
Role of exosomal non‑coding RNAs in cancer‑associated fibroblast‑mediated therapy resistance (Review)
- Authors:
- Published online on: July 11, 2025 https://doi.org/10.3892/ijo.2025.5774
- Article Number: 68
-
Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Cancer is the second leading cause of global mortality and a major modern public health issue. In 2022, ~20 million new cases and 9.7 million cancer-related deaths were reported worldwide (1). Drug resistance remains a major obstacle to extending the overall survival of patients with cancer, contributing to >90% of cancer mortality (2,3). The mechanisms underlying drug resistance include the inhibition of programmed cell death, aberrant metabolism, immune evasion and cancer stem cell (CSC) maintenance. These processes are driven by multiple factors, such as hypoxia and epigenetic regulation (4-8). While previous research has primarily focused on elucidating epigenetic genes and signaling pathway dysregulation in cancer cells (9,10), evidence has highlighted the critical role of the tumor microenvironment (TME) in drug resistance (11,12). The TME is highly complex, consisting of cancer, immune, stromal and endothelial cells, pericytes, and the extracellular matrix (ECM) (13). Cancer-associated fibroblasts (CAFs) are essential stromal cells within the TME, and regulate the TME through paracrine signaling, including the secretion of cytokines, chemokines, growth factors and exosomes, leading to the remodeling of the TME (14,15). This process promotes connective tissue proliferation, angiogenesis and metabolic processes, and intricately interacts with the signaling pathways of cancer cells, resulting in drug resistance (16,17). Furthermore, CAFs induce the formation of immune barriers by influencing the activity of immune cells directly and upregulating immune checkpoint molecules on the cancer cell surface (18,19).
Exosomes, which are membrane vesicles formed by the shedding of the cytoplasmic membrane, serve as crucial mediators for intercellular communication between CAFs and other cells within the TME (20). Non-coding RNAs (ncRNAs), such as long non-coding RNAs (lncRNAs), circular RNAs (circRNAs/circs) and microRNAs (miRNAs/miRs), are non-protein-coding transcripts and are evolutionarily conserved functional components of exosomes (21,22). miRNAs (17-25 nucleotides) bind to the 3′ untranslated region (UTR) of the target mRNA via the seed sequence, leading to translational repression or mRNA degradation (23). lncRNAs (>200 nucleotides) and circRNAs (covalently closed loop) regulate gene expression by interacting with transcription factors (TFs), RNA binding proteins (RBPs) or miRNAs (24,25). Ample evidence has demonstrated that these ncRNAs critically regulate cellular physiological processes and the pathogenesis of diseases, including cancer (21-25).
Recent studies have shown that exosomal ncRNAs act as key regulators, mediating crosstalk between tumor cells and CAFs, tumor-associated macrophages (TAMs), T cells, neutrophils and other cells to regulate tumor growth, metastasis, angiogenesis, immune responses and drug resistance (26-30). For example, M2 polarization of macrophages promotes tumor progression, and exosomal ncRNAs recruit macrophages in the TME and induce M2 polarization by targeting signaling pathways such as the NF-κB or STAT3 signaling pathways (31-33). Exosomal ncRNAs also serve a crucial role in the communication between CAFs and the TME, whereby ncRNAs can alter the phenotype of CAFs. In non-small cell lung cancer (NSCLC), circNOX4 promotes the production of IL-6 via the miR-329-5p/fibroblast-associated protein (FAP) axis, inducing the transition of normal fibroblasts (NFs) to the inflammatory CAF (iCAF) phenotype (34). Furthermore, several publications have demonstrated that CAF-derived exosomes promote the growth and metastasis of colorectal cancer (CRC), head and neck cancer, breast cancer (BC) and melanoma, and are involved in the development of drug resistance (35-38). Recent studies have shown that miRNAs derived from CAFs decrease lipid reactive oxygen species (ROS) levels and inhibit ferroptosis in cancer cells by targeting ChaC glutathione specific γ-glutamyl cyclotransferase 1 and long-chain acyl-CoA synthase 4 (ACSL4), leading to chemotherapy resistance (39,40). Additionally, ncRNAs secreted by CAFs induce stemness in bladder cancer cells by activating the STAT3 and mTOR signaling pathways, thereby creating supportive regions or conditions required for CAFs to exert their biological functions for chemoresistance in bladder cancer (41). Although these studies have revealed the potential role of exosomal ncRNAs in CAF-mediated tumor resistance, there is a lack of systematic elucidation of their core molecular mechanisms and clinical application potential across different tumor types, making it difficult for researchers to achieve a comprehensive understanding. The present review highlights the pivotal role of CAFs in cancer drug resistance. Based on current evidence, to the best of our knowledge, the present review represents the first systematic elaboration of the potential mechanisms through which exosomal ncRNAs mediate resistance to cancer therapies via bidirectional regulation between CAFs and tumor cells. Furthermore, the present review discusses promising research avenues and future directions. Notably, the present review discusses the clinical applications and challenges of exosomal ncRNAs as mediators of CAF-driven cancer resistance, and that targeting CAF-associated exosomal ncRNAs to modify the drug resistance-associated TME holds notable promise for overcoming resistance to chemotherapy, targeted therapy and immunotherapy. This approach thereby offers innovative strategies for the treatment of patients with therapy-resistant cancer in the future.
Origin, heterogeneity and function of CAFs
CAFs are typically elongated cells that lack the mutations commonly found in cancer cells and do not express markers indicating epithelial, endothelial or leukocyte lineages (42). CAFs interact with other cells in the TME through the secretion of metabolites, cytokines, chemokines and exosomes (43), serving a role in tumor cell proliferation and metastasis (44). CAFs are abundantly present in the TME of almost all solid tumors, actively participating in tumor growth and metastasis (42). This section focuses on the heterogeneity and plasticity of CAFs, and discusses their role in cancer (Fig. 1).
Origin, heterogeneity and plasticity of CAFs
The current consensus is that CAFs originate from locally resident fibroblasts activated by various signaling molecules in the TME (42,45). These signaling molecules include TGF-β, hepatocyte growth factor (HGF), platelet-derived growth factor (PDGF), fibroblast growth factor 2, stromal derived factor-1 (SDF-1) and ROS (18). Wu et al (16) summarized the signaling pathways that promote the transformation of fibroblasts into CAFs, including the TGF-β, PI3K/AKT/mTOR and MAPK pathways. Additionally, substantial evidence suggests that CAFs can also differentiate from other cell types. Mesenchymal stem cells (MSCs) can differentiate into CAFs. In BC, bone marrow-derived MSCs are recruited to the TME and differentiate into PDGF receptor (PDGFR) α− CAFs (46,47). Furthermore, endothelial cells (48), epithelial cells (49), adipocytes (50), smooth muscle cells (51) and pericytes (52) are also potential sources of CAFs.
CAFs from different origins exhibit distinct functional roles in tumors. For instance, CAFs differentiated from bone marrow MSCs promote the growth and doxorubicin resistance of acute lymphoblastic leukemia by upregulating TGF-β (53). Endothelial-like and adipocyte-differentiated CAFs enhance cancer invasiveness and metastasis (54,55), whereas mesothelial cell-derived antigen-presenting CAFs induce the differentiation of CD4+ T cells into regulatory T cells (56). These diverse functions highlight the heterogeneity of CAFs. The heterogeneity of CAFs is a hot research topic, and various experts in this field have provided overviews of CAF heterogeneity, aiming to distinguish subtypes of CAFs with different functions (57-59). Based on their functions, CAFs express various markers such as PDGFRα/β, α smooth muscle actin (α-SMA), FAP and fibroblast-specific protein-1 (60). However, it is difficult to establish a unified standard for CAF subtypes. On one hand, CAFs lack specific surface markers (61). α-SMA is widely expressed in smooth muscle cells (62), while FAP can also be detected in macrophages (63). On the other hand, the markers of CAFs lack consistent expression patterns, leading to a vast number of possible marker combinations (64). The consensus is that there are two CAF subtypes: Myofibroblastic CAFs (myCAFs) and iCAFs (42). myCAFs are characterized by the expression of collagen genes and ECM proteins, participating in matrix remodeling and cellular invasion. iCAFs exhibit high expression of cytokine and chemokine genes, promoting a local inflammatory environment in tumors (65,66). In pancreatic cancer, myCAFs with high expression levels of α-SMA and MMP11 are located closer to cancer cells and exhibit a contractile phenotype that produces the ECM, while iCAFs, which are located away from cancer cells, express IL-6 and angiotensin II type 1 receptor, and serve a key role in shaping an immunosuppressive microenvironment (67,68). The heterogeneity of iCAFs and myCAFs is illustrated in Fig. 2 (42,65-68). In recent years, with the application of single-cell and spatial transcriptomics, the heterogeneity of CAFs has been further untangled (69,70). CAFs can be further classified into multiple distinct subtypes based on their differential surface markers. Although the categorization of these subtypes has not yet achieved widespread acceptance, it has deepened the understanding of CAF heterogeneity. For instance, Cords et al (71) classified CAFs into eight categories through pan-cancer single-cell sequencing: myCAFs (high expression of MMP11 and collagen type I α2 chain), iCAFs (high expression of IL-6 and CD34), vascular CAFs (high expression of NOTCH3, collagen type XVIII α1 chain and melanoma cell adhesion molecule), tumor-like CAFs (expressing VEGFA, CD10 and transmembrane protein 158), antigen-presenting CAFs [expressing human leucocyte antigen (HLA)-DRA, HLA-DRB1 and CD74], and the relatively rare interferon-responsive CAFs, reticular-like CAFs and dividing CAFs.
The goal of antitumor therapy based on targeting of CAFs is to reduce the CAFs that promote tumorigenic phenotypes. Therefore, researchers have attempted to determine whether CAF-related molecules can be targeted to transform the phenotype of CAFs (57). Hypoxia-induced hypoxia-inducible factor 1-α (HIF-1α) promotes an increase in the number of α-SMA−/α-SMA-low iCAFs in pancreatic ductal adenocarcinoma (PDAC), while the differentiation of α-SMA+ myCAFs relies on HIF-2α (72). TGF-β inhibits the activation of the janus kinase (JAK)/STAT signaling pathway by suppressing the expression of IL-1 receptor type 1, thereby inhibiting the differentiation of iCAFs (73). These results suggest the plasticity of CAFs.
Role of CAFs in tumor development and progression
CAFs exert a marked effect on various aspects of the TME through the release of exosomes, cytokines, inflammatory ligands and ECM proteins. These actions ultimately manifest as proliferation, invasion and drug resistance in tumor cells (14,15).
The ECM is an important component of the TME and is considered to be a key regulatory factor in promoting tumor malignancy and invasion (74). The ECM is a non-cellular component present in all tissues, composed of collagen, fibronectin, laminin, glycosaminoglycans and proteoglycans (75). The ECM also interacts with factors such as EGF, TGF-β, HGF and VEGF, participating in tumor growth and angiogenesis (76). One of the most prominent abilities of CAFs is the remodeling of the ECM, which is attributed to their high expression of MMP (34), transglutaminase, lysyl oxidase and integrin α11β1 (77,78). ECM remodeling refers to the alteration of the synthesis, degradation and components of the ECM. CAFs alter the density and composition of the ECM, leading to its stiffening and degradation (79). In the TME, the binding of TGF-β to CAF surface receptors activates Smad2/3 and collagen type I α1 chain, thereby enhancing CAF synthesis of collagen, leading to collagen deposition (80). Additionally, the crosslinking enzymes produced by CAFs, in conjunction with force-mediated ECM remodeling, further increase the stiffness of the ECM (42). Higher ECM stiffness is associated with greater tumor invasiveness (81) as it promotes the release of the TF twist family bHLH transcription factor 1 (TWIST1) from the cytoplasmic binding protein G3BP stress granule assembly factor 2 (G3BP2) into the nucleus. The absence of G3BP2 can result in sustained nuclear localization of TWIST1, which, together with increased matrix stiffness, induces epithelial-mesenchymal transition (EMT) (82). Additionally, the stiffening of the ECM can affect the ability of drugs to penetrate cancer cells (83). In addition to increasing ECM stiffness, the MMPs secreted by CAFs lead to ECM degradation, creating pathways within the ECM that promote cancer cell proliferation and invasion, while also providing space for abundant angiogenesis (84-86). Furthermore, the ECM remodeling induced by CAFs maintains an inflammatory state in the TME and prolongs the release of proinflammatory factors such as IL-6, IL-8 and leukemia inhibiting factor (LIF) from cancer cells (87,88). ECM remodeling also affects the migration of infiltrating leukocytes, contributing to immune evasion by cancer cells (89).
