomas have been published, it could be hypothesized that sarcomas are likely to have a higher accumulation of ECM compared with epithelial cancers due to both their shared developmental processes with fibroblasts and the abundance of fibroblasts in connective tissues. However, certain sarcomas, such as RMS, which is a skeletal muscle-like tumor, may exhibit an abundance of ECM similar to that of epithelial tumors due to the lower presence of fibroblasts. In carcinomas, CAFs are able to drive EMT. However, sarcoma cells already exhibit mesenchymal features, and these may need to be induced in carcinomas to enable metastases. Sannino et al (187) proposed that sarcomas are able to undergo phenotypic changes between EMT and mesenchymal-epithelial transition and can retain an intermediate, metastable phenotype. The varying degrees of epithelial/mesenchymal-like differentiation in different types of sarcomas supports this hypothesis.
Lineage plasticity is increasingly acknowledged as the hallmark of cancer, as it is associated not only with cancer cells, but also with other cells in the TME (188). CAFs have been shown to exert an important role in tumoral plasticity and their mutual interactions with cancer cells and other cells of the TME determine tumor behavior, which is translated into cancer progression, invasiveness and therapy resistance (188). In carcinomas, the plasticity of CAFs and cancer cells has been extensively studied both in vitro and in vivo. Via either basic experimental models exploring the changes elicited after co-culture of fibroblasts/CAFs with cancer cells (189,190) or in vivo models and advanced special-based analytic techniques (43), or via the possible implementation of novel strategies and techniques for the evaluation of lineage plasticity (188), our understanding of CAF plasticity in carcinomas has advanced compared with the past.
The research trajectory of CAFs in sarcomas, however, has been different. Pioneering studies on sarcomas and fibroblasts attempted to delineate the association between sarcoma cells and fibroblasts as long ago as the 1950s (191), with a small amount of further progress being made in the 1970s (192,193). These studies already pointed to the behavioral similarities between sarcoma cells and fibroblasts in terms of their in vitro invasiveness (191,192) or the acquisition of their features (193). Since then, attempts that have been made to distinguish fibroblasts/CAFs from sarcomas either morphologically/phenotypically or functionally (194,195), or to delineate their mutual association (193), have been rather sporadic, falling far short of the research efforts dedicated to CAF research in carcinomas. However, novel strategies and technologies are currently changing the opportunity landscape enabling exploration of CAF heterogeneity within sarcomas to be made. The application of novel strategies in combination with metabolic profiling may lead to the identification of novel CAF populations in sarcomas (196); alternatively, applying single-cell sequencing techniques in combination with spatial profiling may serve to unravel sarcoma cell populations with CAF-like properties (174). These novel strategies, in combination with multidimensional high-definition techniques, may now challenge the previously unexplored identities of CAFs or CAF-like cells in sarcomas.
Unraveling the identity of CAF heterogeneity, however, will bring not only opportunities, but also challenges. At present, a number of CAF subsets with various functions and phenotypes have already been identified. This variability may complicate efforts to target them effectively. Furthermore, a universal therapeutic strategy is unlikely to be uncovered without having a comprehensive understanding of the intricate TME. Recent advancements in technology, such as single-cell sequencing and spatial transcriptomics, may facilitate a more detailed exploration of specific CAF subsets (197). However, truly effective targeting of these subsets will require an integrated, deeper and context-dependent understanding of their roles. This understanding will certainly need a greater stress and reliance on an understanding of the functional diversity of CAFs (198). It may be assumed that the functional diversity of CAFs is probably still largely hidden beneath the universal CAF phenotype; moreover, this as yet still-hidden and unexplored functional diversity, driven by the reprogramming forces from the tumor and tumor-infiltrating cells, will presumably complicate the future development of universal therapeutic strategies targeting CAFs or CAF-like cells in sarcomas.
Regardless of the aforementioned discussion, at present, the exact role and underlying mechanisms of CAF involvement in sarcomas have yet to be fully elucidated, which certainly further contributes to the complexity of CAF functionalities and the ensuing challenges with their identity in sarcomas. Owing to such complexity, research based on cancer cell lines and fibroblast lineages needs to be more frequently corroborated both by in vivo analyses and the investigation of CAFs isolated directly from sarcomas and advanced approaches and techniques should be implemented to enhance our understanding of CAF and/or CAF-like cell functions within these challenging tumors.
Diverse ways of targeting CAFs
Considering the commonly pro-tumorigenic role of CAFs and their presence in both primary tumors and metastatic tissues, directing therapeutic efforts against CAFs should hold significant promise even in the advanced stages of the disease. Nevertheless, addressing the suitability of CAFs for this purpose presents challenges due to their very dynamic and context-dependent nature (199). Targeting CAFs is therefore challenging, as the approaches will differ according to individual patients and their separate diagnoses. Therefore, personalized approaches will need to be taken into consideration. However, despite these hurdles of personalization, their targeting is essential for breaking the tumor resistance to a number of the therapeutic interventions currently in use, including immunotherapy. Blocking molecules that are known to induce CAF differentiation and activation, or blocking or neutralizing CAF-derived biologically active molecules that exert an impact on both cancer and immune cells, may offer an easier means of challenging CAF-driven therapy resistance and tumor progression (Fig. 3).
CAF depletion
One particular therapeutic avenue that has come under consideration is to focus on depleting the immunosuppressive FAP+ CAF population. In order to target FAP+ CAFs, a study by Kakarla et al (12) used chimeric antigen receptor T cells (CAR-T), which target FAP. Another study by Wang et al (200) also reported that CAR-T cells were not only able to inhibit tumor growth, but they could also cause an increase in the responsiveness of CD8+ T cells to tumors. A recent study also demonstrated that these CAR-T cells could improve the anti-tumor efficacy of claudin18.2-targeting CAR-T cells via suppressing the recruitment of MDSCs in pancreatic cancer, suggesting that targeting CAFs may be beneficial in combined therapies (201). The strength of the depletion strategy stems from a considerable (antigen-mediated) specificity of the CAF targeting. However, the weakness of this strategy is that CAFs are not a uniform population, consisting of populations with occasionally opposing roles in the tumor microenvironment. Depleting all CAFs may thus inadvertently remove subtypes that have anti-tumorigenic activities, or removal of CAFs with pro-tumorigenic activities may induce compensatory mechanisms. Therefore, depleting CAFs can also lead to unintended consequences and possible weaknesses, as was shown in a preclinical animal study by Ozdemir et al (38). In the authors study involving a transgenic mouse model of PDAC, the removal of α-SMA+ myCAFs led to increased invasiveness of the cells and enhanced EMT. Additionally, this depletion of α-SMA+ myCAFs resulted in decreased survival rates among the experimental animals, and was associated with a suppression of anti-tumor immunity (38). These contrasting findings therefore raise strong awareness of the importance of not employing CAF depletion approaches without a full understanding of how CAF biology operates under specific disease state conditions.
EVs
The therapeutic impact can be attained not only through the direct targeting of CAFs, but also through targeting their products, including EVs. The EVs produced by CAFs are able to support cancer progression, create a pre-metastatic niche for certain cancer diagnoses, and facilitate resistance to therapy. In a study by Gao et al (202), CD63+ CAFs were found to drive the resistance to tamoxifen in breast cancer via the secretion of miR-22-rich EVs. This study further demonstrated that this resistance could be overcome by the simultaneous neutralization of CD63 and miR-22 to enhance the therapeutic effect of tamoxifen (202). Another study (67) showed that CAF-released EVs could mediate CAF-promoted survival of cancer cells as the response to chemotherapy, and inhibiting the release of EVs was shown to overcome the chemotherapy-induced resistance. However, EVs have also been shown to serve dual roles as both targets and tools in cancer therapy (38). Zhou et al (203) recently demonstrated that EVs have significant potential to reprogram CAFs, thereby enhancing therapeutic responses in pancreatic cancer. In their study, these researchers loaded EVs with miR-138-5p and the anti-fibrotic agent pirfenidone. They found that miR-138-5p could inhibit both TGF-β signaling and proline-mediated collagen synthesis. This effect was synergistically enhanced by addition of the anti-fibrotic agent pirfenidone, resulting in the reprogramming of CAFs and the subsequent suppression of their pro-tumorigenic effects. Therefore, targeting EVs represents a promising strategy for elucidating the underlying key pro-tumorigenic mechanisms associated with CAFs. The potential strength of EV targeting may stem from its enhanced specificity. This is because pro-tumorigenic EVs are selectively affected, while the functions of anti-tumorigenic CAFs and their released EVs may remain intact. However, similar to CAF depletion strategies, there are still significant challenges. The inherent heterogeneity of EVs, varying in size, composition, and origin, along with a lack of clear and EV-specific molecular targets that could identify the pro-tumorigenic nature of these EVs, still needs to be addressed.
TGF-β and Wnt signaling
Given the important role of TGF-β in CAF biology, and how CAFs affect ECM deposition, targeting TGF-β signaling has also been proposed as a potential strategy for cancer treatment. However, immune dysregulation, or employing an incorrect dosing of anti-TGF-β agents, may cause potential systemic cytotoxicity (204). Nevertheless, the TGF-β signaling pathway has been shown to be regulated by Wnt signaling (205,206). There is growing evidence to suggest that Wnt signaling in CAFs fulfills a significant role in tumor development. For example, cancer cells located in regions enriched with CAFs were shown to have upregulated Wnt signaling (207) and autocrine Wnt signaling in CAFs was shown to promote cancer cell migration and invasiveness and EMT in breast cancer (208). Another study by Avgustinova et al (209) also demonstrated that non-canonical Wnt7a, which is exclusively secreted by aggressive breast cancer cells, could recruit and activate fibroblasts via a TGF-β-dependent process. Wnt signaling in CAFs has also been shown to be involved in inducing tumor resistance to various types of therapies, including chemotherapy. For example, increased expression of Wnt16B by CAFs was found to attenuate the cytotoxic effects of the chemotherapeutic drugs mitoxantrone and docetaxel in patients with prostate cancer (210). Wnt signaling in CAFs has also been demonstrated to modulate the immune components of the tumor microenvironment; for example, autocrine Wnt2 signaling in fibroblasts was shown to promote: i) Colorectal cancer progression (211); ii) the recruitment and polarization of immune cells, such as tumor-associated macrophages or Tregs [as described in (212)]; and iii) Wnt2 signaling may also inhibit in vitro differentiation and stimulatory activity of dendritic cells (213). Therefore, targeting Wnt2 signaling in CAFs may afford a promising therapeutic strategy, and this has already been corroborated by a study in which targeting Wnt2-producing CAFs was shown to restore dendritic cell-mediated immunity against tumors (213).
IL-2, IL-1 and NLRP signaling
Other potential CAF-targeting therapies may include the stimulation of IL-2 pathways (214,215) or inhibition of IL-1 signaling through IL-1 receptor agonists (28). It has also been demonstrated that TGF-β-activated CAFs contribute to tumor resistance in breast cancer therapy, especially in the case of treatment with trastuzumab, which is often associated with diminished IL-2 activity. This suggests that stimulation of the IL-2 pathway in the stroma may restore the efficacy of trastuzumab (216). To inhibit the IL-1 signaling pathway, the inhibition of NLRP3 in CAFs has been proposed to offer a promising therapeutic strategy. This proposal was based on the finding that activation of the NLRP3/IL-1β signaling axis in CAFs has been associated with both the inflammation-mediated immunosuppressive milieu and the promotion of tumor progression (51,217).
Targeting cell signaling could be a promising approach for future anti-cancer therapies, as new and often highly specific inhibitors of these signaling pathways are being developed. However, a significant challenge with targeting the signaling pathways involved with CAFs is that these targets are essential for maintaining homeostasis. As a result, these inhibitors need to be effective only within the TME. Their full therapeutic potential is likely to be unleashed when combined with effective drug delivery strategies, such as antibody-drug conjugates or nanocarrier-based systems (218).
Targeting CAFs in sarcomas
Currently, various therapeutic approaches are being tested to restrict CAFs within the TME of sarcomas. Similarly to the treatment of other cancers, the focus is mostly on disrupting the CAF-mediated mechanisms of tumor resistance to immunotherapy and/or other therapeutic modalities (Fig. 3). In addition, however, if the same mesenchymal origin and gene expression overlaps with sarcomas, this may allow for targeting CAFs and sarcomas simultaneously via shared targets, which could involve either surface molecules or the associated signaling pathways. For example, the recently established highly-transformed cell line of UPC (JBT19) was shown to express numerous fibroblast/CAFs markers, including the expression of collagen, FAP, nestin, vimentin and a-SMA (32). FAP, a classical marker of CAFs within the epithelial tumors, has been shown to be expressed in a number of histological subtypes of sarcoma cells. Indeed, Crane et al (219) showed that the majority of sarcomas express FAP. Similarly, α-SMA and PDGFR are expressed by sarcomas and CAFs (220-223). Therefore, targeting these molecules allows the targeting of both cell types at the same time, which is not usually possible within carcinomas, and may present a considerable benefit in the treatment of sarcomas. The most advanced drugs targeting PDGFR are imatinib mesylate (Gleevec) and sunitinib malate (Sutent), which are tyrosine-kinase inhibitors of the PDGFR-mediated signaling pathway, and were FDA-approved for the treatment of gastrointestinal stromal tumors (224). Another drug targeting PDGFR is olaratumab, which is an anti-PDGFR monoclonal antibody. A combined treatment of olaratumab with doxorubicin in the treatment of advanced sarcomas has initially shown promising results in a Phase 2 clinical trial (225). However, no improvement in survival outcomes was later observed in the Phase 3 clinical trial (226). Regardless of these findings, novel combinatorial approaches targeting PDGFR are still being tested, including immunotherapy; for example, combinations of anti-PD-1 monoclonal antibody nivolumab with sunitinib or CAR-T cells targeting PDGFR/FAP (227-229).