CAFs secrete various cytokines, such as TGF-β, growth differentiation factor and IL-11, and activate the P38/MAPK, Smad and STAT3 signaling pathways on the surface of tumor cells to promote tumor cell proliferation and invasion (16). Tumor-promoting CAFs highly express integrin subunit β2, which activates the PI3K/AKT/mTOR signaling pathway through the induction of NADH oxidation, thereby facilitating cancer cell proliferation (90). CAFs also secrete exosomes carrying ncRNAs, which serve a profound role in the communication between CAFs and cancer cells by targeting multiple physiologically active mRNAs in cancer cells (91). In addition, CAFs are the primary source of the chemokine C-X-C motif chemokine ligand 12 (CXCL12) in the TME. The binding of CXCL12 to C-X-C motif chemokine receptor 4 (CXCR4) induces an increase in the number of regulatory T cells and a decrease in the number of CD8+ T cells, thereby suppressing the immune microenvironment (92,93). Simultaneously, CXCL12 impedes the recognition of cancer cells by anti-cytotoxic T lymphocyte-associated protein 4 and α-programmed cell death 1 ligand 1, thereby inducing tumor immune evasion (94). Additionally, periostin-positive myCAFs induce macrophage secreted phosphoprotein 1 expression through the IL-6/STAT3 axis and hinder effective T-cell infiltration and the immune response (95,96). Furthermore, crosstalk between CAFs and cancer cells is one of the main mechanisms of cancer metabolic reprogramming. CAF-derived exosomes and metabolic factors such as HIF-1α regulate glycolysis, and amino acid and lipid metabolism in cancer cells by modulating intracellular metabolic pathways (97). This process contributes to cancer growth, metastasis and drug resistance (97). The metabolic secretions of CAFs themselves also mediate tumor development. For instance, lactate produced by CAFs can enhance the expression of discs large homolog 5 in oral squamous cell carcinoma (OSCC) and activate the Hippo signaling pathway, promoting the stem cell properties of OSCC (98). CAF-derived amino acids provide essential nutrients and energy for cancer growth and invasion (99).
In addition, ECM synthesis and remodeling generate solid stress, leading to the compression of blood and lymphatic vessels. This exacerbates hypoxia in the TME, which promotes angiogenesis and transformation through the formation of new blood vessels (100,101). CAFs also secrete various angiogenic molecules, such as VEGF, PDGF, CXCL12 or HGF, to induce angiogenesis in the TME (102,103). The angiogenesis induced by CAFs not only increases the invasive capacity of cancer cells but also represents one of the contributing factors underlying the resistance of cancer cells to anti-EGFR drugs (104).
Exosomal ncRNAs from tumor cells regulate the activation and metabolism of CAFs
Under the stimulation of various factors in the TME, NFs undergo transformation into CAFs, acquiring enhanced proliferative, invasive and metabolic capabilities (105). Notably, this transition equips CAFs with functions that allow them to participate in tumor progression (45). Factors involved in the activation of CAFs include TGF-β, fibroblast growth factor, PDGF, Sonic hedgehog, IL-6, IL-1R and ncRNAs (16,91). These molecular factors activate various signaling pathways within the CAFs, such as the JAK/STAT and EGFR/erb-b2 receptor tyrosine kinase 2 signaling pathways, thereby shaping the heterogeneity of the influence of CAFs on tumor progression (73,106,107). Recent evidence suggests that dysregulation of ncRNAs within CAFs may represent an important mechanism mediating changes in their functional phenotype (108). Biologically functional ncRNAs are abundantly present in exosomes. They are transferred between cells via exosomes and serve a crucial role in cancer growth and drug resistance (109). Notably, CAF-mediated drug resistance often occurs following ncRNA dysregulation. For example, miR-9 secreted by triple-negative BC cells induces the differentiation of tumor-promoting CAFs by targeting EGF containing fibulin extracellular matrix protein 1 (110). miRNAs originating from cancer cells can also stimulate the expression of SDF-1, MMP3, MMP9, PDGF and chemokine CC motif ligand 7 by CAFs (111,112). These findings indicate that the activation of CAFs by exosomal ncRNAs induces drug resistance. Therefore, when discussing the role of ncRNAs in CAF-mediated drug resistance, it is essential to consider the activating effects of exosomal ncRNAs on CAFs (Fig. 3).
Exosomal ncRNAs activate CAFs
Numerous studies have demonstrated that ncRNAs derived from cancer cells serve as critical regulators mediating the differentiation of NFs into CAFs. Treatment of NFs with gastric cancer (GC)-derived exosomes enhances their proliferative capacity and the expression of α-SMA and Vimentin (113). The expression of long intergenic non-protein coding RNA 691 (LINC00691) is increased in NFs, augmenting the ability of NFs to promote cancer cell proliferation and invasion. Subsequent experiments have demonstrated that LINC00691 promotes the differentiation of NFs into CAFs by activating the JAK2/STAT3 signaling pathway (113). Pang et al (114) isolated primary pancreatic fibroblasts from C57 mice and co-cultured them with the BxPC-3 and SW1990 pancreatic cancer cell lines. The results indicated that miR-155 secreted by pancreatic cancer cells in exosomes promoted the transformation of NFs into α-SMA+ and FAP+ CAFs by targeting tumor protein P53 induced nuclear protein 1 (TP53INP1). TP53INP1 is an important functional component of p53, and its inhibition leads to the inactivation of p53, resulting in the activation of CAFs (115,116). Based on clinical data, there is a positive association between exosomal miR-1247-3p and the malignancy and metastasis of hepatocellular carcinoma (HCC). This is due to the fact that exosomal miR-1247-3p secreted by HCC cells targets β-1,4-galactosyltransferase 3, facilitating the differentiation of CAFs and activating their β1-integrin-NF-κB signaling pathway (117). Activated CAFs secrete IL-6 and IL-8, which contribute to the progression of HCC (117). Notably, another study found that miR-21 secreted by HCC cells activated the pyruvate dehydrogenase kinase 1/AKT signaling pathway by targeting PTEN. This process induced the transformation of hepatic stellate cells into CAFs (118). Exosomal miR-21 has also been demonstrated to serve a role in head and neck squamous cell carcinoma (HNSCC); it induces the differentiation of NFs into CAFs by targeting YOD1, thereby promoting lymph node metastasis in HNSCC (119). The lncRNA Gm26809 and miR-211 are upregulated in exosomes from melanoma cells, inducing the formation of CAFs and increasing the expression of CAF markers (α-SMA and FAP) (120). Furthermore, miR-211 targets insulin-like growth factor 2 receptor and leads to the activation of the MAPK signaling pathway in CAFs, which results in increased proliferation and migration, and upregulation of pro-inflammatory genes in CAFs (121). BC cell-secreted exosomes contain various miRNAs, including miR-125b, miR-146a and miR-9, which are transferred to NFs and promote their differentiation into the CAF phenotype (122-124). For example, miR-125b, as an inhibitor of TP53INP1, elevates the expression of α-SMA, MMP and cytokines in NFs (122). Furthermore, NFs differentiate into CAFs following miR-9 uptake, enhancing their proliferation and migration. These CAFs also secrete miR-9, which targets BC cells, forming a positive feedback loop. The miR-9 secreted by CAFs promotes BC metastasis by reducing E-cadherin expression (124). The specific mechanisms of exosomal ncRNAs regulating CAF activation are described in Table I. Activated by these cancer cell-derived ncRNAs, CAFs secrete abundant angiogenic factors, IL-6, TGF-β, TNF-α and CXCL12, which act on cancer cells to promote their proliferation, invasion and drug resistance (118,125,126).
Exosomal ncRNAs induce the glycolysis of CAFs
CAFs maintain their metabolic characteristics of producing lactate and pyruvate through the epigenetic regulation of HIF-1α and glycolytic enzymes (127). This phenomenon is termed the 'reverse Warburg effect' in CAFs, where myCAFs shift to aerobic glycolysis, generating copious amounts of lactate and pyruvate (128). The lactate produced by CAFs is taken up and metabolized by cancer cells to generate NADH, ultimately producing ATP to support cancer cell proliferation and invasion (90). On the other hand, the lactate derived from glycolysis in cancer cells within the TME forms a positive feedback loop with CAFs. The lactate released by cancer cells stimulates the upregulation of IL-6 and collagen triple helix repeat-containing 1 expression in CAFs, which further enhances glycolysis in cancer cells and suppresses the immune microenvironment (129,130). A previous study has reported the role of tumor cell-secreted exosomal ncRNAs in the metabolic reprogramming of CAFs (Table I). In BC, secreted miR-105 induces the metabolic reprogramming of CAFs by targeting the Myc signaling pathway. When nutrients are sufficient, miR-105-reprogrammed CAFs enhance glucose and glutamine metabolism to fuel cancer cells. Under conditions of nutrient deprivation and accumulation of metabolic byproducts, CAFs convert metabolic waste such as lactate and ammonia into high-energy metabolites for detoxification (131). Recently, He et al (132) identified that circABCC4 induces glycolysis in CAFs by enhancing the interaction between pyruvate kinase M1/2 and karyopherin subunit α2, thereby promoting increased IL-8 secretion by CAFs and leading to oxaliplatin resistance in pancreatic cancer. However, to the best of our knowledge, existing evidence has not confirmed whether circABCC4 is delivered to CAFs via exosomes derived from pancreatic cancer cells (132).
CAF-derived exosomal ncRNAs promote drug resistance in cancer
The development of drug resistance in cancer is not only determined by intrinsic factors but also influenced by the complex regulatory network among cells in the TME. CAFs are the most crucial cells involved in the interaction between tumor cells and the stroma, and targeted therapy against CAFs holds great promise for improving cancer drug resistance (133). The use of chemotherapy drugs induces the transformation of NFs into CAFs, which secrete large quantities of cytokines and exosomes (134). It has been suggested that CAF-derived exosomes are ingested by cancer cells, mediating drug resistance in cancer cells by activating drug resistance-related pathways, regulating programmed cell death, EMT, drug transporters and immune evasion (135). These mechanisms are described in Table II and Fig. 4.
Drug resistance-related signaling pathways
In cancer cells, the dysregulation and mutation of key nodal molecules in signaling pathways lead to the aberrant activation of multiple pathways, which form intricate cross-regulatory networks collectively contributing to the development of tumor drug resistance (136). For instance, TGF-β not only induces EMT in cancer cells by activating the core effector Smad, but also promotes PI3K/AKT activation by increasing transforming growth factor β receptor type I kinase activity or suppressing PTEN (137), while the PI3K/AKT pathway further reinforces Wnt signaling by phosphorylating GSK-3β to inhibit β-catenin degradation (138,139). AKT phosphorylates salt-inducible kinase 1, relieving its suppression of STAT3 (140), thereby enhancing JAK/STAT-mediated transcriptional activation of anti-apoptotic and proliferative genes (141). There exists a feedback activation mechanism in several signaling pathways, such as the AKT, ERK and STAT3 signaling pathways. When a single signaling pathway is inhibited, the crosstalk of other signaling pathways activates feedback, further driving the adaptive resistance of cancer cells (142). Functionally, these signaling pathways serve a synergistic role in the formation of tumor resistance by regulating the EMT, the maintenance of CSC stemness and metabolic reprogramming (143,144). There is an interplay of signaling pathways between CAFs and cancer cells (Fig. 5) (144). As crucial players in the regulation of cellular signaling pathways (145), ncRNAs exert critical roles in this regulatory process, which is described in detail subsequently.
Wnt/β-catenin
The Wnt/β-catenin pathway is a classic signaling cascade that drives cancer growth, metastasis and drug resistance in various malignancies, such as GC, HCC and BC (146). In the absence of Wnt ligands, GSK3β, Axin and adenomatous polyposis coli (APC) form a complex, promoting the phosphorylation of β-catenin. When Wnt protein ligands are present, they recruit cytoplasmic Dishevelled proteins to disrupt the Axin/GSK3β/APC complex, thereby rescuing β-catenin from inhibition. Subsequently, accumulated β-catenin enters the nucleus and binds to the TFs T-cell factor (TCF) and lymphoid enhancer factor 1, initiating the expression of a series of oncogenes (147). Activation of the Wnt/β-catenin pathway promotes the expression of glutathione (GSH) peroxidase 4 (GPX4) by forming a β-catenin/TCF4 transcriptional complex, thereby reducing lipid ROS production and drug-induced ferroptosis in GC cells (148). In the TME, HGF secreted by myCAFs enhances the cloning ability and tumorigenicity of colon CSCs by activating the Wnt/β-catenin signaling pathway (149).
Studies have shown that CAF-derived exosomal ncRNAs are also important activators of the Wnt/β-catenin pathway. Ren et al (150) revealed upregulated expression of lncRNA H19 in colitis-associated cancer (CAC). The authors further confirmed that CAF (specifically expressing FAP, ferroptosis suppressor protein 1, ACTA2 and CD90)-derived exosomes were the source of H19 in CAC cells. Overexpression of H19 promoted the proliferation, migration and invasion of CAC cells both in vivo and in vitro. Additionally, H19 increased the expression of CSC markers (Nanog, Oct4 and Sox2) and oxaliplatin resistance of CAC cells. Mechanistically, H19 activated the β-catenin signaling pathway by sponging miR-141, an inhibitor of β-catenin (150). Similarly, lncRNA colorectal cancer-associated lncRNA (CCAL) is transferred between CAFs and CRC cells via exosomes. Upon entering cancer cells, CCAL activates β-catenin by directly interacting with the mRNA-stabilizing protein human antigen R, inducing drug resistance in CRC (151). CAF-derived exosomal miR-148b-3p is a crucial factor driving bladder cancer development. In a xenograft mouse model, injection of CAF-exosomes into mice enhanced their resistance to docetaxel combined with paclitaxel, while the use of a miR-148b-3p inhibitor restored chemotherapy sensitivity (152). PTEN, a direct target of miR-148b-3p, reduces nuclear β-catenin levels by inhibiting AKT phosphorylation (153). Mechanistically, miR-148b-3p promoted the proliferation and EMT of bladder cancer cells through PTEN upregulation and suppression of the β-catenin pathway, thus endowing bladder cancer with drug resistance (152). In cervical cancer (CCa), miR-1323 is secreted by CAFs and is upregulated in CCa cells. Through functional assays, miR-1323 has been demonstrated to promote cell proliferation, migration, invasion and radiation resistance. Further investigation revealed that the effect of miR-1323 was achieved by targeting poly(A) binding protein nuclear 1 (PABPN1). Additionally, PABPN1 can recruit insulin-like growth factor 2 mRNA-binding protein 1 to further regulate GSK-3β, thereby influencing the activity of the WNT/β-catenin signaling pathway (154).
STAT3
The STAT family is one of the most important TF families in tumors, comprising STAT1-6, which share similar structures and exhibit a high degree of conservation (155). Among the STAT family, STAT3 is the most extensively studied member, and serves a pivotal role in tumor cell proliferation, invasion, stem cell characteristics and drug resistance (156). Previous research has indicated that the activation of STAT3 is a crucial pathway for CAFs to exert their effects in cancer progression. CAFs secrete IL-6 to activate the JAK/STAT3 signaling pathway in gallbladder cancer cells, promoting the metastasis of gallbladder cancer. Additionally, the increased number of CAFs is associated with poor prognosis in patients with gallbladder cancer (157). IL-10 derived from CD146+ CAFs facilitates angiogenesis and tumor growth in endometrial cancer via the JAK1/STAT3 axis (158). Cisplatin induces CAFs to produce more IL-11, which activates STAT3 through IL-11 receptor α, thereby enabling NSCLC cells to acquire cisplatin resistance (159). The binding of LIF to the LIF receptor also constitutes an important pathway for activating STAT3 and inducing cancer cell stemness (160). A previous study has revealed that circFARP1 upregulates LIF expression by sponging miR-660-3p and interacts with caveolin 1 to increase LIF secretion. The LIF secreted by CAFs activates STAT3 in pancreatic cancer cells, thereby reducing their sensitivity to gemcitabine (161).
Similarly, exosomal ncRNAs derived from CAFs promote cancer drug resistance by upregulating STAT3. miR-146a-5p expression is elevated in CAFs and miR-146a-5p enters bladder cancer cells via exosomes, and can target and inhibit the expression of AT-rich interactive domain 1A (ARID1A) (41). By targeting ARID1A to relieve the inhibitory effect of suppressor of cytokine signaling 1 on STAT3, miR-146a-5p indirectly activates STAT3, thereby promoting stem cell characteristics and chemotherapy resistance in bladder cancer. Furthermore, high levels of miR-146a-5p are positively associated with bladder cancer staging and recurrence risk (41). Sun et al (162) demonstrated that CAF-derived miR-296-3p in ovarian cancer promotes the proliferation, invasion and migration of ovarian cancer cells, and induces paclitaxel resistance. By analyzing the expression of circulating miR-296-3p in the blood, the chemotherapy sensitivity of patients with ovarian cancer can be predicted. Mechanistically, miR-296-3p directly targets suppressor of cytokine signaling 6 (SOCS6), an inhibitor of STAT3, thereby activating the STAT3 signaling pathway in ovarian cancer cells. Overexpression of SOCS6 can inhibit the activation of STAT3, thereby reversing the drug resistance in cancer cells induced by the elevation of miR-296-3p (162). Zhao et al (163) found that monocytic myeloid-derived suppressor cells (M-MDSCs) contribute to cisplatin resistance in esophageal squamous cell carcinoma (ESCC). CAF-secreted IL-6 and miR-21 synergistically induce the high immunosuppressive function of M-MDSCs. miR-21 promotes tumor α-SMA and CD11b expression, facilitates M-MDSC accumulation and is delivered via exosomes to induce M-MDSC generation (163). Substantial evidence has conclusively demonstrated that PTEN serves as a direct target of miR-21 (118,164,165). Similarly, the study by Zhao et al (163) demonstrated that miR-21 overexpression in M-MDSCs indirectly activated STAT3 by suppressing PTEN, thereby inducing MDSC generation and IL-6 autocrine signaling. Therefore, inhibiting STAT3 could restore cisplatin resistance induced by IL-6 and exosomal miR-21.
PI3K/AKT
The PI3K/AKT signaling pathway serves a crucial role in promoting cancer by regulating cancer cell survival and metabolism. PI3K phosphorylates phosphatidylinositol-4,5-bisphosphate to generate phosphatidylinositol-3,4,5-trisphosphate (PIP3). Subsequently, PIP3 recruits AKT to the plasma membrane. Once AKT is activated, it phosphorylates various substrates (166). mTOR, a common downstream effector of AKT, drives cancer progression by phosphorylating S6 kinase 1 and eIF-4E binding protein 1, and activates HIF-1α and LIPIN1 signaling to mediate cellular glycolysis and lipid metabolism (167).
Certain ncRNAs indirectly activate PI3K/AKT by targeting its upstream regulators. PTEN, a phosphoinositide phosphatase, dephosphorylates phosphatidylinositols (PtdIns) (3-5) P3 at the D3 position of the inositol ring to generate PtdIns (4,5) P2, serving as a direct antagonist of PI3K (168). CAF-derived miR-20a is transferred to cancer cells and upregulates the PI3K/AKT signaling pathway by inhibiting PTEN, thereby enhancing cisplatin resistance in NSCLC. Overexpression of PTEN reverses the upregulation of miR-20a in the PI3K/AKT pathway, inhibiting cell proliferation and cisplatin resistance, while promoting apoptosis (169). Qu et al (170) found that CAF-exosome circN4BP2L2 directly binds to eukaryotic initiation factor 4A (EIF4A3). EIF4A3 is an RNA-binding protein that is involved in the binding and subcellular localization of circRNAs. Studies have reported that EIF4A3 activates the PI3K/AKT signaling pathway through a competing endogenous RNA mechanism after binding with circRNA (171,172). However, the specific mechanisms of circN4BP2L2 in the study by Qu et al (170) have not been thoroughly elucidated. By activating PI3K/AKT, circN4BP2L2 promotes the proliferation and CSC characteristics of CRC cells, inhibits apoptosis and induces resistance to oxaliplatin in CRC cells. Furthermore, an in vivo experiment demonstrated that the inhibition of circN4BP2L2 can suppress the growth and drug resistance of CRC (170). HCC cells uptake CAF-secreted miR-1228-3p, which targets placenta associated 8, an inhibitor of PI3K/AKT, thus promoting proliferation, migration and invasion, and reducing the response of HCC cells to sorafenib (173). In addition, a study by Deng et al (174) demonstrated that HIF-1α was upregulated in CAFs under hypoxic conditions, enriching miR-21 in CAF exosomes by promoting its transcription. Subsequently, miR-21 acted on pancreatic cancer cells and maintained their stemness and gemcitabine resistance through the RAS/AKT/ERK axis.
Other signaling pathways
The TGF-β signaling pathway represents a crucial signaling pathway in the interaction between CAFs and cancer cells (80,175). SMAD is a core effector protein of TGF-β. Phosphorylated SMAD2, SMAD3 and SMAD4 form SMAD multimers, which translocate to the nucleus to regulate the transcription of genes associated with cancer growth and metastasis (175). However, research on the mechanisms by which CAF-derived exosomal ncRNAs regulate TGF-β in cancer cells remains relatively scarce. A study demonstrated that exosomal miR-423-5p could inhibit gremlin2, a SMAD-regulatory protein (176), promoting paclitaxel resistance in prostate cancer. Inhibition of TGF-β partially restored the sensitivity of cancer cells to paclitaxel. Additionally, treatment with exosomal miR-423-5p increased the expression of TGF-β. One study revealed miR-423-5p as a potential target in the TGF-β signaling pathway activated by CAFs (177). Snail is also a TF driven by TGF-β signaling, promoting the transcription of tumor EMT-related genes (178,179). Richards et al (180) demonstrated that exposure to gemcitabine increased the expression of the TF Snail in pancreatic CAFs, leading to increased release of exosomal miR-146a. This resulted in the proliferation and chemoresistance of pancreatic cancer epithelial cells. However, due to the increased expression of Snail in exosomes, the specific role of miR-146a was not determined in the study (180). CDK4/6 inhibitors represent a promising novel therapy for patients with estrogen receptor-positive BC (181). Through single-cell RNA sequencing analysis, miR-20 has been found to be enriched in CD63+ CAFs and to be transported to BC cells via exosomes. miR-20 could directly bind to the 3′ UTR of retinoblastoma susceptibility mRNA, inhibiting the therapeutic effect of CDK4/6 inhibitors on BC. The use of designed cyclic RGD-miR-20 sponge nanoparticles to inhibit miR-20 expression restored the sensitivity of BC to CDK4/6 inhibitors (181). In another study, exosomal miRNA-106b reduced the efficacy of gemcitabine in pancreatic cancer through the TP53INP1/P53 axis. Treatment with an miRNA-106b inhibitor reduced the levels of miRNA-106b in exosomes secreted by CAFs and decreased the resistance of pancreatic cancer to gemcitabine (182). Additionally, in CRC, CAF-derived exosomal miR-181d-5p and circ0067557 promote cell proliferation, migration, invasion and CRC resistance to fluorouracil (5-FU) (183,184). Mechanistically, miR-181d-5p is elevated in CAF-derived exosomes through RNA N6-methyladenosine methylation mediated by methyltransferase-like 3, and subsequently targets neurocalcin δ (NCALD) expression, thereby attenuating NCALD-mediated inactivation of the NF-κB signaling pathway (183,185). circ0067557 physically interacts with the CSC regulator Lin28, and knockdown of circ0067557 reduces Lin28 levels in CRC cells, and suppresses both malignant progression and drug resistance in CRC (184,186). Compared with normoxic CAFs, hypoxic CAFs exhibit deletion of miR-200b-3p, leading to upregulation of its targets zinc finger E-box binding homeobox 1 and E2F transcription factor 3 in CRC cells, which facilitates cell invasion, stemness maintenance and 5-FU resistance (187).