However, targeted therapies aimed at both fibroblasts and cancer cells come with their own set of challenges and possible weaknesses. Although the goal of oncological treatments is to eliminate all cancer cells, that is, the cause of the disease, indiscriminately targeting CAFs without considering their specific subsets can lead to unpredictable therapeutic outcomes. This unpredictability has been observed in studies that have employed CAF-targeting strategies (12,38,200,201). These findings suggest that CAFs have a dual role with respect to tumors, acting either as promoters or inhibitors of tumorigenesis. Deciphering the CAF identity and stratifying this identity according to phenotypical or functional features into subsets with either pro- or anti-tumorigenic roles, is the approach through which CAF targeting strategies should be developed. This stratification may even stand on minute differences, assigning pro-tumorigenic or anti-tumorigenic roles based on even one phenotypic or functional marker. This marker may then be targeted through, for example, new generations of CAR-T cells, the activation of which requires simultaneous recognition of two or more targets (230,231) and their activity enhanced by metabolic reprogramming (232). For this task, CAR T-cells appear to be the most suitable option. Their precise targeting of CAFs and their metabolic reprogramming (enhancement) can not only enable them to effectively and selectively confront the supportive TME but also minimize the risk of off-tumor toxicity, ultimately enhancing overall therapeutic safety. Alternatively, targeting of the CAF subset may also be attained indirectly through neutralizing the CAF-released products, such as EVs and cytokines (202,204). These approaches might challenge the pro-tumorigenic, while preserving the anti-tumorigenic, activities of the CAFs that are elicited by different subsets within the TME. To implement this approach, however, a much more penetrating characterization of CAF heterogeneity and its functional roles would necessarily be required for efficient CAF targeting via different therapeutic interventions.
Immunotherapy holds promise for numerous types of cancers. One of the immunotherapeutic approaches in sarcomas that has been developed is based on the adoptive transfer of membrane-anchored and cell-surface vimentin-targeting IL-12-armed T cells that have been shown to abolish CAF functions in heterogenous osteosarcoma xenograft models (178). The transferred T cells were able to effectively decrease the levels of TGF-β and increases the levels of IFN-γ, which led to CAF deprivation through Fas-mediated apoptosis (178). One potential target for T cell-based immunotherapy, or targeted therapy of STSs, may be leucine-rich repeat containing 15 (233,234); another alternative strategy could be the targeting of FAP, which is expressed in sarcomas and CAFs (219,235). Specific inhibitors or peptides may be utilized to target FAP in sarcomas, also for diagnostic and/or theranostic purposes (236,237). A significant advantage of sarcomas in the field of immunotherapy is that, unlike epithelial tumors, they are often diagnosed as localized tumors and can usually be surgically resected. The resection of these typically large tumors not only provides invaluable material for in-depth analyses, including investigations into the tumor immune microenvironment, but also aids in the prediction and detection of neoantigens (238). This information can help tailor immunotherapeutic approaches. Additionally, these tumors can serve as a vital source of tumor-infiltrating lymphocytes, which are crucial for adoptive cellular immunotherapy (239). A novel strategy to effectively challenge CAFs in sarcomas may also arise in due course through metabolic targeting. A recent study was able not only to identify a new subset of so-called 'glycolytic' CAFs (glyCAFs), which impede T-cell infiltration of tumors, but also to demonstrate that, after targeting glycolysis, the impeded T-cell infiltration was reduced, and the efficacy of the chemotherapy was thereby augmented (196). Moreover, nanocarrier-assisted targeting of the TME (218) may further enhance the specificity and efficacy of the metabolic reprogramming (240). Taken together, these findings thus show new possibilities for targeting CAFs in sarcomas, especially in combination with other therapeutic modalities. However, novel combinations should be approached with caution, as there may be several limitations and weaknesses. For example, combining CAF-targeting CAR-T cells with the metabolic reprogramming of GlyCAFs could be advantageous, as it is expected to increase CAR-T cell infiltration and enhance their therapeutic efficacy. However, if the reprogramming is achieved through the GLUT1 glucose transporter (196), it may also have negative effects on the infiltrating CAR-T cells. GLUT1 is essential for glucose uptake in activated immune cells, and its inhibition in the TME-infiltrated immune cells could inadvertently lead to immunosuppression (241,242).
New avenues for the identification of novel targets in CAFs
Finding new avenues to specifically challenge CAFs for therapeutic purposes requires an understanding both of the intimate crosstalk between tumor cells and CAFs, and how this crosstalk evolves during the course of tumor development. For this purpose, multiple in vitro and in vivo models have been established (Fig. 4).
In vitro models
Although conventional 2D monolayer cell culture is an easy and often used system with high cell viability, it is not as effective as 3D models are in terms of replicating the complexity of the TME. CAFs cultured under 3D conditions secrete relatively more paracrine signaling molecules, and promote the invasive behavior of breast cancer cells compared with 2D conditions (243). Additionally, when CAFs are grown in a 3D model using a mixture of Matrigel and collagen I, the expression of important proteins for cell-ECM contacts, such as tenascin, are upregulated. This was found not to be the case for 2D cultures (244).
Various methods have been employed to enhance 3D tumor spheroid models in order to replicate more closely the complexity of the TME. One way to study the interactions between tumor cells and CAFs is through using a multicellular (heterotypic) spheroid system, where CAFs are co-cultured with tumor cells (245). This system enables an analysis of ECM production and remodeling by CAFs to be made; however, it does not incorporate an important component of the tumor stroma, which is the vasculature. Tumoroids, which consist of tumor cells embedded in a stromal environment comprising monomeric type I rat-tail collagen, patient-specific CAFs and ECM components such as laminin and human umbilical vein endothelial cells, can be utilized to study the impact of CAFs on the vasculature. Additionally, 3D-printed scaffolds may be employed to mimic the intricate architecture of the TME more closely when investigating CAF-cancer cell interactions. Synthetic materials, such as poly(ε-caprolactone) (PCL) (246) and natural biomaterials, such as sodium alginate or gelatin (247), have both been demonstrated to offer multifaceted support for cells and to facilitate direct cell interactions, consequently improving the reproducibility of the 3D tumor models.
To create a realistic 3D model, it is important to consider immune cells. Cytokines and chemokines produced by CAFs are mainly analyzed by ELISA, or via various multiplex cytokine analyses from co-culture supernatants (248) or conditioned media (249). To obviate the need for complex analyses of individual populations in co-culture models, Balachander et al (250) performed a study using only CAFs cultured on engineered fibrous matrices of PCL scaffolds. They demonstrated proliferation of the CAFs, with ECM remodeling, induced stemness and increased invasiveness of the cancer cells, and the CAFs retained a pro-inflammatory phenotype independently of the cancer cells.
In vivo models
The most commonly used in vivo models for studying CAFs are mouse and zebrafish xenografts with an injected mix of tumor cells and CAFs (53,251-253). Additionally, Cre-driver transgenic mice are often used (254-256). The co-injection models have the disadvantage that the injected CAFs become mixed up with the host-derived CAFs as the tumor grows. However, even transgenic models using Cre drivers are not flawless models, as a CAF-specific Cre driver line does not currently exist (8). Therefore, further research is required to develop improved in vivo models that are suited to the study of CAFs.
CAF models in sarcomas
For an improved understanding of the association between sarcomas and CAFs, researchers often utilize the aforementioned strategies. In the case of RMS, both 3D spheroids and xenograft mice have been employed (5). Similarly, in a study by De Vita et al (257), both 3D cultures with a collagen scaffold and a xenotransplantation model were used to study CAFs in UPS, LPS and LMS. Comparatively speaking, the EWS model is quite complex, and requires the inclusion of bone architecture. To create this model, in a study by Molina et al (258), human MSCs were seeded on to and cultured on PCL scaffolds for 12 days in an osteogenic medium to enable their differentiation into osteoblastic cells. Subsequently, the constructs were decellularized and used to inoculate EWS cells (258).
Developing technologies, such as tumor-on-chip models, could further improve the models for CAF/sarcoma studies (259). However, due to the shared mesenchymal origins of CAFs and sarcoma cells, their heterogeneity and significant lineage plasticity, techniques and models that allow for lineage tracing and monitoring should also be incorporated into contemporary models (260,261). This incorporation would help towards an improved understanding of the phenotypical/functional heterogeneities and dynamics of CAFs/CAF-like cells and sarcoma cells in the TME.
Future prospects
Over the last decade, the landscape of clinical oncology has witnessed a significant transformation, shifting its focus away from cancer cells to other cells of the TME. The most significant shift in interest has apparently been towards immune cells, around which novel immunotherapeutic approaches are currently being established. However, after the initial enthusiasm triggered by the explosion of the ICI therapies and the rise of cell-based immunotherapies, some of this enthusiasm is waning as clinical trial results, up to this point, have not matched with the initial expectations. This is particularly true for solid tumors, where the TME is set up to substantially drive tumor resistance to the immunotherapies and other oncological treatments, such as cytotoxic chemotherapy or radiotherapy. Therefore, the current focus in clinical oncology is now on cells that foster tumor resistance and among them, CAFs have become central to this focus.
Although the enormous diversity and plasticity of CAFs may deter researchers from attempting to find new CAF vulnerabilities to therapeutically target, the novel cancer cell-targeting therapeutic approaches that have been developed in recent years may bring new momentum to the research. This momentum could stem from the different goals that cancer cell and CAF targeting should reach. Whereas the goal of cancer cell targeting therapies has been to eliminate cancer cells efficiently, the goal of CAF targeting therapies does not necessarily need to work towards the elimination of CAFs, but rather to efficiently downgrade their cancer cell fostering and protecting activities in order to make the cancer cells more vulnerable to the cancer cell targeting therapies (41). Therefore, the cancer cell targeting therapies that, in spite of having been shown to deliver substantial therapeutic effects, still failed in terms of the efficient elimination of cancer cells, may, once refocused on CAFs, prove to be efficient in breaking down the cancer cell resistance. Among these therapies could be cell-based (12), metabolic pathway-targeting (196) or cell signaling-targeting options (262). However, implementing these therapies will undoubtedly require gaining much broader and more detailed insights into the underlying mechanisms via which CAFs make cancer cells resilient to oncological treatments.
CAFs in sarcomas have long been far removed from the limelight as far as interest in them was concerned, primarily due to even thinner differences between sarcoma cancer cells and CAFs. However, despite these thin differences, a recent study by Broz et al (196) was able to identify novel vulnerabilities of CAF-mediated tumor resistance to therapy. Using a broad and deep characterization of the TME of STS, these researchers were able to characterize a novel subset of CAFs in a murine model that they termed 'glyCAFs'. These glyCAFs were found to rely on glucose transporter 1-dependent expression of CXCL16, and were able to impede cytotoxic T-cell infiltration into the tumor parenchyma. This study also revealed that targeting glycolysis not only was sufficient to enhance T-cell infiltration of the tumors, but it also augmented the efficacy of chemotherapy. These results therefore not only stressed the importance of combinatorial therapeutic interventions in the treatment of sarcoma, including treatments targeting CAFs, but also highlighted that mere metabolic reprogramming of CAFs without their elimination may be sufficient to decrease the resistance of cancer cells. This study therefore demonstrated that the objectives of targeting CAFs should be different from those of targeting cancer cells and that the prospects for CAF studies will more likely depend on the extensive characterization of CAF populations under specific disease conditions, upon which their new vulnerabilities will be revealed and implemented into combined therapies, with the aim of breaking down cancer cell therapeutic resistance. In addition, the extensive characterization of CAF populations under specific disease conditions may reveal targets that could give rise to the contrary; namely, an undesirable enhancement of the CAFs pro-tumorigenic activities. Upon disclosure of these targets, they should then be avoided, not only based on the CAF targeting, but also upon the sarcoma cell targeting.
As has been demonstrated in numerous cancer types, including breast and colorectal cancers, a low proportion of cancer cells relative to stromal components has been associated with poorer overall and disease-free survival outcomes (263,264). Therefore, CAFs hold significant potential in terms of diagnostics and the classification of tumor prognosis. Examining CAFs together with tumor cells after a biopsy or surgical resection may help to improve our understanding of stromal activation within the tumor (265). Another aspect highlights that it is the treatment of solid tumors, particularly through cell-based therapies, that faces distinct challenges, in part due to biological barriers that hinder immune cell infiltration into the tumor tissues. The identification of CAFs according to their specific surface markers and spatial distribution within the TME may therefore serve as valuable diagnostic tools. In a study by De Vlieghere et al (265), it was noted that most investigations have tended to focus either on quantifying the stromal compartment (commonly using broad activation markers such as α-SMA) or on assessing stromal quality through markers of CAF-secreted factors, such as neuregulin. However, these approaches are rarely integrated. For a more comprehensive prognostic assessment of the tumor stroma, these authors suggest that a stepwise method could be advantageous. Initially, hematoxylin-and-eosin staining could be employed to categorize tumors based on stromal content, using a standardized cut-off point (for example, 50%) to distinguish between stroma-rich and stroma-poor tumors. Subsequently, a general CAF marker could be used to evaluate stromal cellularity, followed by the application of markers targeting CAF-secreted proteins to further characterize the activation state of the fibroblastic component. Such integrated tumor characterization could be useful both in terms of stratifying tumor subtypes and in terms of guiding personalized treatment strategies. Moreover, given their established role in mediating resistance to chemotherapy and immunotherapy, the presence and phenotype of CAFs may be of use in terms of selecting patients for clinical trials and in terms of predicting poor responders to conventional therapies.
It is unfortunate that CAF heterogeneity across different tumors continues to present a formidable challenge for diagnosis (266). However, there is potential for new analytical tools to be developed using artificial intelligence (AI) to improve CAF-associated tumor diagnostics and treatment responses. AI-based methods offer the opportunity to process complex diagnostic data, which may be beneficial for dealing with the diagnostic challenges posed by the heterogeneity of CAFs and sarcomas (267-269).
AI holds great potential to shape the future of medicine, and there is a growing interest in applying AI tools to analyze CAFs in various cancer diagnoses. The deep learning method, not only in histological image analyses but also in single-cell RNA sequencing and spatial transcriptomics, may help with the classification of CAF subpopulations and analysis of their gene expression profiles for diagnostic purposes. Moreover, spatial transcriptomic data provide valuable insights into how CAFs interact with tumor and immune cells in sarcoma tissues. Given both the complexity and scale of these datasets, deep learning can facilitate their analysis, increasing the accessibility of CAF-associated diagnostics in clinical practice. However, to develop robust AI tools for sarcomas, further research is required to enhance their accuracy and clinical applicability. Nevertheless, several studies have already highlighted the significant potential of AI in cancer diagnosis, especially with regard to uncovering the roles of CAFs (270-272), and it is therefore likely that this will become the avenue through which the challenging CAF ecosystem in various sarcomas may be unraveled for the implementation of improved personalized treatments and their combinations.