Resistance to cell death
Apoptosis, a form of programmed cell death, serves to maintain tissue homeostasis and eliminate harmful cells (188). Apoptosis is primarily initiated through two distinct pathways: The extrinsic and intrinsic pathways. The extrinsic pathway is mediated by cell surface death receptors, whereas the intrinsic pathway is activated by intracellular stress signals (such as DNA damage and oxidative stress) independent of cell surface receptors, with mitochondria serving as the central regulatory hub (189). Both pathways activate caspases, the key regulators of apoptosis, which are categorized into initiator caspases (such as caspase-8 and -9) and effector caspases (such as caspase-3, -6 and -7) (190). Upon activation, these caspases selectively cleave hundreds of intracellular substrate proteins, ultimately leading to cellular fragmentation into apoptotic bodies and programmed cell death (190,191). Abnormal regulation of the apoptotic system allows cancer cells to escape the apoptotic program, leading to unlimited proliferation and drug resistance (192). Research has highlighted the role of CAF-derived exosomal ncRNAs in cancer cell apoptosis. miR-21 is elevated in CAF exosomes and directly targets ovarian cancer cells, reducing their chemotherapy sensitivity and apoptosis. It has also been demonstrated that miR-21 directly binds to apoptotic peptidase activating factor 1 (APAF1) (193), an apoptotic protein that promotes the formation of apoptotic bodies through interactions with cytochrome c and deoxyadenosine triphosphate. The subsequent activation of caspase-9 and -3 by apoptotic bodies marks the initiation of the apoptotic program (194). By inhibiting APAF1, miR-21 reduces the expression of caspase-9 and -3 in cells, rendering ovarian cancer cells resistant to paclitaxel (193). Furthermore, miR-21 inhibits apoptosis by targeting programmed cell death 4 (PDCD4). In various gastrointestinal tumors, miR-21 has been demonstrated to be upregulated and to suppress PDCD4, thereby promoting tumor cell proliferation, invasion, migration and 5-FU resistance. Overexpression of PDCD4 can reverse the effects of miR-21, consequently reducing cell survival rates and drug resistance (195-197). The Bcl-2 family represents a critical group of proteins governing the intrinsic apoptotic pathway, consisting of anti-apoptotic members (such as Bcl-2, Bcl-xL and Bcl-w) and pro-apoptotic members [Bax/Bcl-2 homologous antagonist/killer (Bak)]. Specifically, Bax and Bak induce the formation of pores in the mitochondrial outer membrane, facilitating the release of cytochrome c and subsequent caspase activation, whereas Bcl-2 exerts its anti-apoptotic function by inhibiting Bax/Bak activation (198,199). Several exosomal ncRNAs are involved in the regulation of the Bcl-2 family. According to the expression levels of collagen type VI α6 chain, collagen type XXV α1 chain and collagen type XXVI α1 chain in OSCC-CAFs, Kang et al (64) classified CAFs into two distinct phenotypes: CAF-Ps (promote cancer development) and CAF-Ds (delay cancer development). CAF-Ps promote cancer progression while exhibiting low expression of the aforementioned genes (64). Subsequently, it was found that the exosomes secreted by CAF-Ps contained elevated levels of miR-876-3p. CAF-P-derived miR-876-3p inhibits the insulin-like growth factor-binding protein-3 (IGFBP-3)/p53 axis by targeting GATA-binding protein 1 in OSCC (200). Inhibition of IGFBP-3 reduces chemotherapy-induced apoptosis by upregulating the expression of Bcl-2, and downregulating Bax and p53. This process reduces the sensitivity of OSCC cells to cisplatin by inhibiting apoptosis (201,202). In NSCLC, CAF-secreted miR-103a-3p directly targets the pro-apoptotic protein Bak1, thereby inhibiting apoptosis in NSCLC cells and inducing cisplatin resistance (203). Additionally, miR-24-3p is transported via exosomes derived from CAFs and is highly expressed in CRC cells, which is associated with malignancy and drug resistance in CRC. Treatment of CAFs with a miR-24-3p inhibitor resulted in decreased expression of miR-24-3p in CRC cells. This led to reduced cell proliferation and enhanced apoptosis. Mechanistically, miR-24-3p inhibits apoptosis by downregulating caudal-type homeobox transcription factor 2/hephaestin, increasing the resistance of CRC cells to methotrexate (204). The packaging of ncRNAs into exosomes may be associated with RBPs such as Pumilio2 (199) and heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) (205). In head and neck cancer CAFs, hnRNPA1 binds to miR-196a, enriching it in CAFs and accelerating its packaging into exosomes through translocation between cells and exosomes. Subsequently, miR-196a targets cyclin-dependent kinase inhibitor 1B and inhibitor of growth family member 5 in head and neck cancer cells, affecting the cell cycle and apoptosis, respectively, and conferring cisplatin resistance to head and neck cancer (205).
DNA damage, particularly DNA double-strand breaks (DSBs), is a crucial factor in activating apoptotic signaling in cancer cells mediated by antitumor drugs (5). Numerous antitumor drugs, such as cisplatin, 5-FU and epirubicin, can induce DNA damage in cancer cells, thereby triggering apoptosis (206). Previous studies have shown that exosomal ncRNAs not only regulate apoptotic signaling pathways but also facilitate the repair of DNA double strands, thereby reducing the occurrence of apoptosis. In radiation-resistant ESCC, FAP+ CAFs transport lncRNA actin filament associated protein 1 antisense RNA 1 (AFAP1-AS1) to cancer cells via exosomes. AFAP1-AS1 promotes the expression of the DNA repair proteins phosphorylated (p-)ataxia telangiectasia mutated and p-checkpoint kinase 2, reducing the DNA damage marker protein γ-H2AX and inducing resistance to radiotherapy in cancer cells (207). Although not directly transmitted via exosomes, the overexpression of lncRNA uroplakin 1A antisense RNA 1 (UPK1A-AS1) in PDAC cells is induced by CAF-secreted IL-8. IL-8 promotes the expression of p65, which directly activates the transcription of UPK1A-AS1. Subsequently, UPK1A-AS1 serves as a molecular scaffold for the DSB repair proteins X-ray repair cross-complementing protein 5 and X-ray repair cross-complementing protein 6, mediating DNA repair through non-homologous end-joining, thus conferring cisplatin resistance to PDAC (208).
Ferroptosis, a regulated form of cell death distinct from apoptosis, was first proposed in 2012 (209,210). Ferroptosis is characterized by an increase in the small pool of Fe2+ and lipid peroxidation (8). The accumulation of Fe2+ within cells generates free radicals through the Fenton reaction and contributes to the production of phospholipid hydroperoxides (211). Simultaneously, Fe2+ catalyzes the generation of reactive ROS, thereby promoting lipid peroxidation. This overwhelming lipid peroxidation ultimately leads to cell death (212). The canonical regulatory pathway of ferroptosis relies on the GSH peroxidase GPX4, whose normal biological function depends on the support of GSH (213). Inhibition of GPX4 and GSH leads to the accumulation of lipid ROS, resulting in rupture of the cell membrane (214). Sorafenib can inhibit solute carrier family 3 member 2 and solute carrier family 7 member 11 (SLC7A11), promoting lipid peroxidation in HCC. This hinders the production of GSH and inhibits GPX4 expression, thereby upregulating ferroptosis in HCC cells (215). However, the regulatory network within the TME may enhance cancer cell resistance to ferroptosis by strengthening the antioxidant system, thus inducing drug resistance (215).
The activation of the cystine-glutamate antiporter SLC7A11 increases GSH production by transporting cysteine (216). In glioma, CAF-derived exosomes promote deleted in lymphocytic leukemia 1 (DLEU1) transcription through heat shock transcription factor 1 activation, thereby upregulating DLEU1 expression (217). Subsequently, DLEU1 interacts directly with ZFP36 and inhibits activating transcription factor 3 (ATF3), which binds to the SLC7A11 promoter and inhibits SLC7A11 in a p53-independent manner (218). This process confers ferroptosis resistance to glioma cells (217). Conversely, lncRNA DACT3-AS1 is transferred from CAF exosomes to GC cells. DACT3-AS1 inhibits cell proliferation, migration and invasion both in vivo and in vitro by sponging miR-181a-5p, which targets sirtuin 1 (SIRT1) (219). In this context, SIRT1 activates ATF3 by depleting NAD+, thereby inhibiting SLC7A11 and GPX. By suppressing miR-181a-5p, DACT3-AS1 promotes SIRT1-mediated ferroptosis, consequently increasing cellular sensitivity to 5-FU (220). γ-glutamylcyclotransferase 1 (ChaC1) is a key enzyme in the GSH degradation process (221). CAF-derived exosomes can hinder ferroptosis induced by erastin (a GSH inhibitor) via miR-432-5p, which directly targets to ChaC1. By inhibiting ChaC1 expression, miR-432-5p attenuates GSH depletion and ferroptosis in prostate cancer cells, thereby inducing docetaxel resistance (39). Additionally, ACSL4 and arachidonic acid 15-lipoxygenase-1 (ALOX15) are key enzymes in phospholipid metabolism and induction of lipid peroxidation, mediating the specific oxidation of polyunsaturated fatty acids-phosphates (222,223). In digestive system cancers, CAFs secrete miR-522 and miR-3173-5p, which target ALOX15 and ACSL4, respectively, leading to the accumulation of lipid ROS in cancer cells and inducing acquired drug resistance (40,224).
EMT in cancer cells
EMT is a process driven by EMT-related TFs that leads to the transformation of cells from an epithelial-like morphology to a mesenchymal morphology. Following EMT, cancer cells exhibit an elongated morphology and upregulated expression of mesenchymal markers such as N-cadherin and vimentin, which respectively reduce intercellular adhesion and enhance cell motility, and thus, the cells become more invasive (225). EMT has a strong association with cancer drug resistance. Multiple signaling pathways, such as Wnt/β-catenin and TGF-β, that promote EMT also activate multidrug resistance (MDR) genes (226). The adaptive changes in autophagy and metabolism during EMT in cancer cells also promote the occurrence of MDR (226,227). CAFs can promote EMT in cancer cells, with exosomal ncRNAs serving a role in this process (19). In CRC, CAFs directly transfer miR-92a-3p to CRC cells via exosomes. Upregulation of miR-92a-3p activates the Wnt/β-catenin signaling pathway (228). Furthermore, miR-92a-3p has been demonstrated to directly target and suppress F-box and WD repeat domain containing 7, an E3-ubiquitin ligase responsible for degrading the EMT TF zinc finger E-box binding homeobox 2 (229). Experimental results revealed that miR-92a-3p promoted cell proliferation, invasion and migration, while inducing CSC generation, EMT and resistance to 5-FU in CRC (228). Crk-like adapter protein (CrkL) has been identified as a key regulator of EMT, mediating C-C motif chemokine ligand 20/C-C motif chemokine receptor 6-induced EMT through activation of the AKT pathway (230). Long intergenic non-protein coding RNA 355 (LINC00355), present in CAF-derived exosomes, acts as a sponge for miR-34b-5p, which can target and inhibit CrkL. LINC00355 indirectly upregulates the expression of CrkL, promoting EMT and MDR in CRC cells (231). Additionally, exosomal miR-625-3p inhibits the tumor suppressor gene WW domain containing oxidoreductase (WWOX) by targeting CUGBP, Elav-like family member 2 (CELF2), which regulates SMAD3, a component of the TGF-β signaling pathway. Through the CELF2/WWOX axis, miR-625-3p promotes cell EMT and chemoresistance in CRC (232,233).
Drug transporters
The ability of cancer cells to acquire drug resistance manifests as reduced drug uptake and enhanced drug efflux, which is achieved through the abnormal upregulation of drug transporter proteins such as ABC transporters (234). Numerous ncRNAs, such as lncRNA cancer susceptibility 9, miR-27a and miR-451, serve a critical role in regulating drug transporters, and targeting these proteins may be an effective way to alleviate drug resistance in cancer cells (235). Exosomal LINC00355 derived from CAFs in BC promotes the expression of ATP-binding cassette subfamily B member 1 (ABCB1), a drug efflux transporter and a target of miR-34b-5p, by sponging miR-34b-5p. This increased expression enhances the efflux of chemotherapy drugs from cancer cells. Overexpression of miR-34b-5p or knockdown of ABCB1 can eliminate the effect of LINC00355 in CAF exosomes, thereby restoring the sensitivity of BC cells to cisplatin (236). Similarly, midkine in exosomes increases the expression levels of lncRNA antisense non-coding RNA in the INK4 locus (ANRIL) in OSCC. Elevated ANRIL expression is associated with advanced TNM staging and lymph node metastasis in OSCC. Quantitative PCR analysis has demonstrated that ANRIL upregulated the drug transporters multidrug resistance-associated protein 1 (MRP1) and ATP-binding cassette subfamily C member 2 (ABCC2), while ANRIL knockdown reduced their expression, indicating positive regulation of MRP1/ABCC2 by ANRIL. Additionally, ANRIL suppresses caspase-3 activation to inhibit apoptotic pathways, thereby promoting tumor cell proliferation and reducing cisplatin-induced apoptosis (237). Additionally, anti-pyrimidine drugs such as gemcitabine enter cancer cells through balanced uptake by nucleoside transporter 2 (ENT2). miR-4717-5p, one of the most abundant miRNAs in CAF-derived exosomes in lymphoma, enters lymphoma cells and inhibits ENT2 expression to reduce gemcitabine uptake, thereby decreasing the sensitivity of lymphoma to gemcitabine (238).
Immune evasion
Cytokines secreted by CAFs serve a crucial role in immune cell regulation within the TME (239). However, to the best of our knowledge, the function of ncRNAs derived from CAF-sourced exosomes, which serve as important mediators of intercellular communication in the TME, remains unexplored. HLA is a type of protein found on cell surfaces, and is essential for the immune recognition mechanism of CD8+ T cells (240). In PDAC, lncRNA RP11-161H23.5 derived from CAFs promotes immune evasion via downregulation of HLA-A expression through binding to CCR4-NOT transcription complex subunit 4, which enhances the degradation of HLA-A mRNA by shortening its poly(A) tail. Engineered extracellular vesicles loaded with RP11-161H23.5 small interfering RNA (siRNA) have been constructed to enhance immunotherapy effectiveness, emerging as a potential strategy for the treatment of pancreatic cancer (241). In lung cancer, CAFs interact with programmed cell death protein 1 (PD-1) on the surface of peripheral blood mononuclear cells (PBMCs) by upregulating the expression of programmed death-ligand 1 (PD-L1) in lung cancer cells (242). This process inhibits signaling pathways in PBMCs, such as the PI3K/AKT and Ras/MEK/ERK pathways, thus blocking the proliferation, differentiation and metabolic reprogramming of PBMCs (243). A study (242) has shown that the lncRNA OIP5-AS1 serves an important role by functioning as a sponge to adsorb miR-142-5p, which directly targets and inhibits PD-L1 expression. OIP5-AS1 enters lung cancer cells through exosomes secreted by CAFs, upregulating the expression levels of PD-L1 in lung cancer cells by inhibiting miR-142-5p. This process subsequently suppresses PBMC-induced cell death via the PD-L1/PD-1 axis (242,243).
Association between exosomal ncRNAs and soluble factors
The regulation of tumor drug resistance by CAFs is not mediated by a single pathway but rather involves the synergistic effects of multiple mechanisms. Evidence indicates that cytokines and chemoattractants secreted by CAFs also contribute to tumor progression and drug resistance (42). Activated CAFs secrete IL-6, which stimulates the activation of the JAK/STAT3 pathway in cancer cells and enhances TGF-β-induced p-Smad3 and Snail expression, thereby promoting EMT (244). Additionally, IL-6-activated JAK/STAT3 suppresses p53, attenuating the response of prostate cancer cells to doxorubicin (245). HGF derived from α-SMA+ CAFs activates c-Met and further promotes the PI3K/AKT and FRA1/hes related family bHLH transcription factor with YRPW motif 1 signaling cascades, inducing drug resistance in HCC, ovarian cancer and BC (246-248). Furthermore, SDF-1 secreted by CAFs upregulates SATB homeobox 1 expression in pancreatic cancer cells, enhancing their resistance to gemcitabine (249). The chemoattractant CXCL12, also secreted by CAFs, binds to the CXCR4 receptor on cancer cells, activating the Wnt/β-catenin pathway, and inducing EMT and cisplatin resistance in ovarian cancer (250). These findings suggest that various soluble factors secreted by CAFs and exosomal ncRNAs share common mechanisms of action, such as activating the TGF-β and Wnt/β-catenin pathways, thereby exerting synergistic effects. Compared with soluble factors, exosomal ncRNAs may serve as key regulatory nodes in signaling pathways, modulating multiple cascades and avoiding feedback activation upon inhibition of a single pathway (21,22). Furthermore, ncRNAs can induce cancer drug resistance through diverse mechanisms. For example, miR-21 mediates the activation of multiple drug resistance-related signaling pathways and promotes apoptosis resistance (163,174,193). Given the tissue- and cell-specific expression of ncRNAs, targeting of ncRNAs may represent a more comprehensive and personalized therapeutic strategy compared with targeting of soluble factors.
Unresolved issues and research visions
The present review summarizes the role of exosomal ncRNAs in CAF-mediated tumor drug resistance, noting the diversity of their mechanisms of action. However, current research does not encompass all the mechanisms associated with CAF-derived exosomal ncRNAs, and a number of these mechanisms remain to be validated by further studies. CAFs serve a crucial role in the metabolic regulation of cancer cells (97). Cytokines secreted by CAFs stimulate glycolysis in cancer cells by activating the HIF-1 and PI3K/Akt signaling pathways (251,252). Glycolysis provides tumors with sufficient energy in nutrient-deprived environments, supporting their survival and supplying ATP for drug efflux pumps (253). Concurrently, CAFs secrete substantial amounts of pyruvate and lactate, synergizing with cancer cell glycolysis to suppress the immune microenvironment (97). Only a limited number of studies have elucidated the association between CAFs and cancer lipid metabolism. Ippolito et al (254) demonstrated that lactate secreted by CAFs upregulated the expression levels of lipid metabolism-related genes in cancer cells. Aberrant lipid metabolism supplies energy and metabolic substrates to cancer cells. Additionally, its catabolic products act as signaling molecules to modulate drug resistance-related pathways (255). Furthermore, CAFs exhibit high expression of glutamine synthetase, leading to increased secretion of glutamine. The glutamine molecules are catabolized into glutamate in cancer cells to support their proliferation (256). CAF-derived exosomal ncRNA is also a crucial factor regulating metabolism. Targeting ncRNA can overcome cancer growth, metastasis and drug resistance mediated by the metabolic reprogramming of cancer cells (257). In oral cancer, lncRNA H19 secreted by CAFs upregulates the expression of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3, a key enzyme regulating the cellular glycolytic pathway, by acting as a sponge for miR-675-5p. This interaction allows H19 to induce increased glycolysis in oral cancer, thereby promoting tumor growth (258). In BC, exosomes derived from CAFs have been found to indirectly upregulate negative elongation factor complex member E (NELFE) by acting as a sponge for miR-4510. Long intergenic non-protein coding RNA 1711 activates thioredoxin through the miR-4510/NELFE axis, thereby promoting glycolysis, proliferation and invasion of BC cells (259). In addition to glycolysis, exosomes derived from CRC cells that contain HSPC111 could promote the formation of a pre-metastatic niche and facilitate CRC liver metastasis in mouse models. This occurs through the upregulation of acetyl-CoA levels in CAFs, which is mediated by lipid metabolism promoted by ATP citrate lyase phosphorylation (260). The accumulation of acetyl-CoA enhances H3K27 acetylation in CAFs, thus facilitating the expression and secretion of C-X-C motif chemokine ligand 5 (CXCL5). Furthermore, the CXCL5-C-X-C motif chemokine receptor 2 axis increases the excretion of CRC cell exosomes containing HSPC111, thereby establishing a positive feedback loop that promotes CRC cell liver metastasis (260). Although the role of ncRNAs derived from CAFs in drug resistance through metabolic reprogramming has been hypothesized, to the best of our knowledge, the association between the two has not been specifically confirmed by experimental data.
Various stresses such as antitumor drugs promote tumor autophagy (261). Autophagy facilitates tumor growth by recycling metabolic substances and regulating mitochondrial function (261). Autophagy involves >20 proteins encoded by autophagy-related genes. These proteins encapsulate cytoplasmic materials into the double-membrane vesicles of autophagosomes. After engulfing the materials, autophagosomes fuse with lysosomes, utilizing pH-sensitive hydrolases to degrade the engulfed substances (262). Although Zhu et al (263) demonstrated that CAF-derived lncRNA FAL1 promoted oxaliplatin resistance by inhibiting autophagy, it was only under conditions of excessive autophagy activation that it could exert its antitumor effect by inducing cell death (264). CAF-mediated autophagy in cancer cells contributes to drug resistance (265,266). The role and mechanism of exosomal ncRNAs in this process remain to be further investigated.
Exosomal ncRNAs secreted by CAFs may also influence other cells within the TME, including TAMs and endothelial cells (227). The role of crosstalk mediated by exosomal ncRNAs between CAFs and these cells in the context of drug resistance has not been extensively studied. Given that much of the research on the TME has concentrated on individual cell types, further exploration in this area could enhance the understanding of the complex regulatory networks within the TME and their impact on tumors. Additionally, the subtypes of CAFs when examining the functions of CAF-derived ncRNA have not been thoroughly investigated. This limitation impedes a more comprehensive understanding of the roles of CAFs expressing different surface receptors and their secreted ncRNAs in cancer, leading to issues of irreproducibility in research.