Furthermore, CAFs can also be obtained from the bloodstream (273,274). A study by Ao et al (273) found that circulating CAFs were present in the majority of patients with metastatic breast cancer, whereas no circulating CAFs were detected in healthy donors. This suggested that circulating CAFs may be a promising diagnostic marker. Similar findings were observed in an in vivo animal model of breast cancer (275) and in other diagnoses, such as prostate cancer (274). When approved for other cancers, including sarcomas, the detection of circulating CAFs in peripheral blood may be implemented in diagnostics in the future.
To date, the heterogeneity of CAFs across various tumor types continues to pose a significant challenge for accurate diagnosis (266). Future studies should emphasize the application of advanced methodologies, such as comprehensive single-cell profiling and spatial transcriptomics, to elucidate the complexity of CAF populations and to characterize functionally distinct subtypes. Existing therapeutic strategies aimed at CAF depletion or inhibition have demonstrated limited efficacy and, in certain cases, even detrimental outcomes, largely due to the context-dependent tumor-promoting or tumor-suppressing roles of specific CAF subsets. Therefore, future efforts should focus on developing selective and context-specific interventions, including the phenotypic reprogramming of CAFs, as well as targeting CAF population-derived signaling pathways.
In addition, emerging analytical tools leveraging AI hold a promise for advancing CAF-associated tumor diagnostics and improving predictions of treatment response. AI-based approaches have been shown to facilitate the interpretation of complex, high-dimensional datasets, offering potential solutions to the diagnostic challenges posed by the heterogeneity of CAFs and the broader sarcoma landscape (267-269).
Conclusion
In conclusion, CAFs in sarcomas pose a significant challenge. This challenge arises not only from their dual role in tumors (pro- or anti-tumorigenic activities), as observed in carcinomas, but also from their similarities to sarcoma cells. As a result, our understanding of CAFs in sarcomas may still lag far behind our understanding of CAFs in carcinomas. However, looking at this challenge from a different vantage point, the resemblance between CAFs and sarcoma cells yet presents a new opportunity. The similarities suggest that insights gained from studying sarcomas/sarcoma cells, including their plasticity, communication with the immune system, impact on the TME and vulnerabilities or resistance mechanisms, might also apply to understanding CAFs. In today's era of AI, a deeper understanding of CAFs in sarcomas could enable the use of diagnostic, prognostic and predictive AI tools that are currently being designed for cancers for an improved understanding of the role of CAFs. Therefore, it is hoped that advancing our knowledge of sarcomas and their treatment could contribute to an improved understanding of CAFs, whether in sarcomas or carcinomas.
Availability of data and materials
Not applicable.
Authors' contributions
IB, KKa, ZOS and DSm conceived and designed the present study. YSK, PT, DSt, KKr, JS, AO and JB contributed to the writing of the manuscript. IB and KKa share first authorship. ZOS and DSm share senior authorship. All authors read and approved the final manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
JB is a part-time employee and a minority shareholder of Sotio, a.s., a biotech company that is developing novel immunotherapies. The other authors declare that they have no competing interests.
Acknowledgements
Not applicable.
Funding
The research was supported by funding from the Charles University (project GA UK No. 94323); the Ministry of Health, Czech Republic (projects AZV NU23J-08-00031, NU22-03-00300 and NU23-08-00071); and the Institutional grant no. RVO 61388971 of the Institute of Microbiology AS CR, v. v. i.
References
Riley RS, June CH, Langer R and Mitchell MJ: Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov. 18:175–196. 2019. View Article : Google Scholar : PubMed/NCBI | |
Mondal M, Guo J, He P and Zhou D: Recent advances of oncolytic virus in cancer therapy. Hum Vaccin Immunother. 16:2389–2402. 2020. View Article : Google Scholar : PubMed/NCBI | |
Soerjomataram I and Bray F: Planning for tomorrow: Global cancer incidence and the role of prevention 2020-2070. Nat Rev Clin Oncol. 18:663–672. 2021. View Article : Google Scholar : PubMed/NCBI | |
Chen X and Song E: Turning foes to friends: Targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 18:99–115. 2019. View Article : Google Scholar | |
D'Agostino S, Tombolan L, Saggioro M, Frasson C, Rampazzo E, Pellegrini S, Favaretto F, Biz C, Ruggieri P, Gamba P, et al: Rhabdomyosarcoma cells produce their own extracellular matrix with minimal involvement of Cancer-associated fibroblasts: A preliminary study. Front Oncol. 10:6009802020. View Article : Google Scholar | |
Sarkar M, Nguyen T, Gundre E, Ogunlusi O, El-Sobky M, Giri B and Sarkar TR: Cancer-associated fibroblasts: The chief architect in the tumor microenvironment. Front Cell Dev Biol. 11:10890682023. View Article : Google Scholar : PubMed/NCBI | |
Pillozzi S, Bernini A, Palchetti I, Crociani O, Antonuzzo L, Campanacci D and Scoccianti G: Soft tissue sarcoma: An insight on biomarkers at molecular, metabolic and cellular level. Cancers. 13:30442021. View Article : Google Scholar : PubMed/NCBI | |
Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, Fearon D, Greten FR, Hingorani SR, Hunter T, et al: A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 20:174–186. 2020. View Article : Google Scholar : PubMed/NCBI | |
Maia A, Schollhorn A, Schuhmacher J and Gouttefangeas C: CAF-immune cell crosstalk and its impact in immunotherapy. Semin Immunopathol. 45:203–214. 2023. View Article : Google Scholar : | |
Kennel KB, Bozlar M, De Valk AF and Greten FR: Cancer-associated fibroblasts in inflammation and antitumor immunity. Clin Cancer Res. 29:1009–1016. 2023. View Article : Google Scholar : | |
Huang H, Wang Z, Zhang Y, Pradhan RN, Ganguly D, Chandra R, Murimwa G, Wright S, Gu X, Maddipati R, et al: Mesothelial cell-derived antigen-presenting cancer-associated fibroblasts induce expansion of regulatory T cells in pancreatic cancer. Cancer Cell. 40:656–673.e7. 2022. View Article : Google Scholar : | |
Kakarla S, Chow KK, Mata M, Shaffer DR, Song XT, Wu MF, Liu H, Wang LL, Rowley DR, Pfizenmaier K and Gottschalk S: Antitumor effects of chimeric receptor engineered human T cells directed to tumor stroma. Mol Ther. 21:1611–1620. 2013. View Article : Google Scholar : PubMed/NCBI | |
Chen B, Chan WN, Xie F, Mui CW, Liu X, Cheung AHK, Lung RWM, Chow C, Zhang Z, Fang C, et al: The molecular classification of cancer-associated fibroblasts on a pan-cancer single-cell transcriptional atlas. Clin Transl Med. 13:e15162023. View Article : Google Scholar : PubMed/NCBI | |
Ohlund D, Elyada E and Tuveson D: Fibroblast heterogeneity in the cancer wound. J Exp Med. 211:1503–1523. 2014. View Article : Google Scholar : PubMed/NCBI | |
Tsoumakidou M: The advent of immune stimulating CAFs in cancer. Nat Rev Cancer. 23:258–269. 2023. View Article : Google Scholar : PubMed/NCBI | |
Choi KJ, Nam JK, Kim JH, Choi SH and Lee YJ: Endothelial-to-mesenchymal transition in anticancer therapy and normal tissue damage. Exp Mol Med. 52:781–792. 2020. View Article : Google Scholar : PubMed/NCBI | |
Lamouille S, Xu J and Derynck R: Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 15:178–196. 2014. View Article : Google Scholar : PubMed/NCBI | |
Taguchi A, Kawana K, Tomio K, Yamashita A, Isobe Y, Nagasaka K, Koga K, Inoue T, Nishida H, Kojima S, et al: Matrix metalloproteinase (MMP)-9 in cancer-associated fibroblasts (CAFs) is suppressed by omega-3 polyunsaturated fatty acids in vitro and in vivo. PLoS One. 9:e896052014. View Article : Google Scholar : PubMed/NCBI | |
Cavazzoni A and Digiacomo G: Role of cytokines and other soluble factors in tumor development: Rationale for new therapeutic strategies. Cells. 12:25322023. View Article : Google Scholar : PubMed/NCBI | |
O'Connell JT, Sugimoto H, Cooke VG, MacDonald BA, Mehta AI, LeBleu VS, Dewar R, Rocha RM, Brentani RR, Resnick MB, et al: VEGF-A and Tenascin-C produced by S100A4+ stromal cells are important for metastatic colonization. Proc Natl Acad Sci USA. 108:16002–16007. 2011. View Article : Google Scholar : PubMed/NCBI | |
Liu T, Han C, Wang S, Fang P, Ma Z, Xu L and Yin R: Cancer-associated fibroblasts: An emerging target of Anti-cancer immunotherapy. J Hematol Oncol. 12:862019. View Article : Google Scholar : PubMed/NCBI | |
Robertson-Tessi M, Gillies RJ, Gatenby RA and Anderson AR: Impact of metabolic heterogeneity on tumor growth, invasion, and treatment outcomes. Cancer Res. 75:1567–1579. 2015. View Article : Google Scholar : PubMed/NCBI | |
Wu D, Zhuo L and Wang X: Metabolic reprogramming of carcinoma-associated fibroblasts and its impact on metabolic heterogeneity of tumors. Semin Cell Dev Biol. 64:125–131. 2017. View Article : Google Scholar | |
Martinez-Outschoorn UE, Lisanti MP and Sotgia F: Catabolic cancer-associated fibroblasts transfer energy and biomass to anabolic cancer cells, fueling tumor growth. Semin Cancer Biol. 25:47–60. 2014. View Article : Google Scholar : PubMed/NCBI | |
Tavares-Valente D, Baltazar F, Moreira R and Queiros O: Cancer cell bioenergetics and pH regulation influence breast cancer cell resistance to paclitaxel and doxorubicin. J Bioenerg Biomembr. 45:467–475. 2013. View Article : Google Scholar : PubMed/NCBI | |
Guo Z, Zhang H, Fu Y, Kuang J, Zhao B, Zhang L, Lin J, Lin S, Wu D and Xie G: Cancer-associated fibroblasts induce growth and radioresistance of breast cancer cells through paracrine IL-6. Cell Death Discov. 9:62023. View Article : Google Scholar : PubMed/NCBI | |
Yoshida GJ: Regulation of heterogeneous cancer-associated fibroblasts: The molecular pathology of activated signaling pathways. J Exp Clin Cancer Res. 39:1122020. View Article : Google Scholar : PubMed/NCBI | |
Biffi G, Oni TE, Spielman B, Hao Y, Elyada E, Park Y, Preall J and Tuveson D: IL1-Induced JAK/STAT signaling is antagonized by TGFβ to Shape CAF heterogeneity in pancreatic ductal adenocarcinomacinoma. Cancer Discov. 9:282–301. 2019. View Article : Google Scholar | |
Rimal R, Desai P, Daware R, Hosseinnejad A, Prakash J, Lammers T and Singh S: Cancer-associated fibroblasts: Origin, function, imaging, and therapeutic targeting. Adv Drug Deliv Rev. 189:1145042022. View Article : Google Scholar : PubMed/NCBI | |
Strizova Z, Bartunkova J and Smrz D: The challenges of adoptive cell transfer in the treatment of human renal cell carcinoma. Cancer Immunol Immunother. 68:1831–1838. 2019. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Yang S, Tavormina J, Tampe D, Zeisberg M, Wang H, Mahadevan KK, Wu CJ, Sugimoto H, Chang CC, et al: Oncogenic collagen I homotrimers from cancer cells bind to α3β1 integrin and impact tumor microbiome and immunity to promote pancreatic cancer. Cancer Cell. 40:818–834.e9. 2022. View Article : Google Scholar | |
Taborska P, Lukac P, Stakheev D, Rajsiglova L, Kalkusova K, Strnadova K, Lacina L, Dvorankova B, Novotny J, Kolar M, et al: Novel PD-L1- and collagen-expressing patient-derived cell line of undifferentiated pleomorphic sarcoma (JBT19) as a model for cancer immunotherapy. Sci Rep. 13:190792023. View Article : Google Scholar : PubMed/NCBI | |
Sun X, Wu B, Chiang HC, Deng H, Zhang X, Xiong W, Liu J, Rozeboom AM, Harris BT, Blommaert E, et al: Tumour DDR1 promotes collagen fibre alignment to instigate immune exclusion. Nature. 599:673–678. 2021. View Article : Google Scholar : PubMed/NCBI | |
Meyaard L: The inhibitory collagen receptor LAIR-1 (CD305). J Leukoc Biol. 83:799–803. 2008. View Article : Google Scholar | |
Horn LA, Chariou PL, Gameiro SR, Qin H, Iida M, Fousek K, Meyer TJ, Cam M, Flies D, Langermann S, et al: Remodeling the tumor microenvironment via blockade of LAIR-1 and TGF-β signaling enables PD-L1-mediated tumor eradication. J Clin Invest. 132:e1551482022. View Article : Google Scholar | |
Cheng Y, Li H, Deng Y, Tai Y, Zeng K, Zhang Y, Liu W, Zhang Q and Yang Y: Cancer-associated fibroblasts induce PDL1+ neutrophils through the IL6-STAT3 pathway that foster immune suppression in hepatocellular carcinoma. Cell Death Dis. 9:4222018. View Article : Google Scholar : PubMed/NCBI | |
Cao H, Cheng HS, Wang JK, Tan NS and Tay CY: A 3D physio-mimetic interpenetrating network-based platform to decode the pro and anti-tumorigenic properties of cancer-associated fibroblasts. Acta Biomater. 132:448–460. 2021. View Article : Google Scholar : PubMed/NCBI | |
Ozdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, Laklai H, Sugimoto H, Kahlert C, Novitskiy SV, et al: Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 25:719–734. 2014. View Article : Google Scholar : PubMed/NCBI | |
Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, Dekleva EN, Saunders T, Becerra CP, Tattersall IW, et al: Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 25:735–747. 2014. View Article : Google Scholar : PubMed/NCBI | |
Alkasalias T, Flaberg E, Kashuba V, Alexeyenko A, Pavlova T, Savchenko A, Szekely L, Klein G and Guven H: Inhibition of tumor cell proliferation and motility by fibroblasts is both contact and soluble factor dependent. Proc Natl Acad Sci USA. 111:17188–17193. 2014. View Article : Google Scholar : PubMed/NCBI | |
Gorchs L, Ahmed S, Mayer C, Knauf A, Fernández Moro C, Svensson M, Heuchel R, Rangelova E, Bergman P and Kaipe H: The vitamin D analogue calcipotriol promotes an anti-tumorigenic phenotype of human pancreatic CAFs but reduces T cell mediated immunity. Sci Rep. 10:174442020. View Article : Google Scholar : PubMed/NCBI | |
Mao X, Xu J, Wang W, Liang C, Hua J, Liu J, Zhang B, Meng Q, Yu X and Shi S: Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Mol Cancer. 20:1312021. View Article : Google Scholar : PubMed/NCBI | |
Bartoschek M, Oskolkov N, Bocci M, Lövrot J, Larsson C, Sommarin M, Madsen CD, Lindgren D, Pekar G, Karlsson G, et al: Spatially and functionally distinct subclasses of breast cancer-associated fibroblasts revealed by single cell RNA sequencing. Nat Commun. 9:51502018. View Article : Google Scholar : PubMed/NCBI | |
Newman L: Oncologic anthropology: Global variations in breast cancer risk, biology, and outcome. J Surg Oncol. 128:959–966. 2023. View Article : Google Scholar : PubMed/NCBI | |
Ma J, Chan JJ, Toh CH and Yap YS: Emerging systemic therapy options beyond CDK4/6 inhibitors for hormone receptor-positive HER2-negative advanced breast cancer. NPJ Breast Cancer. 9:742023. View Article : Google Scholar : PubMed/NCBI | |
Pandey K, Katuwal NB, Park N, Hur J, Cho YB, Kim SK, Lee SA, Kim I, Lee SR and Moon YW: Combination of abemaciclib following eribulin overcomes Palbociclib-resistant breast cancer by inhibiting the G2/M cell cycle phase. Cancers (Basel). 14:2102022. View Article : Google Scholar : PubMed/NCBI | |
Piwocka O, Musielak M, Piotrowski I, Kulcenty K, Adamczyk B, Fundowicz M, Suchorska WM and Malicki J: Primary cancer-associated fibroblasts exhibit high heterogeneity among breast cancer subtypes. Rep Pract Oncol Radiother. 28:159–171. 2023. View Article : Google Scholar : PubMed/NCBI | |
Kieffer Y, Hocine HR, Gentric G, Pelon F, Bernard C, Bourachot B, Lameiras S, Albergante L, Bonneau C, Guyard A, et al: Single-cell analysis reveals fibroblast clusters linked to immunotherapy resistance in cancer. Cancer Discov. 10:1330–1351. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yamashita M, Ogawa T, Zhang X, Hanamura N, Kashikura Y, Takamura M, Yoneda M and Shiraishi T: Role of stromal myofibroblasts in invasive breast cancer: Stromal expression of alpha-smooth muscle actin correlates with worse clinical outcome. Breast Cancer. 19:170–176. 2012. View Article : Google Scholar | |
Pelon F, Bourachot B, Kieffer Y, Magagna I, Mermet-Meillon F, Bonnet I, Costa A, Givel AM, Attieh Y, Barbazan J, et al: Cancer-associated fibroblast heterogeneity in axillary lymph nodes drives metastases in breast cancer through complementary mechanisms. Nat Commun. 11:4042020. View Article : Google Scholar : PubMed/NCBI | |
Ershaid N, Sharon Y, Doron H, Raz Y, Shani O, Cohen N, Monteran L, Leider-Trejo L, Ben-Shmuel A, Yassin M, et al: NLRP3 inflammasome in fibroblasts links tissue damage with inflammation in breast cancer progression and metastasis. Nat Commun. 10:43752019. View Article : Google Scholar : PubMed/NCBI | |
Yang SS, Ma S, Dou H, Liu F, Zhang SY, Jiang C, Xiao M and Huang YX: Breast cancer-derived exosomes regulate cell invasion and metastasis in breast cancer via miR-146a to activate cancer associated fibroblasts in tumor microenvironment. Exp Cell Res. 391:1119832020. View Article : Google Scholar : PubMed/NCBI | |
Ren J, Smid M, Iaria J, Salvatori DCF, van Dam H, Zhu HJ, Martens JWM and Ten Dijke P: Cancer-associated fibroblast-derived Gremlin 1 promotes breast cancer progression. Breast Cancer Res. 21:1092019. View Article : Google Scholar : PubMed/NCBI | |
Wu HJ, Hao M, Yeo SK and Guan JL: FAK signaling in cancer-associated fibroblasts promotes breast cancer cell migration and metastasis by exosomal miRNAs-mediated intercellular communication. Oncogene. 39:2539–2549. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chatterjee A, Jana S, Chatterjee S, Wastall LM, Mandal G, Nargis N, Roy H, Hughes TA and Bhattacharyya A: MicroRNA-222 reprogrammed cancer-associated fibroblasts enhance growth and metastasis of breast cancer. Br J Cancer. 121:679–689. 2019. View Article : Google Scholar : PubMed/NCBI | |
Mizrahi JD, Surana R, Valle JW and Shroff RT: Pancreatic cancer. Lancet. 395:2008–2020. 2020. View Article : Google Scholar : PubMed/NCBI | |
Sohal DPS, Kennedy EB, Cinar P, Conroy T, Copur MS, Crane CH, Garrido-Laguna I, Lau MW, Johnson T, Krishnamurthi S, et al: Metastatic pancreatic cancer: ASCO guideline update. J Clin Oncol. 38:3217–3230. 2020. View Article : Google Scholar : PubMed/NCBI | |
Bekkali NLH and Oppong KW: Pancreatic ductal adenocarcinoma epidemiology and risk assessment: Could we prevent? Possibility for an early diagnosis. Endosc Ultrasound. 6(Suppl 3): S58–S61. 2017. View Article : Google Scholar | |
Hosein AN, Brekken RA and Maitra A: Pancreatic cancer stroma: An update on therapeutic targeting strategies. Nat Rev Gastroenterol Hepatol. 17:487–505. 2020. View Article : Google Scholar : PubMed/NCBI | |
Manoukian P, Bijlsma M and van Laarhoven H: The cellular origins of Cancer-associated fibroblasts and their opposing contributions to pancreatic cancer growth. Front Cell Dev Biol. 9:7439072021. View Article : Google Scholar : PubMed/NCBI | |
Geng X, Chen H, Zhao L, Hu J, Yang W, Li G, Cheng C, Zhao Z, Zhang T, Li L and Sun B: Cancer-associated fibroblast (CAF) heterogeneity and targeting therapy of CAFs in pancreatic cancer. Front Cell Dev Biol. 9:6551522021. View Article : Google Scholar : PubMed/NCBI | |
Ohlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, Corbo V, Oni TE, Hearn SA, Lee EJ, et al: Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med. 214:579–596. 2017. View Article : Google Scholar : PubMed/NCBI | |
Kim HW, Lee JC, Paik KH, Kang J, Kim J and Hwang JH: Serum interleukin-6 is associated with pancreatic ductal adenocarcinoma progression pattern. Medicine (Baltimore). 96:e59262017. View Article : Google Scholar : PubMed/NCBI | |
Goulet CR, Champagne A, Bernard G, Vandal D, Chabaud S, Pouliot F and Bolduc S: Cancer-associated fibroblasts induce epithelial-mesenchymal transition of bladder cancer cells through paracrine IL-6 signalling. BMC Cancer. 19:1372019. View Article : Google Scholar : PubMed/NCBI | |
Mirzaei S, Saghari S, Bassiri F, Raesi R, Zarrabi A, Hushmandi K, Sethi G and Tergaonkar V: NF-κB as a regulator of cancer metastasis and therapy response: A focus on epithelial-mesenchymal transition. J Cell Physiol. 237:2770–2795. 2022. View Article : Google Scholar : PubMed/NCBI | |
Rebelo R, Xavier CPR, Giovannetti E and Vasconcelos MH: Fibroblasts in pancreatic cancer: Molecular and clinical perspectives. Trends Mol Med. 29:439–453. 2023. View Article : Google Scholar : PubMed/NCBI | |
Richards KE, Zeleniak AE, Fishel ML, Wu J, Littlepage LE and Hill R: Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene. 36:1770–1778. 2017. View Article : Google Scholar : | |
Aronsson L, Bengtsson A, Toren W, Andersson R and Ansari D: Intraductal papillary mucinous carcinoma versus pancreatic ductal adenocarcinoma: A systematic review and meta-analysis. Int J Surg. 71:91–99. 2019. View Article : Google Scholar : PubMed/NCBI | |
Aronsson L, Andersson R and Ansari D: Intraductal papillary mucinous neoplasm of the pancreas-epidemiology, risk factors, diagnosis, and management. Scand J Gastroenterol. 52:803–815. 2017. View Article : Google Scholar : PubMed/NCBI | |
Bernard V, Semaan A, Huang J, San Lucas FA, Mulu FC, Stephens BM, Guerrero PA, Huang Y, Zhao J, Kamyabi N, et al: Single-cell transcriptomics of pancreatic cancer precursors demonstrates epithelial and microenvironmental heterogeneity as an early event in neoplastic progression. Clin Cancer Res. 25:2194–2205. 2019. View Article : Google Scholar | |
Storandt MH, Zemla TM, Patell K, Naleid N, Gile JJ, Tran NH, Chakrabarti S, Jin Z, Borad M and Mahipal A: Atezolizumab plus bevacizumab as first-line systemic therapy for hepatocellular carcinoma: A multi-institutional cohort study. Oncologist. 29:986–996. 2024. View Article : Google Scholar : PubMed/NCBI | |
Selene II, Ozen M and Patel RA: Hepatocellular carcinoma: Advances in systemic therapy. Semin Intervent Radiol. 41:56–62. 2024. View Article : Google Scholar : PubMed/NCBI | |
Singal AG, Llovet JM, Yarchoan M, Mehta N, Heimbach JK, Dawson LA, Jou JH, Kulik LM, Agopian VG, Marrero JA, et al: AASLD practice guidance on prevention, diagnosis, and treatment of hepatocellular carcinoma. Hepatology. 78:1922–1965. 2023. View Article : Google Scholar : PubMed/NCBI | |
Loh JJ, Li TW, Zhou L, Wong TL, Liu X, Ma VWS, Lo CM, Man K, Lee TK, Ning W, et al: FSTL1 secreted by activated fibroblasts promotes hepatocellular carcinoma metastasis and stemness. Cancer Res. 81:5692–5705. 2021. View Article : Google Scholar : PubMed/NCBI | |
Liu J, Chen S, Wang W, Ning BF, Chen F, Shen W, Ding J, Chen W, Xie WF, Zhang X, et al: Cancer-associated fibroblasts promote hepatocellular carcinoma metastasis through chemokine-activated hedgehog and TGF-β pathways. Cancer Lett. 379:49–59. 2016. View Article : Google Scholar : PubMed/NCBI | |
Jia CC, Wang TT, Liu W, Fu BS, Hua X, Wang GY, Li TJ, Li X, Wu XY, Tai Y, et al: Cancer-associated fibroblasts from hepatocellular carcinoma promote malignant cell proliferation by HGF secretion. PLoS One. 8:e632432013. View Article : Google Scholar : PubMed/NCBI | |
Xiong S, Wang R, Chen Q, Luo J, Wang J, Zhao Z, Li Y, Wang Y, Wang X and Cheng B: Cancer-associated fibroblasts promote stem cell-like properties of hepatocellular carcinoma cells through IL-6/STAT3/Notch signaling. Am J Cancer Res. 8:302–316. 2018.PubMed/NCBI | |
Li Y, Wang R, Xiong S, Wang X, Zhao Z, Bai S, Wang Y, Zhao Y and Cheng B: Cancer-associated fibroblasts promote the stemness of CD24+ liver cells via paracrine signaling. J Mol Med (Berl). 97:243–255. 2019. View Article : Google Scholar | |
Zhu GQ, Tang Z, Huang R, Qu WF, Fang Y, Yang R, Tao CY, Gao J, Wu XL, Sun HX, et al: CD36+ cancer-associated fibroblasts provide immunosuppressive microenvironment for hepatocellular carcinoma via secretion of macrophage migration inhibitory factor. Cell Discov. 9:252023. View Article : Google Scholar : | |
Eun JW, Yoon JH, Ahn HR, Kim S, Kim YB, Lim SB, Park W, Kang TW, Baek GO, Yoon MG, et al: Cancer-associated fibroblast-derived secreted phosphoprotein 1 contributes to resistance of hepatocellular carcinoma to sorafenib and lenvatinib. Cancer Commun (Lond). 43:455–479. 2023. View Article : Google Scholar : PubMed/NCBI | |
Xia S, Pan Y, Liang Y, Xu J and Cai X: The microenvironmental and metabolic aspects of sorafenib resistance in hepatocellular carcinoma. EBioMedicine. 51:1026102020. View Article : Google Scholar : PubMed/NCBI | |