Clinical application and challenges
Given the various roles of CAFs in tumors, targeting CAFs (promoting their transformation into NFs or anticancer phenotypic CAFs) may benefit patients with tumors (267). In practice, achieving clinical benefits may not necessarily require the elimination or reprogramming of CAFs, but can also be achieved by blocking signals from CAFs. For example, targeting CXCL12 signaling can be considered as targeting CAFs, as they are the primary source of chemokines in numerous tumors (94). Indeed, targeting the communication process between CAFs and cancer cells is also an effective method to alter the function of CAFs (267), making ncRNAs important candidate targets for the treatment of CAF-mediated cancer drug resistance. Therapies targeting ncRNAs are based on RNA modification technologies that regulate their expression: Upregulating cancer-suppressing ncRNAs or downregulating cancer-promoting ncRNAs (268). Multiple methods have been used to target ncRNA expression to regulate tumor progression (268). For instance, the CRISPR-CRISPR associated protein 9 (Cas9) system (269), siRNA (270) and antisense DNA oligonucleotides containing locked nucleic acid (271) have been used to inhibit cancer-promoting ncRNAs, while mimics can be employed to overexpress cancer-suppressing ncRNAs (272). Furthermore, small activating RNA (saRNA), which induces gene expression by targeting promoter regions, has also been employed to upregulate target ncRNAs (273,274).
Given the inherent instability and high molecular weight of these RNA-based therapeutics in vivo, the delivery systems for these 'RNA cargos' are increasingly being optimized to ensure their precise and efficient transport to specific cells (275). Delivery systems are generally divided into viral delivery systems and non-viral delivery systems. Based on the type of carrier, the non-viral delivery systems can be categorized into nanoparticles (including lipid, polymer and inorganic nanoparticles), liposomes, polymer micelles and exosomes (275). Nanoparticles can enhance their efficacy through crosslinking agents or stimulus-responsive systems. For instance, some studies have utilized GSH and pH-sensitive nanoparticles. The pH-sensitive properties allow nanoparticles to accumulate in the acidic environment of tumor tissues, where they can further reduce the abundant GSH in cells to thiols, leading to micelle degradation and promoting the release of their contents (276,277). Simultaneously, these nanoparticles can also co-deliver chemotherapeutic drugs with ncRNA-targeted drugs to achieve a synergistic antitumor effect (278). Additionally, using viral vectors such as adenoviruses, adeno-associated viruses and lentiviruses to deliver nucleic acids that regulate ncRNAs is an effective strategy (268). Gene therapies targeting ncRNAs have entered clinical trials, and have demonstrated good tolerability and efficacy in some studies (279,280).
The timing of cancer diagnosis and the initiation of treatment are critical determinants of patient prognosis (281). Exosomes secreted by CAFs enter the circulatory system and are abundantly present in bodily fluids such as blood, urine and saliva, and remain stable in suitable environments (282,283). This allows the ncRNAs specifically expressed in exosomes to serve as important biomarkers to monitor the progression, prognosis and drug response of patients with cancer (284). Xu et al (285) developed a CAF-related miRNA model and found that miR-106b-5p was associated with the growth and recurrence of prostate cancer. CAF-derived exosomal miR-146a-5p has been detected in the bodily fluids of patients with bladder cancer, and its elevated levels are positively associated with the recurrence rate of bladder cancer (41). The findings of Zhou et al (207) indicated that AFAP1-AS1 was secreted by CAFs, and high levels of AFAP1-AS1 in plasma were associated with low sensitivity to definitive chemoradiotherapy in patients with ESCC.
Targeting the exosomal ncRNA derived from CAFs provides a promising strategy to overcome drug resistance in cancer. However, several urgent issues hinder its clinical application. In ncRNA-targeted therapies, off-target effects and systemic toxicity present formidable challenges (286). Despite the considerable potential of ncRNAs in cancer treatment, their off-target effects continue to be a barrier that must be addressed (286). Off-target effects can arise with any delivery system employed to target ncRNAs (286). Furthermore, systemic toxicity leading to patient intolerance remains a primary reason for the discontinuation of clinical trials (287). A phase I clinical trial was conducted to evaluate the efficacy and safety of a miR-34a liposomal mimetic in patients with advanced solid tumors. Among the 85 enrolled patients, a partial response was observed in three cases, while stable disease was achieved in 16 cases. However, all participants experienced adverse reactions, including fever, nausea and pain. The trial was ultimately terminated due to four fatalities attributed to severe adverse events (288). On the other hand, although delivery systems have advanced, different types of delivery systems still exhibit certain disadvantages. For instance, while liposomes and polymer nanoparticles can safely and effectively transfer large genes, their transfection efficiency and transgene expression are relatively low (289). Conversely, although viral vectors address the issue of transfection efficiency and allow for sustained gene expression, they are limited in their development due to high immunogenicity and low payload capacity (290). Despite these challenges, it is hypothesized that with the emergence of novel, innovative research and interdisciplinary collaboration, therapies targeting ncRNAs will achieve breakthroughs.
Conclusion
The TME serves a crucial role in cancer growth, metastasis and drug resistance. CAFs are a critical component of the TME, constructing a drug-resistant TME through a complex intercellular communication network. Accumulating evidence suggests that ncRNAs exert extensive biological functions in regulating cancer progression mediated by CAFs. The present review focuses on the bidirectional regulatory mechanisms of exosomal ncRNAs in CAF-mediated drug resistance. Exosomal ncRNAs derived from tumor cells activate multiple signaling pathways, inducing the differentiation of NFs into CAFs. This establishes a suitable niche for CAFs to regulate cancer drug resistance. Conversely, CAF-derived exosomal ncRNAs can be internalized by cancer cells, activating drug resistance-related signaling pathways such as Wnt/β-catenin, STAT3, TGF-β and PI3K/AKT. Furthermore, exosomal ncRNAs regulate cell death, EMT and drug transporter expression levels, and promote immune evasion, all of which contribute to cancer drug resistance. Additionally, CAF-derived exosomal ncRNAs may mediate drug resistance by regulating autophagy, metabolic reprogramming and the immune microenvironment, which are promising areas for future research
It is noteworthy that the specifically expressed ncRNAs in CAF-derived exosomes hold potential for the development of novel clinical diagnostic and therapeutic approaches. Exosomal ncRNAs in bodily fluids are readily accessible and quantifiable, serving as emerging biomarkers that reflect tumor TNM staging and therapeutic drug sensitivity in patients with cancer. Furthermore, RNA modification technologies, including siRNA, saRNA and CRISPR-Cas9, can disrupt intercellular communication and signaling cascades by regulating exosomal ncRNAs, thereby effectively reversing tumor drug resistance. Various delivery systems have been designed by researchers to achieve precise and efficient delivery of ncRNA-targeting therapeutics. Currently, ncRNA-targeting strategies are undergoing continuous advancement and entering clinical trials. With further research progress, intervention strategies targeting CAF-derived exosomal ncRNAs are expected to overcome the limitations of conventional therapies and provide innovative treatment approaches to combat tumor drug resistance.
Availability of data and materials
Not applicable.
Authors' contributions
JL wrote the majority of the manuscript and created the figures and tables. YH contributed to the manuscript writing. LF and MS assisted in figure preparation and manuscript revision. GH, ZW and FD revised the manuscript. YZ and YX revised the manuscript and prepared tables. YL revised the manuscript and supervised the process. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Acknowledgements
Not applicable.
Funding
No funding was received.
References
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Zhang H and Chen X: Drug resistance and combating drug resistance in cancer. Cancer Drug Resist. 2:141–160. 2019.PubMed/NCBI | |
Dong D, Yu X, Xu J, Yu N, Liu Z and Sun Y: Cellular and molecular mechanisms of gastrointestinal cancer liver metastases and drug resistance. Drug Resist Updat. 77:1011252024. View Article : Google Scholar : PubMed/NCBI | |
Hussain S, Singh A, Nazir SU, Tulsyan S, Khan A, Kumar R, Bashir N, Tanwar P and Mehrotra R: Cancer drug resistance: A fleet to conquer. J Cell Biochem. 120:14213–14225. 2019. View Article : Google Scholar : PubMed/NCBI | |
Carneiro BA and El-Deiry WS: Targeting apoptosis in cancer therapy. Nat Rev Clin Oncol. 17:395–417. 2020. View Article : Google Scholar : PubMed/NCBI | |
Rezayatmand H, Razmkhah M and Razeghian-Jahromi I: Drug resistance in cancer therapy: The Pandora's Box of cancer stem cells. Stem Cell Res Ther. 13:1812022. View Article : Google Scholar : PubMed/NCBI | |
Wang Z, Wang Y, Li Z, Xue W, Hu S and Kong X: Lipid metabolism as a target for cancer drug resistance: Progress and prospects. Front Pharmacol. 14:12743352023. View Article : Google Scholar : PubMed/NCBI | |
Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B, Liu J, Yu X and Shi S: Targeting cell death pathways for cancer therapy: Recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol. 15:1742022. View Article : Google Scholar : PubMed/NCBI | |
Gao L, Wu ZX, Assaraf YG, Chen ZS and Wang L: Overcoming anti-cancer drug resistance via restoration of tumor suppressor gene function. Drug Resist Updat. 57:1007702021. View Article : Google Scholar : PubMed/NCBI | |
Nussinov R, Tsai CJ and Jang H: Anticancer drug resistance: An update and perspective. Drug Resist Updat. 59:1007962021. View Article : Google Scholar : PubMed/NCBI | |
Pitt JM, Marabelle A, Eggermont A, Soria JC, Kroemer G and Zitvogel L: Targeting the tumor microenvironment: Removing obstruction to anticancer immune responses and immunotherapy. Ann Oncol. 27:1482–1492. 2016. View Article : Google Scholar : PubMed/NCBI | |
Kundu M, Butti R, Panda VK, Malhotra D, Das S, Mitra T, Kapse P, Gosavi SW and Kundu GC: Modulation of the tumor microenvironment and mechanism of immunotherapy-based drug resistance in breast cancer. Mol Cancer. 23:922024. View Article : Google Scholar : PubMed/NCBI | |
Giraldo NA, Sanchez-Salas R, Peske JD, Vano Y, Becht E, Petitprez F, Validire P, Ingels A, Cathelineau X, Fridman WH and Sautès-Fridman C: The clinical role of the TME in solid cancer. Br J Cancer. 120:45–53. 2019. View Article : Google Scholar : | |
Peng Z, Tong Z, Ren Z, Ye M and Hu K: Cancer-associated fibroblasts and its derived exosomes: A new perspective for reshaping the tumor microenvironment. Mol Med. 29:662023. View Article : Google Scholar : PubMed/NCBI | |
Loh JJ and Ma S: The role of cancer-associated fibroblast as a dynamic player in mediating cancer stemness in the tumor microenvironment. Front Cell Dev Biol. 9:7276402021. View Article : Google Scholar : PubMed/NCBI | |
Wu F, Yang J, Liu J, Wang Y, Mu J, Zeng Q, Deng S and Zhou H: Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct Target Ther. 6:2182021. View Article : Google Scholar : PubMed/NCBI | |
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 | |
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 | |
Fiori ME, Di Franco S, Villanova L, Bianca P, Stassi G and De Maria R: Cancer-associated fibroblasts as abettors of tumor progression at the crossroads of EMT and therapy resistance. Mol Cancer. 18:702019. View Article : Google Scholar : PubMed/NCBI | |
Zhang L and Yu D: Exosomes in cancer development, metastasis, and immunity. Biochim Biophys Acta Rev Cancer. 1871:455–468. 2019. View Article : Google Scholar : PubMed/NCBI | |
Li C, Ni YQ, Xu H, Xiang QY, Zhao Y, Zhan JK, He JY, Li S and Liu YS: Roles and mechanisms of exosomal non-coding RNAs in human health and diseases. Signal Transduct Target Ther. 6:3832021. View Article : Google Scholar : PubMed/NCBI | |
Nemeth K, Bayraktar R, Ferracin M and Calin GA: Non-coding RNAs in disease: From mechanisms to therapeutics. Nat Rev Genet. 25:211–232. 2024. View Article : Google Scholar | |
Hill M and Tran N: miRNA interplay: Mechanisms and consequences in cancer. Dis Model Mech. 14:dmm0476622021. View Article : Google Scholar : PubMed/NCBI | |
Bhan A, Soleimani M and Mandal SS: Long noncoding RNA and cancer: A new paradigm. Cancer Res. 77:3965–3981. 2017. View Article : Google Scholar : PubMed/NCBI | |
Liu X, Zhang Y, Zhou S, Dain L, Mei L and Zhu G: Circular RNA: An emerging frontier in RNA therapeutic targets, RNA therapeutics, and mRNA vaccines. J Control Release. 348:84–94. 2022. View Article : Google Scholar : PubMed/NCBI | |
Ibrahim FM, Saleh RO, Uinarni H, Bokov DO, Menon SV, Zarifovich KB, Misra N, Al-Hamdani MM, Husseen B and Jawad MA: Exosomal noncoding RNA (ncRNA) in breast cancer pathogenesis and therapy; two sides of the same coin. Exp Cell Res. 444:1143592025. View Article : Google Scholar | |
Saadh MJ, Allela OQB, Kareem RA, Ballal S, Chahar M, Saini S, Prasad GVS, Sameer HN, Hamad AK, Athab ZH and Adil M: The role of exosomal non-coding RNAs in the breast cancer tumor microenvironment. Funct Integr Genomics. 25:322025. View Article : Google Scholar : PubMed/NCBI | |
Alipoor SD and Chang H: Exosomal miRNAs in the tumor microenvironment of multiple myeloma. Cells. 12:10302023. View Article : Google Scholar : PubMed/NCBI | |
Huang Q, Zhong X, Li J, Hu R, Yi J, Sun J, Xu Y and Zhou X: Exosomal ncRNAs: Multifunctional contributors to the immunosuppressive tumor microenvironment of hepatocellular carcinoma. Biomed Pharmacother. 173:1164092024. View Article : Google Scholar : PubMed/NCBI | |
Hussen BM, Abdullah ST, Abdullah SR, Younis YM, Hidayat HJ, Rasul MF and Mohamadtahr S: Exosomal non-coding RNAs: Blueprint in colorectal cancer metastasis and therapeutic targets. Noncoding RNA Res. 8:615–632. 2023. View Article : Google Scholar : PubMed/NCBI | |
Li W, Wang X, Li C, Chen T and Yang Q: Exosomal non-coding RNAs: Emerging roles in bilateral communication between cancer cells and macrophages. Mol Ther. 30:1036–1053. 2022. View Article : Google Scholar : | |
Entezari M, Sadrkhanloo M, Rashidi M, Asnaf SE, Taheriazam A, Hashemi M, Ashrafizadeh M, Zarrabi A, Rabiee N, Hushmandi K, et al: Non-coding RNAs and macrophage interaction in tumor progression. Crit Rev Oncol Hematol. 173:1036802022. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Yu D, Ji C, Wang M, Fu M, Qian Y and Zhang X, Ji R, Li C, Gu J and Zhang X: Exosomal miR-4745-5p/3911 from N2-polarized tumor-associated neutrophils promotes gastric cancer metastasis by regulating SLIT2. Mol Cancer. 23:1982024. View Article : Google Scholar : PubMed/NCBI | |
Zhao Y, Jia Y, Wang J, Chen X, Han J, Zhen S, Yin S, Lv W, Yu F, Wang J, et al: circNOX4 activates an inflammatory fibroblast niche to promote tumor growth and metastasis in NSCLC via FAP/IL-6 axis. Mol Cancer. 23:472024. View Article : Google Scholar : PubMed/NCBI | |
Golestannejad P, Monkaresi M, Zhian Zargaran F, Khosravani M, Asgari P, Mobaraki H, Gorjizad M, Hasany S, Senobari Ghezeljehmeidan A, Hemmati S, et al: Role of cancer associated fibroblast (CAF) derived miRNAs on head and neck malignancies microenvironment: A systematic review. BMC Cancer. 25:5822025. View Article : Google Scholar : PubMed/NCBI | |
Ye F, Liang Y, Wang Y, Le Yang R, Luo D, Li Y, Jin Y, Han D, Chen B, Zhao W, et al: Cancer-associated fibroblasts facilitate breast cancer progression through exosomal circTBPL1-mediated intercellular communication. Cell Death Dis. 14:4712023. View Article : Google Scholar : PubMed/NCBI | |
Shelton M, Anene CA, Nsengimana J, Roberts W, Newton-Bishop J and Boyne JR: The role of CAF derived exosomal microRNAs in the tumour microenvironment of melanoma. Biochim Biophys Acta Rev Cancer. 1875:1884562021. View Article : Google Scholar | |
Sun S, Zhang Y, Li Y and Wei L: Crosstalk between colorectal cancer cells and cancer-associated fibroblasts in the tumor microenvironment mediated by exosomal noncoding RNAs. Front Immunol. 14:11616282023. View Article : Google Scholar : PubMed/NCBI | |
Zhao J, Shen J, Mao L, Yang T, Liu J and Hongbin S: Cancer associated fibroblast secreted miR-432-5p targets CHAC1 to inhibit ferroptosis and promote acquired chemoresistance in prostate cancer. Oncogene. 43:2104–2114. 2024. View Article : Google Scholar : PubMed/NCBI | |
Qi R, Bai Y, Li K, Liu N, Xu Y, Dal E, Wang Y, Lin R, Wang H, Liu Z, et al: Cancer-associated fibroblasts suppress ferroptosis and induce gemcitabine resistance in pancreatic cancer cells by secreting exosome-derived ACSL4-targeting miRNAs. Drug Resist Updat. 68:1009602023. View Article : Google Scholar : PubMed/NCBI | |
Zhuang J, Shen L, Li M, Sun J, Hao J, Li J, Zhu Z, Ge S, Zhang D, Guo H, et al: Cancer-associated fibroblast-derived miR-146a-5p generates a niche that promotes bladder cancer stemness and chemoresistance. Cancer Res. 83:1611–1627. 2023. 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 | |
Farhood B, Najafi M and Mortezaee K: Cancer-associated fibroblasts: Secretions, interactions, and therapy. J Cell Biochem. 120:2791–2800. 2019. View Article : Google Scholar | |
Joshi RS, Kanugula SS, Sudhir S, Pereira MP, Jain S and Aghi MK: The role of cancer-associated fibroblasts in tumor progression. Cancers. 13:13992021. View Article : Google Scholar : PubMed/NCBI | |
Arina A, Idel C, Hyjek EM, Alegre ML, Wang Y, Bindokas VP, Weichselbaum RR and Schreiber H: Tumor-associated fibroblasts predominantly come from local and not circulating precursors. Proc Natl Acad Sci USA. 113:7551–7556. 2016. View Article : Google Scholar : PubMed/NCBI | |
Raz Y, Cohen N, Shani O, Bell RE, Novitskiy SV, Abramovitz L, Levy C, Milyavsky M, Leider-Trejo L, Moses HL, et al: Bone marrow-derived fibroblasts are a functionally distinct stromal cell population in breast cancer. J Exp Med. 215:3075–3093. 2018. View Article : Google Scholar : PubMed/NCBI | |
Peng Y and Li Z and Li Z: GRP78 secreted by tumor cells stimulates differentiation of bone marrow mesenchymal stem cells to cancer-associated fibroblasts. Biochem Biophys Res Commun. 440:558–563. 2013. View Article : Google Scholar : PubMed/NCBI | |
Potenta S, Zeisberg E and Kalluri R: The role of endothelial-to-mesenchymal transition in cancer progression. Br J Cancer. 99:1375–1379. 2008. View Article : Google Scholar : PubMed/NCBI | |
Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong ST, Choi H, El Rayes T, Ryu S, Troeger J, et al: Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 527:472–476. 2015. View Article : Google Scholar : PubMed/NCBI | |
Jotzu C, Alt E, Welte G, Li J, Hennessy BT, Devarajan E, Krishnappa S, Pinilla S, Droll L and Song YH: Adipose tissue derived stem cells differentiate into carcinoma-associated fibroblast-like cells under the influence of tumor derived factors. Cell Oncol (Dordr). 34:55–67. 2011. View Article : Google Scholar : PubMed/NCBI | |
Rinkevich Y, Mori T, Sahoo D, Xu PX, Bermingham JR Jr and Weissman IL: Identification and prospective isolation of a mesothelial precursor lineage giving rise to smooth muscle cells and fibroblasts for mammalian internal organs, and their vasculature. Nat Cell Biol. 14:1251–1260. 2012. 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 | |
Pan C, Liu P, Ma D, Zhang S, Ni M, Fang Q and Wang J: Bone marrow mesenchymal stem cells in microenvironment transform into cancer-associated fibroblasts to promote the progression of B-cell acute lymphoblastic leukemia. Biomed Pharmacother. 130:1106102020. View Article : Google Scholar | |
Sun X, Cai W, Li H, Gao C, Ma X, Guo Y, Fu D, Xiao D, Zhang Z, Wang Y, et al: Endothelial-like cancer-associated fibroblasts facilitate pancreatic cancer metastasis via vasculogenic mimicry and paracrine signalling. Gut. Mar 23–2025.Epub ahead of print. View Article : Google Scholar | |
Hamabe-Horiike T, Harada SI, Yoshida K, Kinoshita J, Yamaguchi T and Fushida S: Adipocytes contribute to tumor progression and invasion of peritoneal metastasis by interacting with gastric cancer cells as cancer associated fibroblasts. Cancer Rep (Hoboken). 6:e16472023. | |
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 : | |
Bu L, Baba H, Yoshida N, Miyake K, Yasuda T, Uchihara T, Tan P and Ishimoto T: Biological heterogeneity and versatility of cancer-associated fibroblasts in the tumor microenvironment. Oncogene. 38:4887–4901. 2019. View Article : Google Scholar : PubMed/NCBI | |
Biffi G and Tuveson DA: Diversity and biology of cancer-associated fibroblasts. Physiol Rev. 101:147–176. 2021. View Article : Google Scholar : | |
Chhabra Y and Weeraratna AT: Fibroblasts in cancer: Unity in heterogeneity. Cell. 186:1580–1609. 2023. View Article : Google Scholar : PubMed/NCBI | |
Sugimoto H, Mundel TM, Kieran MW and Kalluri R: Identification of fibroblast heterogeneity in the tumor microenvironment. Cancer Biol Ther. 5:1640–1646. 2006. View Article : Google Scholar : PubMed/NCBI | |
Cortez E, Roswall P and Pietras K: Functional subsets of mesenchymal cell types in the tumor microenvironment. Semin Cancer Biol. 25:3–9. 2014. View Article : Google Scholar : PubMed/NCBI | |
Kim SJ, Kim SA, Choi YA, Park DY and Lee J: Alpha-smooth muscle actin-positive perivascular cells in diabetic retina and choroid. Int J Mol Sci. 21:21582020. View Article : Google Scholar : PubMed/NCBI | |