Yu L, Liu Q, Huo J, Wei F and Guo W: Cancer-associated fibroblasts induce immunotherapy resistance in hepatocellular carcinoma animal model. Cell Mol Biol (Noisy-le-Grand). 66:36–40. 2020. View Article : Google Scholar : PubMed/NCBI | |
Lotfollahzadeh S, Recio-Boiles A and Cagir B: Colon cancer. StatPearls. Treasure Island (FL) with ineligible companies. Disclosure: Alejandro Recio-Boiles declares no relevant financial relationships with ineligible companies. Disclosure: Burt Cagir declares no relevant financial relationships with ineligible companies.2024. | |
Constantinou V and Constantinou C: Focusing on colorectal cancer in young adults (Review). Mol Clin Oncol. 20:82024. View Article : Google Scholar | |
Siegel RL, Wagle NS, Cercek A, Smith RA and Jemal A: Colorectal cancer statistics, 2023. CA Cancer J Clin. 73:233–254. 2023.PubMed/NCBI | |
Zeineddine FA, Zeineddine MA, Yousef A, Gu Y, Chowdhury S, Dasari A, Huey RW, Johnson B, Kee B, Lee MS, et al: Survival improvement for patients with metastatic colorectal cancer over twenty years. NPJ Precis Oncol. 7:162023. View Article : Google Scholar : PubMed/NCBI | |
Morris VK, Kennedy EB, Baxter NN, Benson AB III, Cercek A, Cho M, Ciombor KK, Cremolini C, Davis A, Deming DA, et al: Treatment of metastatic colorectal cancer: ASCO Guideline. J Clin Oncol. 41:678–700. 2023. View Article : Google Scholar | |
Deng L, Jiang N, Zeng J, Wang Y and Cui H: The versatile roles of Cancer-associated fibroblasts in colorectal cancer and therapeutic implications. Front Cell Dev Biol. 9:7332702021. View Article : Google Scholar : PubMed/NCBI | |
Wikberg ML, Edin S, Lundberg IV, Van Guelpen B, Dahlin AM, Rutegård J, Stenling R, Oberg A and Palmqvist R: High intratumoral expression of fibroblast activation protein (FAP) in colon cancer is associated with poorer patient prognosis. Tumour Biol. 34:1013–1020. 2013. View Article : Google Scholar : PubMed/NCBI | |
Hu JL, Wang W, Lan XL, Zeng ZC, Liang YS, Yan YR, Song FY, Wang FF, Zhu XH, Liao WJ, et al: CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Mol Cancer. 18:912019. View Article : Google Scholar : PubMed/NCBI | |
Aizawa T, Karasawa H, Funayama R, Shirota M, Suzuki T, Maeda S, Suzuki H, Yamamura A, Naitoh T, Nakayama K and Unno M: Cancer-associated fibroblasts secrete Wnt2 to promote cancer progression in colorectal cancer. Cancer Med. 8:6370–6382. 2019. View Article : Google Scholar : PubMed/NCBI | |
Siegel RL, Giaquinto AN and Jemal A: Cancer statistics, 2024. CA Cancer J Clin. 74:12–49. 2024.PubMed/NCBI | |
Gridelli C, Rossi A, Carbone DP, Guarize J, Karachaliou N, Mok T, Petrella F, Spaggiari L and Rosell R: Non-small-cell lung cancer. Nat Rev Dis Primers. 1:150092015. View Article : Google Scholar : PubMed/NCBI | |
Herbst RS, Morgensztern D and Boshoff C: The biology and management of Non-small cell lung cancer. Nature. 553:446–454. 2018. View Article : Google Scholar : PubMed/NCBI | |
Kim SH, Choe C, Shin YS, Jeon MJ, Choi SJ, Lee J, Bae GY, Cha HJ and Kim J: Human lung cancer-associated fibroblasts enhance motility of Non-small cell lung cancer cells in co-culture. Anticancer Res. 33:2001–2009. 2013.PubMed/NCBI | |
An J, Enomoto A, Weng L, Kato T, Iwakoshi A, Ushida K, Maeda K, Ishida-Takagishi M, Ishii G, Ming S, et al: Significance of cancer-associated fibroblasts in the regulation of gene expression in the leading cells of invasive lung cancer. J Cancer Res Clin Oncol. 139:379–388. 2013. View Article : Google Scholar | |
Chen WJ, Ho CC, Chang YL, Chen HY, Lin CA, Ling TY, Yu SL, Yuan SS, Chen YJ, Lin CY, et al: Cancer-associated fibroblasts regulate the plasticity of lung cancer stemness via paracrine signalling. Nat Commun. 5:34722014. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Cao L, Wang H, Liu B, Zhang Q, Meng Z, Wu X, Zhou Q and Xu K: Cancer-associated fibroblasts enhance metastatic potential of lung cancer cells through IL-6/STAT3 signaling pathway. Oncotarget. 8:76116–76128. 2017. View Article : Google Scholar : PubMed/NCBI | |
Vicent S, Sayles LC, Vaka D, Khatri P, Gevaert O, Chen R, Zheng Y, Gillespie AK, Clarke N, Xu Y, et al: Cross-species functional analysis of cancer-associated fibroblasts identifies a critical role for CLCF1 and IL-6 in non-small cell lung cancer in vivo. Cancer Res. 72:5744–5756. 2012. View Article : Google Scholar : PubMed/NCBI | |
Yang F, Yan Y, Yang Y, Hong X, Wang M, Yang Z, Liu B and Ye L: MiR-210 in exosomes derived from CAFs promotes non-small cell lung cancer migration and invasion through PTEN/PI3K/AKT pathway. Cell Signal. 73:1096752020. View Article : Google Scholar : PubMed/NCBI | |
Xiang H, Ramil CP, Hai J, Zhang C, Wang H, Watkins AA, Afshar R, Georgiev P, Sze MA, Song XS, et al: Cancer-associated fibroblasts promote immunosuppression by inducing ROS-generating monocytic MDSCs in lung squamous cell carcinoma. Cancer Immunol Res. 8:436–450. 2020. View Article : Google Scholar : PubMed/NCBI | |
Kanwal B, Biswas S, Seminara RS and Jeet C: Immunotherapy in advanced Non-small cell lung cancer patients: Ushering chemotherapy through the checkpoint inhibitors? Cureus. 10:e32542018.PubMed/NCBI | |
Herzog BH, Baer JM, Borcherding N, Kingston NL, Belle JI, Knolhoff BL, Hogg GD, Ahmad F, Kang LI, Petrone J, et al: Tumor-associated fibrosis impairs immune surveillance and response to immune checkpoint blockade in non-small cell lung cancer. Sci Transl Med. 15:eadh80052023. View Article : Google Scholar : PubMed/NCBI | |
Jenkins L, Jungwirth U, Avgustinova A, Iravani M, Mills A, Haider S, Harper J and Isacke CM: Cancer-associated fibroblasts suppress CD8+ T-cell infiltration and confer resistance to Immune-checkpoint blockade. Cancer Res. 82:2904–2917. 2022. View Article : Google Scholar : PubMed/NCBI | |
Tauriello DVF: Targeting CAFs to improve Anti-PD-1 checkpoint immunotherapy. Cancer Res. 83:655–656. 2023. View Article : Google Scholar : PubMed/NCBI | |
Shintani Y, Kimura T, Funaki S, Ose N, Kanou T and Fukui E: Therapeutic targeting of Cancer-associated fibroblasts in the Non-small cell lung cancer tumor microenvironment. Cancers (Basel). 15:3352023. View Article : Google Scholar : PubMed/NCBI | |
Li M, Wu B, Li L, Lv C and Tian Y: Reprogramming of cancer-associated fibroblasts combined with immune checkpoint inhibitors: A potential therapeutic strategy for cancers. Biochimica et biophysica acta. Rev Cancer. 1878:1889452023. View Article : Google Scholar | |
Rawla P: Epidemiology of prostate cancer. World J Oncol. 10:63–89. 2019. View Article : Google Scholar : PubMed/NCBI | |
Culp MB, Soerjomataram I, Efstathiou JA, Bray F and Jemal A: Recent global patterns in prostate cancer incidence and mortality rates. Eur Urol. 77:38–52. 2020. View Article : Google Scholar | |
Hammerer P and Manka L: Androgen deprivation therapy for advanced prostate cancer. Urologic Oncology. Merseburger AS and Burger M: Springer International Publishing; Cham: pp. 255–276. 2019, View Article : Google Scholar | |
Figueiredo A, Costa L, Mauricio MJ, Figueira L, Ramos R and Martins-da-Silva C: Nonmetastatic castration-resistant prostate cancer: Current challenges and trends. Clin Drug Investig. 42:631–642. 2022. View Article : Google Scholar : PubMed/NCBI | |
Bubendorf L, Schopfer A, Wagner U, Sauter G, Moch H, Willi N, Gasser TC and Mihatsch MJ: Metastatic patterns of prostate cancer: An autopsy study of 1,589 patients. Hum Pathol. 31:578–583. 2000. View Article : Google Scholar : PubMed/NCBI | |
Begley LA, Kasina S, MacDonald J and Macoska JA: The inflammatory microenvironment of the aging prostate facilitates cellular proliferation and hypertrophy. Cytokine. 43:194–199. 2008. View Article : Google Scholar : PubMed/NCBI | |
Levesque C and Nelson PS: Cellular constituents of the prostate stroma: Key contributors to prostate cancer progression and therapy resistance. Cold Spring Harb Perspect Med. 8:a0305102018. View Article : Google Scholar | |
ChallaSivaKanaka S, Vickman RE, Kakarla M, Hayward SW and Franco OE: Fibroblast heterogeneity in prostate carcinogenesis. Cancer Lett. 525:76–83. 2022. View Article : Google Scholar : | |
Franco OE and Hayward SW: Targeting the tumor stroma as a novel therapeutic approach for prostate cancer. Adv Pharmacol. 65:267–313. 2012. View Article : Google Scholar : PubMed/NCBI | |
Jung Y, Kim JK, Shiozawa Y, Wang J, Mishra A, Joseph J, Berry JE, McGee S, Lee E, Sun H, et al: Recruitment of mesenchymal stem cells into prostate tumours promotes metastasis. Nat Commun. 4:17952013. View Article : Google Scholar : PubMed/NCBI | |
Bavik C, Coleman I, Dean JP, Knudsen B, Plymate S and Nelson PS: The gene expression program of prostate fibroblast senescence modulates neoplastic epithelial cell proliferation through paracrine mechanisms. Cancer Res. 66:794–802. 2006. View Article : Google Scholar : PubMed/NCBI | |
Bedeschi M, Marino N, Cavassi E, Piccinini F and Tesei A: Cancer-associated fibroblast: Role in prostate cancer progression to metastatic disease and therapeutic resistance. Cells. 12:8022023. View Article : Google Scholar : PubMed/NCBI | |
Bonollo F, Thalmann GN, Kruithof-de Julio M and Karkampouna S: The role of Cancer-Associated fibroblasts in prostate cancer tumorigenesis. Cancers (Basel). 12:18872020. View Article : Google Scholar : PubMed/NCBI | |
Lavie D, Ben-Shmuel A, Erez N and Scherz-Shouval R: Cancer-associated fibroblasts in the single-cell era. Nat Cancer. 3:793–807. 2022. View Article : Google Scholar : PubMed/NCBI | |
Nguyen EV, Pereira BA, Lawrence MG, Ma X, Rebello RJ, Chan H, Niranjan B, Wu Y, Ellem S, Guan X, et al: Proteomic profiling of human prostate Cancer-associated fibroblasts (CAF) reveals LOXL2-dependent regulation of the tumor microenvironment. Mol Cell Proteomics. 18:1410–1427. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wadosky KM and Koochekpour S: Molecular mechanisms underlying resistance to androgen deprivation therapy in prostate cancer. Oncotarget. 7:64447–64470. 2016. View Article : Google Scholar : PubMed/NCBI | |
Boudadi K and Antonarakis ES: Resistance to novel antiandrogen therapies in metastatic Castration-resistant prostate cancer. Clin Med Insights Oncol. 10(Suppl 1): S1–S9. 2016. | |
Chandrasekar T, Yang JC, Gao AC and Evans CP: Mechanisms of resistance in castration-resistant prostate cancer (CRPC). Transl Androl Urol. 4:365–380. 2015. | |
Leach DA and Buchanan G: Stromal androgen receptor in prostate cancer development and progression. Cancers (Basel). 9:102017. View Article : Google Scholar : PubMed/NCBI | |
Cioni B, Nevedomskaya E, Melis MHM, van Burgsteden J, Stelloo S, Hodel E, Spinozzi D, de Jong J, van der Poel H, de Boer JP, et al: Loss of androgen receptor signaling in prostate cancer-associated fibroblasts (CAFs) promotes CCL2- and CXCL8-mediated cancer cell migration. Mol Oncol. 12:1308–1323. 2018. View Article : Google Scholar : PubMed/NCBI | |
Eder T, Weber A, Neuwirt H, Grünbacher G, Ploner C, Klocker H, Sampson N and Eder IE: Cancer-associated fibroblasts modify the response of prostate cancer cells to androgen and Anti-androgens in Three-dimensional spheroid culture. Int J Mol Sci. 17:14582016. View Article : Google Scholar : PubMed/NCBI | |
Cheteh EH, Augsten M, Rundqvist H, Bianchi J, Sarne V, Egevad L, Bykov VJ, Östman A and Wiman KG: Human cancer-associated fibroblasts enhance glutathione levels and antagonize drug-induced prostate cancer cell death. Cell Death Dis. 8:e28482017. View Article : Google Scholar : PubMed/NCBI | |
Baumhoer D, Hench J and Amary F: Recent advances in molecular profiling of bone and soft tissue tumors. Skeletal Radiol. 53:1925–1936. 2024. View Article : Google Scholar : PubMed/NCBI | |
Anderson WJ and Doyle LA: Updates from the 2020 World health organization classification of soft tissue and bone tumours. Histopathology. 78:644–657. 2021. View Article : Google Scholar : PubMed/NCBI | |
Ehnman M, Chaabane W, Haglund F and Tsagkozis P: The tumor microenvironment of pediatric sarcoma: Mesenchymal mechanisms regulating cell migration and metastasis. Curr Oncol Rep. 21:902019. View Article : Google Scholar : PubMed/NCBI | |
Plikus MV, Wang X, Sinha S, Forte E, Thompson SM, Herzog EL, Driskell RR, Rosenthal N, Biernaskie J and Fibroblasts Horsley V: Origins, definitions, and functions in health and disease. Cell. 184:3852–3872. 2021. View Article : Google Scholar : PubMed/NCBI | |
Resag A, Toffanin G, Benesova I, Müller L, Potkrajcic V, Ozaniak A, Lischke R, Bartunkova J, Rosato A, Jöhrens K, et al: The immune contexture of liposarcoma and its clinical implications. Cancers. 14:45782022. View Article : Google Scholar : PubMed/NCBI | |
Jones JEC, Rabhi N, Orofino J, Gamini R, Perissi V, Vernochet C and Farmer SR: The adipocyte acquires a fibroblast-like transcriptional signature in response to a high fat diet. Sci Rep. 10:23802020. View Article : Google Scholar : PubMed/NCBI | |
Lendahl U, Muhl L and Betsholtz C: Identification, discrimination and heterogeneity of fibroblasts. Nat Commun. 13:34092022. View Article : Google Scholar : PubMed/NCBI | |
Bochet L, Lehuede C, Dauvillier S, Wang YY, Dirat B, Laurent V, Dray C, Guiet R, Maridonneau-Parini I, Le Gonidec S, et al: Adipocyte-derived fibroblasts promote tumor progression and contribute to the desmoplastic reaction in breast cancer. Cancer Res. 73:5657–5668. 2013. View Article : Google Scholar : PubMed/NCBI | |
Bouche C and Quail DF: Fueling the tumor microenvironment with cancer-associated adipocytes. Cancer Res. 83:1170–1172. 2023. View Article : Google Scholar : PubMed/NCBI | |
Harati K, Daigeler A, Hirsch T, Jacobsen F, Behr B, Wallner C, Lehnhardt M and Becerikli M: Tumor-associated fibroblasts promote the proliferation and decrease the doxorubicin sensitivity of liposarcoma cells. Int J Mol Med. 37:1535–1541. 2016. View Article : Google Scholar : PubMed/NCBI | |
Xu C, Yan L, Guan X, Wang Z, Wu J, Lv A, Liu D, Liu F, Dong B, Zhao M, et al: Tsp2 facilitates tumor-associated fibroblasts formation and promotes tumor progression in retroperitoneal liposarcoma. Int J Biol Sci. 18:5038–5055. 2022. View Article : Google Scholar : PubMed/NCBI | |
Kalluri R: The biology and function of fibroblasts in cancer. Nat Rev Cancer. 16:582–598. 2016. View Article : Google Scholar : PubMed/NCBI | |
Skapek SX, Ferrari A, Gupta AA, Lupo PJ, Butler E, Shipley J, Barr FG and Hawkins DS: Rhabdomyosarcoma. Nat Rev Dis Primers. 5:12019. View Article : Google Scholar : PubMed/NCBI | |
Hettmer S and Wagers AJ: Muscling in: Uncovering the origins of rhabdomyosarcoma. Nat Med. 16:171–173. 2010. View Article : Google Scholar : PubMed/NCBI | |
Chapman MA, Meza R and Lieber RL: Skeletal muscle fibroblasts in health and disease. Differentiation. 92:108–115. 2016. View Article : Google Scholar : PubMed/NCBI | |
Tarnowski M, Grymula K, Liu R, Tarnowska J, Drukala J, Ratajczak J, Mitchell RA, Ratajczak MZ and Kucia M: Macrophage migration inhibitory factor is secreted by rhabdomyosarcoma cells, modulates tumor metastasis by binding to CXCR4 and CXCR7 receptors and inhibits recruitment of cancer-associated fibroblasts. Mol Cancer Res. 8:1328–1343. 2010. View Article : Google Scholar : PubMed/NCBI | |
Wysoczynski M, Shin DM, Kucia M and Ratajczak MZ: Selective upregulation of interleukin-8 by human rhabdomyosarcomas in response to hypoxia: Therapeutic implications. Int J Cancer. 126:371–381. 2010. View Article : Google Scholar | |
Awaji M, Saxena S, Wu L, Prajapati DR, Purohit A, Varney ML, Kumar S, Rachagani S, Ly QP, Jain M, et al: CXCR2 signaling promotes secretory cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. FASEB J. 34:9405–9418. 2020. View Article : Google Scholar : PubMed/NCBI | |
Dhayni K, Zibara K, Issa H, Kamel S and Bennis Y: Targeting CXCR1 and CXCR2 receptors in cardiovascular diseases. Pharmacol Ther. 237:1082572022. View Article : Google Scholar : PubMed/NCBI | |
Ghayad SE, Rammal G, Ghamloush F, Basma H, Nasr R, Diab-Assaf M, Chelala C and Saab R: Exosomes derived from embryonal and alveolar rhabdomyosarcoma carry differential miRNA cargo and promote invasion of recipient fibroblasts. Sci Rep. 6:370882016. View Article : Google Scholar : PubMed/NCBI | |
Fahs A, Hussein N, Zalzali H, Ramadan F, Ghamloush F, Tamim H, El Homsi M, Badran B, Boulos F, Tawil A, et al: CD147 promotes tumorigenesis via Exosome-mediated signaling in rhabdomyosarcoma. Cells. 11:22672022. View Article : Google Scholar : PubMed/NCBI | |
Burns J, Wilding CP, Krasny L, Zhu X, Chadha M, Tam YB, Ps H, Mahalingam AH, Lee ATJ, Arthur A, et al: The proteomic landscape of soft tissue sarcomas. Nature Commun. 14:38342023. View Article : Google Scholar | |
Stork T, Hegedus B, Guder W, Hamacher R, Hardes J, Kaths M, Plönes T, Pöttgen C, Schildhaus HU, Streitbürger A, et al: Prognostic factors for leiomyosarcoma with isolated metastases to the lungs: Impact of metastasectomy. Ann Surg Oncol. 29:4429–4436. 2022. View Article : Google Scholar : PubMed/NCBI | |
Tseng WW, Swallow CJ, Strauss DC, Bonvalot S, Rutkowski P, Ford SJ, Gonzalez RJ, Gladdy RA, Gyorki DE, Fairweather M, et al: Management of locally recurrent retroperitoneal sarcoma in the Adult: An updated consensus approach from the transatlantic australasian retroperitoneal sarcoma working group. Ann Surg Oncol. 29:7335–7348. 2022. View Article : Google Scholar : PubMed/NCBI | |
Tran V and Slavin J: Immunohistochemistry in bone and soft tissue tumours. Sarcoma: A Practical Guide to Multidisciplinary Management. Choong PFM: Springer Singapore; Singapore: pp. 119–134. 2021, View Article : Google Scholar | |
Porrello G, Cannella R, Randazzo A, Badalamenti G, Brancatelli G and Vernuccio F: CT and MR imaging of retroperitoneal sarcomas: A practical guide for the radiologist. Cancers (Basel). 15:29852023. View Article : Google Scholar : PubMed/NCBI | |
Kerrison WGJ, Thway K, Jones RL and Huang PH: The biology and treatment of leiomyosarcomas. Crit Rev Oncol Hematol. 184:1039552023. View Article : Google Scholar : PubMed/NCBI | |
Tai Y, Woods EL, Dally J, Kong D, Steadman R, Moseley R and Midgley AC: Myofibroblasts: Function, formation, and scope of molecular therapies for skin fibrosis. Biomolecules. 11:10952021. View Article : Google Scholar : PubMed/NCBI | |
Nagao Y, Yokoi A, Yoshida K, Kitagawa M, Asano-Inami E, Kato T, Ishikawa M, Yamamoto Y and Kajiyama H: Uterine leiomyosarcoma cell-derived extracellular vesicles induce the formation of cancer-associated fibroblasts. Biochim Biophys Acta Mol Basis Dis. 1870:1671032024. View Article : Google Scholar : PubMed/NCBI | |
Canter RJ, Beal S, Borys D, Martinez SR, Bold RJ and Robbins AS: Interaction of histologic subtype and histologic grade in predicting survival for soft-tissue sarcomas. J Am Coll Surg. 210:191–198.e2. 2010. View Article : Google Scholar : PubMed/NCBI | |
Sun H, Liu J, Hu F, Xu M, Leng A, Jiang F and Chen K: Current research and management of undifferentiated pleomorphic sarcoma/myofibrosarcoma. Front Genet. 14:11094912023. View Article : Google Scholar : PubMed/NCBI | |
Ozzello L, Stout AP and Murray MR: Cultural characteristics of malignant histiocytomas and fibrous xanthomas. Cancer. 16:331–344. 1963. View Article : Google Scholar : PubMed/NCBI | |
O'Brien JE and Stout AP: Malignant fibrous xanthomas. Cancer. 17:1445–1455. 1964. View Article : Google Scholar : PubMed/NCBI | |
Iwasaki H, Isayama T, Johzaki H and Kikuchi M: Malignant fibrous histiocytoma. Evidence of perivascular mesenchymal cell origin immunocytochemical studies with monoclonal anti-MFH antibodies. Am J Pathol. 128:528–537. 1987.PubMed/NCBI | |
Erlandson RA and Antonescu CR: The rise and fall of malignant fibrous histiocytoma. Ultrastruct Pathol. 28:283–289. 2004. View Article : Google Scholar | |
Widemann BC and Italiano A: Biology and management of undifferentiated pleomorphic sarcoma, myxofibrosarcoma, and malignant peripheral nerve sheath tumors: State of the art and perspectives. J Clin Oncol. 36:160–167. 2018. View Article : Google Scholar : | |
Osanai T, Yamakawa M, Suda A and Watanabe Y: Metamorphosed fibroblasts and their relation to the histogenesis of malignant fibrous histiocytoma in experimental murine model. Histol Histopathol. 15:697–705. 2000.PubMed/NCBI | |
Taxy JB and Battifora H: Malignant fibrous histiocytoma. An electron microscopic study. Cancer. 40:254–267. 1977. View Article : Google Scholar : PubMed/NCBI | |
Wood GS, Beckstead JH, Turner RR, Hendrickson MR, Kempson RL and Warnke RA: Malignant fibrous histiocytoma tumor cells resemble fibroblasts. Am J Surg Pathol. 10:323–335. 1986. View Article : Google Scholar : PubMed/NCBI | |
Chang Y, Cho B, Kim S and Kim J: Direct conversion of fibroblasts to osteoblasts as a novel strategy for bone regeneration in elderly individuals. Exp Mol Med. 51:1–8. 2019.PubMed/NCBI | |
Neumann E, Lefevre S, Zimmermann B, Gay S and Muller-Ladner U: Rheumatoid arthritis progression mediated by activated synovial fibroblasts. Trends Mol Med. 16:458–468. 2010. View Article : Google Scholar : PubMed/NCBI | |
Setty BA, Gikandi A and DuBois SG: Ewing sarcoma drug therapy: Current standard of care and emerging agents. Paediatr Drugs. 25:389–397. 2023. View Article : Google Scholar : PubMed/NCBI | |
Riggi N, Suva ML and Stamenkovic I: Ewing's Sarcoma. N Engl J Med. 384:154–164. 2021. View Article : Google Scholar : PubMed/NCBI | |
Volchenboum SL, Andrade J, Huang L, Barkauskas DA, Krailo M, Womer RB, Ranft A, Potratz J, Dirksen U, Triche TJ and Lawlor ER: Gene expression profiling of ewing sarcoma tumors reveals the prognostic importance of Tumor-stromal interactions: A report from the Children's oncology group. J Pathol Clin Res. 1:83–94. 2015. View Article : Google Scholar : PubMed/NCBI | |
Wrenn ED, Apfelbaum AA, Rudzinski ER, Deng X, Jiang W, Sud S, Van Noord RA, Newman EA, Garcia NM, Miyaki A, et al: Cancer-associated Fibroblast-like tumor cells remodel the ewing sarcoma tumor microenvironment. Clin Cancer Res. 29:5140–5154. 2023. View Article : Google Scholar : PubMed/NCBI | |
Li S, Zhang H, Liu J and Shang G: Targeted therapy for osteosarcoma: A review. J Cancer Res Clin Oncol. 149:6785–6797. 2023. View Article : Google Scholar : PubMed/NCBI | |
Panez-Toro I, Munoz-Garcia J, Vargas-Franco JW, Renodon-Cornière A, Heymann MF, Lézot F and Heymann D: Advances in osteosarcoma. Curr Osteoporos Rep. 21:330–343. 2023. View Article : Google Scholar : PubMed/NCBI | |
Misaghi A, Goldin A, Awad M and Kulidjian AA: Osteosarcoma: A comprehensive review. SICOT J. 4:122018. View Article : Google Scholar : PubMed/NCBI | |
Hu J, Lazar AJ, Ingram D, Wang WL, Zhang W, Jia Z, Ragoonanan D, Wang J, Xia X, Mahadeo K, et al: Cell membrane-anchored and tumor-targeted IL-12 T-cell therapy destroys cancer-associated fibroblasts and disrupts extracellular matrix in heterogenous osteosarcoma xenograft models. J Immunother Cancer. 12:e0069912024. View Article : Google Scholar : PubMed/NCBI | |
Wang JW, Wu XF, Gu XJ and Jiang XH: Exosomal miR-1228 from Cancer-associated fibroblasts promotes cell migration and invasion of osteosarcoma by directly targeting SCAI. Oncol Res. 27:979–986. 2019. View Article : Google Scholar | |
Mazumdar A, Urdinez J, Boro A, Migliavacca J, Arlt MJE, Muff R, Fuchs B, Snedeker JG and Gvozdenovic A: Osteosarcoma-derived extracellular vesicles induce lung fibroblast reprogramming. Int J Mol Sci. 21:54512020. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Liu Z, Yang X, Lu W, Chen Y, Lin Y, Wang J, Lin S and Yun JP: H3K27 acetylation activated-COL6A1 promotes osteosarcoma lung metastasis by repressing STAT1 and activating pulmonary cancer-associated fibroblasts. Theranostics. 11:1473–1492. 2021. View Article : Google Scholar : PubMed/NCBI | |
David MS, Kelly E and Zoellner H: Opposite cytokine synthesis by fibroblasts in contact co-culture with osteosarcoma cells compared with transwell co-cultures. Cytokine. 62:48–51. 2013. View Article : Google Scholar : PubMed/NCBI | |
Xu Y, Chen P, Liu D, Xu Q, Meng H and Wang X: Exploration of s new biomarker in osteosarcoma and association with clinical outcomes: TOP2A+ cancer associated fibroblasts. J Gene Med. 25:e35282023. View Article : Google Scholar | |
Liu Y, Han X, Han Y, Bi J, Wu Y, Xiang D, Zhang Y, Bi W, Xu M and Li J: Integrated transcriptomic analysis systematically reveals the heterogeneity and molecular characterization of cancer-associated fibroblasts in osteosarcoma. Gene. 907:1482862024. View Article : Google Scholar : PubMed/NCBI | |
LeBleu VS and Neilson EG: Origin and functional heterogeneity of fibroblasts. FASEB. 34:3519–3536. 2020. View Article : Google Scholar | |
Pittenger MF, Discher DE, Peault BM, Phinney DG, Hare JM and Caplan AI: Mesenchymal stem cell perspective: Cell biology to clinical progress. NPJ Regen Med. 4:222019. View Article : Google Scholar : PubMed/NCBI | |
Sannino G, Marchetto A, Kirchner T and Grunewald TGP: Epithelial-to-mesenchymal and mesenchymal-to-epithelial transition in mesenchymal tumors: A paradox in sarcomas? Cancer Res. 77:4556–4561. 2017. View Article : Google Scholar : PubMed/NCBI | |
Mehta A and Stanger BZ: Lineage Plasticity: The new cancer hallmark on the block. Cancer Res. 84:184–191. 2024. View Article : Google Scholar : | |
Strating E, Verhagen MP, Wensink E, Dünnebach E, Wijler L, Aranguren I, De la Cruz AS, Peters NA, Hageman JH, van der Net MMC, et al: Co-cultures of colon cancer cells and cancer-associated fibroblasts recapitulate the aggressive features of mesenchymal-like colon cancer. Front Immunol. 14:10539202023. View Article : Google Scholar : PubMed/NCBI | |
Jeong SY, Lee JH, Shin Y, Chung S and Kuh HJ: Co-culture of tumor spheroids and fibroblasts in a collagen Matrix-incorporated microfluidic chip mimics reciprocal activation in solid tumor microenvironment. PLoS One. 11:e01590132016. View Article : Google Scholar : PubMed/NCBI | |
Abercrombie M, Heaysman JE and Karthauser HM: Social behaviour of cells in tissue culture. III. Mutual influence of sarcoma cells and fibroblasts. Exp Cell Res. 13:276–291. 1957. View Article : Google Scholar : PubMed/NCBI | |
Abercrombie M and Heaysman JE: Invasive behavior between sarcoma and fibroblast populations in cell culture. J Natl Cancer Inst. 56:561–570. 1976. View Article : Google Scholar : PubMed/NCBI | |
Johnson GS, Friedman RM and Pastan I: Restoration of several morphological characteristics of normal fibroblasts in sarcoma cells treated with adenosine-3': 5'-cyclic monophosphate and its derivatives. Proc Natl Acad Sci USA. 68:425–429. 1971. View Article : Google Scholar | |
Dahlberg WK, Little JB, Fletcher JA, Suit HD and Okunieff P: Radiosensitivity in vitro of human soft tissue sarcoma cell lines and skin fibroblasts derived from the same patients. Int J Radiat Biol. 63:191–198. 1993. View Article : Google Scholar : PubMed/NCBI | |
Fisher C: Low-grade sarcomas with CD34-positive fibroblasts and Low-grade myofibroblastic sarcomas. Ultrastruct Pathol. 28:291–305. 2004. View Article : Google Scholar | |
Broz MT, Ko EY, Ishaya K, Xiao J, De Simone M, Hoi XP, Piras R, Gala B, Tessaro FHG, Karlstaedt A, et al: Metabolic targeting of cancer associated fibroblasts overcomes T-cell exclusion and chemoresistance in Soft-tissue sarcomas. Nat Commun. 15:24982024. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Sinjab A, Min J, Han G, Paradiso F, Zhang Y, Wang R, Pei G, Dai Y, Liu Y, et al: Conserved spatial subtypes and cellular neighborhoods of cancer-associated fibroblasts revealed by single-cell spatial multi-omics. Cancer Cell. 43:905–924.e6. 2025. View Article : Google Scholar : PubMed/NCBI | |
Kazakova AN, Lukina MM, Anufrieva KS, Bekbaeva IV, Ivanova OM, Shnaider PV, Slonov A, Arapidi GP and Shender VO: Exploring the diversity of cancer-associated fibroblasts: Insights into mechanisms of drug resistance. Front Cell Dev Biol. 12:14031222024. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, McAndrews KM and Kalluri R: Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat Rev Clin Oncol. 18:792–804. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang LC, Lo A, Scholler J, Sun J, Majumdar RS, Kapoor V, Antzis M, Cotner CE, Johnson LA, Durham AC, 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. 2:154–166. 2014. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Sun Y, Wang P, Li S, Dong Y, Zhou M, Shi B, Jiang H, Sun R and Li Z: FAP-targeted CAR-T suppresses MDSCs recruitment to improve the antitumor efficacy of claudin18.2-targeted CAR-T against pancreatic cancer. J Transl Med. 21:2552023. View Article : Google Scholar : PubMed/NCBI | |
Gao Y, Li X, Zeng C, Liu C, Hao Q, Li W, Zhang K, Zhang W, Wang S, Zhao H, et al: CD63+ Cancer-associated fibroblasts confer tamoxifen resistance to breast cancer cells through exosomal miR-22. Adv Sci (Weinh). 7:20025182020. View Article : Google Scholar | |
Zhou P, Du X, Jia W, Feng K and Zhang Y: Engineered extracellular vesicles for targeted reprogramming of cancer-associated fibroblasts to potentiate therapy of pancreatic cancer. Signal Transduct Target Ther. 9:1512024. View Article : Google Scholar : PubMed/NCBI | |
Peng D, Fu M, Wang M, Wei Y and Wei X: Targeting TGF-β signal transduction for fibrosis and cancer therapy. Mol Cancer. 21:1042022. View Article : Google Scholar | |
Akhmetshina A, Palumbo K, Dees C, Bergmann C, Venalis P, Zerr P, Horn A, Kireva T, Beyer C, Zwerina J, et al: Activation of canonical Wnt signalling is required for TGF-beta-mediated fibrosis. Nat Commun. 3:7352012. View Article : Google Scholar | |
Xu L, Cui WH, Zhou WC, Li DL, Li LC, Zhao P, Mo XT, Zhang Z and Gao J: Activation of Wnt/β-catenin signalling is required for TGF-β/Smad2/3 signalling during myofibroblast proliferation. J Cell Mol Med. 21:1545–1554. 2017. View Article : Google Scholar : PubMed/NCBI | |
Giguelay A, Turtoi E, Khelaf L, Tosato G, Dadi I, Chastel T, Poul MA, Pratlong M, Nicolescu S, Severac D, et al: The landscape of cancer-associated fibroblasts in colorectal cancer liver metastases. Theranostics. 12:7624–7639. 2022. View Article : Google Scholar : PubMed/NCBI | |
Li H, Liu W, Zhang X and Wang Y: Cancer-associated fibroblast-secreted collagen triple helix repeat containing-1 promotes breast cancer cell migration, invasiveness and epithelial-mesenchymal transition by activating the Wnt/β-catenin pathway. Oncol Lett. 22:8142021. | |
Avgustinova A, Iravani M, Robertson D, Fearns A, Gao Q, Klingbeil P, Hanby AM, Speirs V, Sahai E, Calvo F and Isacke CM: Tumour cell-derived Wnt7a recruits and activates fibroblasts to promote tumour aggressiveness. Nat Commun. 7:103052016. View Article : Google Scholar : PubMed/NCBI | |
Sun Y, Campisi J, Higano C, Beer TM, Porter P, Coleman I, True L and Nelson PS: Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B. Nat Med. 18:1359–1368. 2012. View Article : Google Scholar : PubMed/NCBI | |
Kramer N, Schmollerl J, Unger C, Nivarthi H, Rudisch A, Unterleuthner D, Scherzer M, Riedl A, Artaker M, Crncec I, et al: Autocrine WNT2 signaling in fibroblasts promotes colorectal cancer progression. Oncogene. 36:5460–5472. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhang C, Fei Y, Wang H, Hu S, Liu C, Hu R and Du Q: CAFs orchestrates tumor immune microenvironment-A new target in cancer therapy? Front Pharmacol. 14:11133782023. View Article : Google Scholar : PubMed/NCBI | |
Huang TX, Tan XY, Huang HS, Li YT, Liu BL, Liu KS, Chen X, Chen Z, Guan XY, Zou C and Fu L: Targeting cancer-associated fibroblast-secreted WNT2 restores dendritic cell-mediated antitumour immunity. Gut. 71:333–344. 2022. View Article : Google Scholar | |
Moynihan KD, Kumar MP, Sultan H, Pappas DC, Park T, Chin SM, Bessette P, Lan RY, Nguyen HC, Mathewson ND, et al: IL-2 targeted to CD8+ T cells promotes robust effector T cell responses and potent antitumor immunity. Cancer Discov. 14:1206–1225. 2024. View Article : Google Scholar : PubMed/NCBI | |
Siebert N, Leopold J, Zumpe M, Troschke-Meurer S, Biskupski S, Zikoridse A and Lode HN: The immunocytokine FAP-IL-2v enhances anti-neuroblastoma efficacy of the anti-GD2 antibody dinutuximab beta. Cancers. 14:48422022. View Article : Google Scholar : | |
Rivas EI, Linares J, Zwick M, Gómez-Llonin A, Guiu M, Labernadie A, Badia-Ramentol J, Lladó A, Bardia L, Pérez-Núñez I, et al: Targeted immunotherapy against distinct cancer-associated fibroblasts overcomes treatment resistance in refractory HER2+ breast tumors. Nat Commun. 13:53102022. View Article : Google Scholar : PubMed/NCBI | |
Karthik R, Deshpande NU, Iago de Castro S, Anna B, Ifeanyichukwu O, Haleh A, Andrews A, Vanessa G, Siddharth M, Samara S, et al: Abstract C021: Granulocytic MDSC-derived NLRP3 inflammasome activation is a novel regulator of inflammatory CAF skewness in pancreatic cancer. In: Proceedings of the AACR Special Conference in Cancer Research: Pancreatic Cancer; 2023 Sep 27-30; Boston, Massachusetts. Philadelphia (PA), AACR. Cancer Res. 84(Suppl 2)2024.Abstract nr C021. | |
Shao X, Zhao X, Wang B, Fan J, Wang J and An H: Tumor microenvironment targeted nano-drug delivery systems for multidrug resistant tumor therapy. Theranostics. 15:1689–1714. 2025. View Article : Google Scholar : PubMed/NCBI | |
Crane JN, Graham DS, Mona CE, Nelson SD, Samiei A, Dawson DW, Dry SM, Masri MG, Crompton JG, Benz MR, et al: Fibroblast activation protein expression in sarcomas. Sarcoma. 2023:24804932023. View Article : Google Scholar : PubMed/NCBI | |
Miettinen M: Immunohistochemistry of soft tissue tumours-review with emphasis on 10 markers. Histopathology. 64:101–118. 2014. View Article : Google Scholar | |
Zhang H, Yue X, Chen Z, Liu C, Wu W, Zhang N, Liu Z, Yang L, Jiang Q, Cheng Q, et al: Define cancer-associated fibroblasts (CAFs) in the tumor microenvironment: New opportunities in cancer immunotherapy and advances in clinical trials. Mol Cancer. 22:1592023. View Article : Google Scholar : PubMed/NCBI | |
Brahmi M, Lesluyes T, Dufresne A, Toulmonde M, Italiano A, Mir O, Le Cesne A, Valentin T, Chevreau C, Bonvalot S, et al: Expression and prognostic significance of PDGF ligands and receptors across soft tissue sarcomas. ESMO Open. 6:1000372021. View Article : Google Scholar : PubMed/NCBI | |
Robin YM, Penel N, Perot G, Neuville A, Vélasco V, Ranchère-Vince D, Terrier P and Coindre JM: Transgelin is a novel marker of smooth muscle differentiation that improves diagnostic accuracy of leiomyosarcomas: A comparative immunohistochemical reappraisal of myogenic markers in 900 soft tissue tumors. Mod Pathol. 26:502–510. 2013. View Article : Google Scholar | |
Dagher R, Cohen M, Williams G, Rothmann M, Gobburu J, Robbie G, Rahman A, Chen G, Staten A, Griebel D and Pazdur R: Approval summary: Imatinib mesylate in the treatment of metastatic and/or unresectable malignant gastrointestinal stromal tumors. Clin Cancer Res. 8:3034–3038. 2002.PubMed/NCBI | |
Tap WD, Jones RL, Van Tine BA, Chmielowski B, Elias AD, Adkins D, Agulnik M, Cooney MM, Livingston MB, Pennock G, et al: Olaratumab and doxorubicin versus doxorubicin alone for treatment of soft-tissue sarcoma: An open-label phase 1b and randomised phase 2 trial. Lancet. 388:488–497. 2016. View Article : Google Scholar : PubMed/NCBI | |
Tap WD, Wagner AJ, Schoffski P, Martin-Broto J, Krarup-Hansen A, Ganjoo KN, Yen CC, Abdul Razak AR, Spira A, Kawai A, 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. 323:1266–1276. 2020. View Article : Google Scholar : PubMed/NCBI | |
Martin-Broto J, Hindi N, Grignani G, Martinez-Trufero J, Redondo A, Valverde C, Stacchiotti S, Lopez-Pousa A, D'Ambrosio L, Gutierrez A, et al: Nivolumab and sunitinib combination in advanced soft tissue sarcomas: A multicenter, Single-arm, phase Ib/II trial. J Immunother Cancer. 8:e0015612020. View Article : Google Scholar : PubMed/NCBI | |
Shahvali S, Rahiman N, Jaafari MR and Arabi L: Targeting fibroblast activation protein (FAP): Advances in CAR-T cell, antibody, and vaccine in cancer immunotherapy. Drug Deliv Transl Res. 13:2041–2056. 2023. View Article : Google Scholar : PubMed/NCBI | |
Xiao W, Wang J, Wen X, Xu B, Que Y, Yu K, Xu L, Zhao J, Pan Q, Zhou P and Zhang X: Chimeric antigen receptor-modified T-cell therapy for platelet-derived growth factor receptor α-positive rhabdomyosarcoma. Cancer. 126(Suppl 9): S2093–S2100. 2020. View Article : Google Scholar | |
Vogt KC, Silberman PC, Lin Q, Han JE, Laflin A, Gellineau HA, Heller DA and Scheinberg DA: Microenvironment actuated CAR T cells improve solid tumor efficacy without toxicity. Sci Adv. 11:eads34032025. View Article : Google Scholar : PubMed/NCBI | |
Dharani S, Cho H, Fernandez JP, Juillerat A, Valton J, Duchateau P, Poirot L and Das S: TALEN-edited allogeneic inducible dual CAR T cells enable effective targeting of solid tumors while mitigating off-tumor toxicity. Mol Ther. 32:3915–3931. 2024. View Article : Google Scholar : PubMed/NCBI | |
Liu H, Yang W and Jiang J: Targeting tumor metabolism to augment CD8+ T cell anti-tumor immunity. J Pharm Anal. 15:1011502025. View Article : Google Scholar : | |
Ben-Ami E, Perret R, Huang Y, Courgeon F, Gokhale PC, Laroche-Clary A, Eschle BK, Velasco V, Le Loarer F, Algeo MP, et al: LRRC15 targeting in Soft-tissue sarcomas: Biological and clinical implications. Cancers (Basel). 12:572020. View Article : Google Scholar | |
Slemmons KK, Mukherjee S, Meltzer P, Purcell JW and Helman LJ: LRRC15 antibody-drug conjugates show promise as osteosarcoma therapeutics in preclinical studies. Pediatr Blood Cancer. 68:e287712021. View Article : Google Scholar | |
Brennen WN, Isaacs JT and Denmeade SR: Rationale behind targeting fibroblast activation protein-expressing carcinoma-associated fibroblasts as a novel chemotherapeutic strategy. Mol Cancer Ther. 11:257–266. 2012. View Article : Google Scholar : PubMed/NCBI | |
Giammarile F, Knoll P, Paez D, Estrada Lobato E, Calapaqui Teran AK and Delgado Bolton RC: Fibroblast activation protein inhibitor (FAPI) PET imaging in sarcomas: A new frontier in nuclear medicine. Semin Nucl Med. 54:340–344. 2024. View Article : Google Scholar : PubMed/NCBI | |
Greifenstein L, Gunkel A, Hoehne A, Osterkamp F, Smerling C, Landvogt C, Mueller C and Baum RP: 3BP-3940, a highly potent FAP-targeting peptide for theranostics-production, validation and first in human experience with Ga-68 and Lu-177. iScience. 26:1085412023. View Article : Google Scholar | |
Xie N, Shen G, Gao W, Huang Z, Huang C and Fu L: Neoantigens: Promising targets for cancer therapy. Signal Transduct Target Ther. 8:92023. View Article : Google Scholar : PubMed/NCBI | |
Kirane A, Lee D and Ariyan C: Surgical considerations in Tumor-infiltrating lymphocyte therapy: Challenges and opportunities. Transplant Cell Ther. 31(Suppl 1): S591–S598. 2025. View Article : Google Scholar : PubMed/NCBI | |
Fan S, Wang W, Che W, Xu Y, Jin C, Dong L and Xia Q: Nanomedicines targeting metabolic pathways in the tumor microenvironment: Future perspectives and the role of AI. Metabolites. 15:2012025. View Article : Google Scholar : PubMed/NCBI | |
Di Dedda C, Vignali D, Piemonti L and Monti P: Pharmacological targeting of GLUT1 to control Autoreactive T cell responses. Int J Mol Sci. 20:49622019. View Article : Google Scholar : PubMed/NCBI | |
Macintyre AN, Gerriets VA, Nichols AG, Michalek RD, Rudolph MC, Deoliveira D, Anderson SM, Abel ED, Chen BJ, Hale LP and Rathmell JC: The glucose transporter Glut1 is selectively essential for CD4 T cell activation and effector function. Cell Metab. 20:61–72. 2014. View Article : Google Scholar : PubMed/NCBI | |
Sung KE, Su X, Berthier E, Pehlke C, Friedl A and Beebe DJ: Understanding the impact of 2D and 3D fibroblast cultures on in vitro breast cancer models. PLoS One. 8:e763732013. View Article : Google Scholar : PubMed/NCBI | |
Tolle RC, Gaggioli C and Dengjel J: Three-dimensional cell culture conditions affect the proteome of cancer-associated fibroblasts. J Proteome Res. 17:2780–2789. 2018. View Article : Google Scholar : PubMed/NCBI | |
Shao H, Moller M, Wang D, Ting A, Boulina M and Liu ZJ: A Novel stromal Fibroblast-modulated 3D tumor spheroid model for studying Tumor-stroma interaction and drug discovery. J Vis Exp. Feb 28–2020. View Article : Google Scholar | |
Chen H, Cheng Y, Wang X, Wang J, Shi X, Li X, Tan W and Tan Z: 3D printed in vitro tumor tissue model of colorectal cancer. Theranostics. 10:12127–12143. 2020. View Article : Google Scholar : PubMed/NCBI | |
Mondal A, Gebeyehu A, Miranda M, Bahadur D, Patel N, Ramakrishnan S, Rishi AK and Singh M: Characterization and printability of Sodium alginate-Gelatin hydrogel for bioprinting NSCLC co-culture. Sci Rep. 9:199142019. View Article : Google Scholar | |
Kuen J, Darowski D, Kluge T and Majety M: Pancreatic cancer cell/fibroblast co-culture induces M2 like macrophages that influence therapeutic response in a 3D model. PLoS One. 12:e01820392017. View Article : Google Scholar : PubMed/NCBI | |
Jobe NP, Rosel D, Dvořánková B, Kodet O, Lacina L, Mateu R, Smetana K and Brábek J: Simultaneous blocking of IL-6 and IL-8 is sufficient to fully inhibit CAF-induced human melanoma cell invasiveness. Histochem Cell Biol. 146:205–217. 2016. View Article : Google Scholar : PubMed/NCBI | |
Balachander GM, Talukdar PM, Debnath M, Rangarajan A and Chatterjee K: Inflammatory role of Cancer-associated fibroblasts in invasive breast tumors revealed using a fibrous polymer scaffold. ACS Appl Mater Interfaces. 10:33814–33826. 2018. View Article : Google Scholar : PubMed/NCBI | |
Linxweiler J, Hajili T, Körbel C, Berchem C, Zeuschner P, Müller A, Stöckle M, Menger MD, Junker K and Saar M: Cancer-associated fibroblasts stimulate primary tumor growth and metastatic spread in an orthotopic prostate cancer xenograft model. Sci Reps. 10:125752020. View Article : Google Scholar | |
Miyazaki Y, Oda T, Inagaki Y, Kushige H, Saito Y, Mori N, Takayama Y, Kumagai Y, Mitsuyama T and Kida YS: Adipose-derived mesenchymal stem cells differentiate into heterogeneous cancer-associated fibroblasts in a stroma-rich xenograft model. Sci Rep. 11:46902021. View Article : Google Scholar : PubMed/NCBI | |
Wahbi W, Awad S, Salo T and Al-Samadi A: Stroma modulation of radiation response in head and neck squamous cell carcinoma: Insights from zebrafish larvae xenografts. Exp Cell Res. 435:1139112024. View Article : Google Scholar : PubMed/NCBI | |
Foster DS, Januszyk M, Delitto D, Yost KE, Griffin M, Guo J, Guardino N, Delitto AE, Chinta M, Burcham AR, et al: Multiomic analysis reveals conservation of cancer-associated fibroblast phenotypes across species and tissue of origin. Cancer Cell. 40:1392–1406.e7. 2022. View Article : Google Scholar : PubMed/NCBI | |
Cho C, Mukherjee R, Peck AR, Sun Y, McBrearty N, Katlinski KV, Gui J, Govindaraju PK, Puré E, Rui H and Fuchs SY: Cancer-associated fibroblasts downregulate type I interferon receptor to stimulate intratumoral stromagenesis. Oncogene. 39:6129–6137. 2020. View Article : Google Scholar : PubMed/NCBI | |
Helms EJ, Berry MW, Chaw RC, DuFort CC, Sun D, Onate MK, Oon C, Bhattacharyya S, Sanford-Crane H, Horton W, et al: Mesenchymal lineage heterogeneity underlies nonredundant functions of pancreatic Cancer-associated fibroblasts. Cancer Discov. 12:484–501. 2022. View Article : Google Scholar | |
De Vita A, Recine F, Miserocchi G, Pieri F, Spadazzi C, Cocchi C, Vanni S, Liverani C, Farnedi A, Fabbri F, et al: The potential role of the extracellular matrix in the activity of trabectedin in UPS and L-sarcoma: Evidences from a patient-derived primary culture case series in tridimensional and zebrafish models. J Exp Clin Cancer Res. 40:1652021. View Article : Google Scholar : PubMed/NCBI | |
Molina ER, Chim LK, Salazar MC, Koons GL, Menegaz BA, Ruiz-Velasco A, Lamhamedi-Cherradi SE, Vetter AM, Satish T, Cuglievan B, et al: 3D Tissue-engineered tumor model for Ewing's sarcoma that incorporates Bone-like ECM and mineralization. ACS Biomater Sci Eng. 6:539–552. 2020. View Article : Google Scholar | |
Moghimi N, Hosseini SA, Dalan AB, Mohammadrezaei D, Goldman A and Kohandel M: Controlled tumor heterogeneity in a co-culture system by 3D bio-printed tumor-on-chip model. Sci Rep. 13:136482023. View Article : Google Scholar : PubMed/NCBI | |
Yang D, Jones MG, Naranjo S, Rideout WM III, Min KHJ, Ho R, Wu W, Replogle JM, Page JL, Quinn JJ, et al: Lineage tracing reveals the phylodynamics, plasticity, and paths of tumor evolution. Cell. 185:1905–1923.e25. 2022. View Article : Google Scholar : PubMed/NCBI | |
Simeonov KP, Byrns CN, Clark ML, Norgard RJ, Martin B, Stanger BZ, Shendure J, McKenna A and Lengner CJ: Single-cell lineage tracing of metastatic cancer reveals selection of hybrid EMT states. Cancer Cell. 39:1150–1162.e9. 2021. View Article : Google Scholar : PubMed/NCBI | |
Liu J, Liu C, Ma Y, Pan X, Chu R, Yao S, Chen J, Liu C, Chen Z, Sheng C, et al: STING inhibitors sensitize platinum chemotherapy in ovarian cancer by inhibiting the CGAS-STING pathway in cancer-associated fibroblasts (CAFs). Cancer Lett. 588:2167002024. View Article : Google Scholar : PubMed/NCBI | |
de Kruijf EM, van Nes JG, van de Velde CJ, Putter H, Smit VT, Liefers GJ, Kuppen PJ, Tollenaar RA and Mesker WE: Tumor-stroma ratio in the primary tumor is a prognostic factor in early breast cancer patients, especially in triple-negative carcinoma patients. Breast Cancer Res Treat. 125:687–696. 2011. View Article : Google Scholar | |
Huijbers A, Tollenaar RA, v Pelt GW, Zeestraten EC, Dutton S, McConkey CC, Domingo E, Smit VT, Midgley R, Warren BF, et al: The proportion of tumor-stroma as a strong prognosticator for stage II and III colon cancer patients: Validation in the VICTOR trial. Ann Oncol. 24:179–185. 2013. View Article : Google Scholar | |
De Vlieghere E, Verset L, Demetter P, Bracke M and De Wever O: Cancer-associated fibroblasts as target and tool in cancer therapeutics and diagnostics. Virchows Arch. 467:367–382. 2015. View Article : Google Scholar : PubMed/NCBI | |
Kanzaki R and Pietras K: Heterogeneity of cancer-associated fibroblasts: Opportunities for precision medicine. Cancer Sci. 111:2708–2717. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wang Q, Zhang X, Du K, Wu X, Zhou Y, Chen D and Zeng L: Machine learning identifies characteristics molecules of cancer associated fibroblasts significantly correlated with the prognosis, immunotherapy response and immune microenvironment in lung adenocarcinoma. Front Oncol. 12:10592532022. View Article : Google Scholar : PubMed/NCBI | |
Shen C, Rawal S, Brown R, Zhou H, Agarwal A, Watson MA, Cote RJ and Yang C: Automatic detection of circulating tumor cells and cancer associated fibroblasts using deep learning. Sci Rep. 13:57082023. View Article : Google Scholar : PubMed/NCBI | |
Agnoletto C, Caruso C and Garofalo C: Heterogeneous circulating tumor cells in sarcoma: Implication for clinical practice. Cancers (Basel). 13:21892021. View Article : Google Scholar : PubMed/NCBI | |
Yang Z, Zhou D and Huang J: Identifying explainable machine learning models and a novel SFRP2+ fibroblast signature as predictors for precision medicine in ovarian cancer. Int J Mol Sci. 24:169422023. View Article : Google Scholar : | |
Min KW, Kim DH, Noh YK, Son BK, Kwon MJ and Moon JY: Cancer-associated fibroblasts are associated with poor prognosis in solid type of lung adenocarcinoma in a machine learning analysis. Sci Rep. 11:167792021. View Article : Google Scholar : PubMed/NCBI | |
Huang B, Chen Q, Ye Z, Zeng L, Huang C, Xie Y, Zhang R and Shen H: Construction of a matrix Cancer-associated fibroblast signature Gene-based risk prognostic signature for directing immunotherapy in patients with breast cancer using Single-cell analysis and machine learning. Int J Mol Sci. 24:131752023. View Article : Google Scholar : PubMed/NCBI | |
Ao Z, Shah SH, Machlin LM, Parajuli R, Miller PC, Rawal S, Williams AJ, Cote RJ, Lippman ME, Datar RH and El-Ashry D: Identification of Cancer-associated fibroblasts in circulating blood from patients with metastatic breast cancer. Cancer Res. 75:4681–4687. 2015. View Article : Google Scholar : PubMed/NCBI | |
Booijink R, Terstappen LWMM, Dathathri E, Isebia K, Kraan J, Martens J and Bansal R: Identification of functional and diverse circulating cancer-associated fibroblasts in metastatic Castration-naive prostate cancer patients. Mol Oncol. 19:2074–2091. 2024. View Article : Google Scholar | |
Lu T, Oomens L, Terstappen L and Prakash J: In vivo detection of circulating cancer-associated fibroblasts in breast tumor mouse xenograft: Impact of tumor stroma and chemotherapy. Cancers (Basel). 15:11272023. View Article : Google Scholar : PubMed/NCBI |