Wu Y, Wu C, Shi T, Cai Q, Wang T, Xiong Y, Zhang Y, Jiang W, Lu M, Chen Z, et al: FAP expression in adipose tissue macrophages promotes obesity and metabolic inflammation. Proc Natl Acad Sci USA. 120:e23030751202023. View Article : Google Scholar : PubMed/NCBI | |
Kang SH, Oh SY, Lee HJ, Kwon TG, Kim JW, Lee ST, Choi SY and Hong SH: Cancer-associated fibroblast subgroups showing differential promoting effect on HNSCC progression. Cancers (Basel). 13:6542021. View Article : Google Scholar : PubMed/NCBI | |
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 : | |
Caligiuri G and Tuveson DA: Activated fibroblasts in cancer: Perspectives and challenges. Cancer Cell. 41:434–449. 2023. View Article : Google Scholar : PubMed/NCBI | |
Öhlund 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 | |
Elyada E, Bolisetty M, Laise P, Flynn WF, Courtois ET, Burkhart RA, Teinor JA, Belleau P, Biffi G, Lucito MS, et al: Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 9:1102–1123. 2019. View Article : Google Scholar : PubMed/NCBI | |
Costa A, Kieffer Y, Scholer-Dahirel A, Pelon F, Bourachot B, Cardon M, Sirven P, Magagna I, Fuhrmann L, Bernard C, et al: Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell. 33:463–479.e10. 2018. View Article : Google Scholar : PubMed/NCBI | |
Shi T, Yao L, Han Y, Hao P and Lu P: Quantitative phosphoproteomics reveals system-wide phosphorylation network altered by spry in mouse mammary stromal fibroblasts. Int J Mol Sci. 20:54002019. View Article : Google Scholar : PubMed/NCBI | |
Cords L, Tietscher S, Anzeneder T, Langwieder C, Rees M, de Souza N and Bodenmiller B: Cancer-associated fibroblast classification in single-cell and spatial proteomics data. Nat Commun. 14:42942023. View Article : Google Scholar : PubMed/NCBI | |
Schwörer S, Cimino FV, Ros M, Tsanov KM, Ng C, Lowe SW, Carmona-Fontaine C and Thompson CB: Hypoxia potentiates the inflammatory fibroblast phenotype promoted by pancreatic cancer cell-derived cytokines. Cancer Res. 83:1596–1610. 2023. View Article : Google Scholar : PubMed/NCBI | |
Biffi G, Oni TE, Spielman B, Hao Y, Elyada E, Park Y, Preall J and Tuveson DA: IL1-induced JAK/STAT signaling is antagonized by TGFβ to shape CAF heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov. 9:282–301. 2019. View Article : Google Scholar | |
Huang J, Zhang L, Wan D, Zhou L, Zheng S, Lin S and Qiao Y: Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct Target Ther. 6:1532021. View Article : Google Scholar : PubMed/NCBI | |
Piersma B, Hayward MK and Weaver VM: Fibrosis and cancer: A strained relationship. Biochim Biophys Acta Rev Cancer. 1873:1883562020. View Article : Google Scholar : PubMed/NCBI | |
Naba A, Clauser KR, Ding H, Whittaker CA, Carr SA and Hynes RO: The extracellular matrix: Tools and insights for the 'omics' era. Matrix Biol. 49:10–24. 2016. View Article : Google Scholar | |
Zeltz C, Primac I, Erusappan P, Alam J, Noel A and Gullberg D: Cancer-associated fibroblasts in desmoplastic tumors: Emerging role of integrins. Semin Cancer Biol. 62:166–181. 2020. View Article : Google Scholar | |
Santi A, Kugeratski FG and Zanivan S: Cancer associated fibroblasts: The architects of stroma remodeling. Proteomics. 18:e17001672018. View Article : Google Scholar | |
Najafi M, Farhood B and Mortezaee K: Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J Cell Biochem. 120:2782–2790. 2019. View Article : Google Scholar | |
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 | |
Patwardhan S, Mahadik P, Shetty O and Sen S: ECM stiffness-tuned exosomes drive breast cancer motility through thrombospondin-1. Biomaterials. 279:1211852021. View Article : Google Scholar : PubMed/NCBI | |
Wei SC, Fattet L, Tsai JH, Guo Y, Pai VH, Majeski HE, Chen AC, Sah RL, Taylor SS, Engler AJ and Yang J: Matrix stiffness drives epithelial-mesenchymal transition and tumour metastasis through a TWIST1-G3BP2 mechanotransduction pathway. Nat Cell Biol. 17:678–688. 2015. View Article : Google Scholar : PubMed/NCBI | |
Jiang Y, Zhang H, Wang J, Liu Y, Luo T and Hua H: Targeting extracellular matrix stiffness and mechanotransducers to improve cancer therapy. J Hematol Oncol. 15:342022. View Article : Google Scholar : PubMed/NCBI | |
Li ZL, Wang ZJ, Wei GH, Yang Y and Wang XW: Changes in extracellular matrix in different stages of colorectal cancer and their effects on proliferation of cancer cells. World J Gastrointest Oncol. 12:267–275. 2020. View Article : Google Scholar : PubMed/NCBI | |
Erdogan B and Webb DJ: Cancer-associated fibroblasts modulate growth factor signaling and extracellular matrix remodeling to regulate tumor metastasis. Biochem Soc Trans. 45:229–236. 2017. View Article : Google Scholar : PubMed/NCBI | |
Jabłońska-Trypuć A, Matejczyk M and Rosochacki S: Matrix metalloproteinases (MMPs), the main extracellular matrix (ECM) enzymes in collagen degradation, as a target for anticancer drugs. J Enzyme Inhib Med Chem. 31:177–183. 2016. View Article : Google Scholar | |
Cazet AS, Hui MN, Elsworth BL, Wu SZ, Roden D, Chan CL, Skhinas JN, Collot R, Yang J, Harvey K, et al: Targeting stromal remodeling and cancer stem cell plasticity overcomes chemoresistance in triple negative breast cancer. Nat Commun. 9:28972018. View Article : Google Scholar : PubMed/NCBI | |
Shi Y, Gao W, Lytle NK, Huang P, Yuan X, Dann AM, Ridinger-Saison M, DelGiorno KE, Antal CE, Liang G, et al: Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring. Nature. 569:131–135. 2019. View Article : Google Scholar : PubMed/NCBI | |
Kaur A, Ecker BL, Douglass SM, Kugel CH III, Webster MR, Almeida FV, Somasundaram R, Hayden J, Ban E, Ahmadzadeh H, et al: Remodeling of the collagen matrix in aging skin promotes melanoma metastasis and affects immune cell motility. Cancer Discov. 9:64–81. 2019. View Article : Google Scholar : | |
Zhang X, Dong Y, Zhao M, Ding L, Yang X, Jing Y, Song Y, Chen S, Hu Q and Ni Y: ITGB2-mediated metabolic switch in CAFs promotes OSCC proliferation by oxidation of NADH in mitochondrial oxidative phosphorylation system. Theranostics. 10:12044–12059. 2020. View Article : Google Scholar : PubMed/NCBI | |
Fang Z, Xu J, Zhang B, Wang W, Liu J, Liang C, Hua J, Meng Q, Yu X and Shi S: The promising role of noncoding RNAs in cancer-associated fibroblasts: An overview of current status and future perspectives. J Hematol Oncol. 13:1542020. View Article : Google Scholar : PubMed/NCBI | |
Zeng Y, Li B, Liang Y, Reeves PM, Qu X, Ran C, Liu Q, Callahan MV, Sluder AE, Gelfand JA, et al: Dual blockade of CXCL12-CXCR4 and PD-1-PD-L1 pathways prolongs survival of ovarian tumor-bearing mice by prevention of immunosuppression in the tumor microenvironment. FASEB J. 33:6596–6608. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wei R, Li J, Lin W, Pang X, Yang H, Lai S, Wei X, Jiang X, Yuan Y and Yang R: Nanoparticle-mediated blockade of CXCL12/CXCR4 signaling enhances glioblastoma immunotherapy: Monitoring early responses with MRI radiomics. Acta Biomater. 177:414–430. 2024. View Article : Google Scholar : PubMed/NCBI | |
Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, Connell CM, Roberts EW, Zhao Q, Caballero OL, et al: Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci USA. 110:20212–20217. 2013. View Article : Google Scholar : PubMed/NCBI | |
Wang H, Liang Y, Liu Z, Zhang R, Chao J, Wang M, Liu M, Qiao L, Xuan Z, Zhao H and Lu L: POSTN+ cancer-associated fibroblasts determine the efficacy of immunotherapy in hepatocellular carcinoma. J Immunother Cancer. 12:e0087212024. View Article : Google Scholar : | |
Chen C, Guo Q, Liu Y, Hou Q, Liao M, Guo Y, Zang Y, Wang F, Liu H, Luan X, et al: Single-cell and spatial transcriptomics reveal POSTN+ cancer-associated fibroblasts correlated with immune suppression and tumour progression in non-small cell lung cancer. Clin Transl Med. 13:e15152023. View Article : Google Scholar | |
Li Z, Sun C and Qin Z: Metabolic reprogramming of cancer-associated fibroblasts and its effect on cancer cell reprogramming. Theranostics. 11:8322–8336. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhang S, Wang J, Chen Y, Liang W, Liu H, Du R, Sun Y, Hu C and Shang Z: CAFs-derived lactate enhances the cancer stemness through inhibiting the MST1 ubiquitination degradation in OSCC. Cell Biosci. 14:1442024. View Article : Google Scholar : PubMed/NCBI | |
Bertero T, Oldham WM, Grasset EM, Bourget I, Boulter E, Pisano S, Hofman P, Bellvert F, Meneguzzi G, Bulavin DV, et al: Tumor-stroma mechanics coordinate amino acid availability to sustain tumor growth and malignancy. Cell Metab. 29:124–140.e10. 2019. View Article : Google Scholar : | |
Kim I, Choi S, Yoo S, Lee M and Kim IS: Cancer-associated fibroblasts in the hypoxic tumor microenvironment. Cancers (Basel). 14:33212022. View Article : Google Scholar : PubMed/NCBI | |
Wei X, Chen Y, Jiang X, Peng M, Liu Y, Mo Y, Ren D, Hua Y, Yu B, Zhou Y, et al: Mechanisms of vasculogenic mimicry in hypoxic tumor microenvironments. Mol Cancer. 20:72021. View Article : Google Scholar : PubMed/NCBI | |
Cadamuro M, Brivio S, Mertens J, Vismara M, Moncsek A, Milani C, Fingas C, Cristina Malerba M, Nardo G, Dall'Olmo L, et al: Platelet-derived growth factor-D enables liver myofibroblasts to promote tumor lymphangiogenesis in cholangiocarcinoma. J Hepatol. 70:700–709. 2019. View Article : Google Scholar | |
Li P, Zhang H, Chen T, Zhou Y, Yang J and Zhou J: Cancer-associated fibroblasts promote proliferation, angiogenesis, metastasis and immunosuppression in gastric cancer. Matrix Biol. 132:59–71. 2024. View Article : Google Scholar : PubMed/NCBI | |
Dai S, Liu Y, Liu Z, Li R, Luo F, Li Y, Dai L and Peng X: Cancer-associated fibroblasts mediate resistance to anti-EGFR therapies in cancer. Pharmacol Res. 206:1073042024. View Article : Google Scholar : PubMed/NCBI | |
Kobayashi H, Enomoto A, Woods SL, Burt AD, Takahashi M and Worthley DL: Cancer-associated fibroblasts in gastrointestinal cancer. NaNat Rev Gastroenterol Hepatol. 16:282–295. 2019. View Article : Google Scholar | |
Mucciolo G, Araos Henríquez J, Jihad M, Pinto Teles S, Manansala JS, Li W, Ashworth S, Lloyd EG, Cheng PSW, Luo W, et al: EGFR-activated myofibroblasts promote metastasis of pancreatic cancer. Cancer Cell. 42:101–118.e11. 2024. View Article : Google Scholar : PubMed/NCBI | |
Bordignon P, Bottoni G, Xu X, Popescu AS, Truan Z, Guenova E, Kofler L, Jafari P, Ostano P, Röcken M, et al: Dualism of FGF and TGF-β signaling in heterogeneous cancer-associated fibroblast activation with ETV1 as a critical determinant. Cell Rep. 28:2358–2372.e6. 2019. View Article : Google Scholar | |
Likonen D, Pinchasi M, Beery E, Sarsor Z, Signorini LF, Gervits A, Sharan R, Lahav M, Raanani P and Uziel O: Exosomal telomerase transcripts reprogram the microRNA transcriptome profile of fibroblasts and partially contribute to CAF formation. Sci Rep. 12:164152022. View Article : Google Scholar : PubMed/NCBI | |
Tang XH, Guo T, Gao XY, Wu XL, Xing XF, Ji JF and Li ZY: Exosome-derived noncoding RNAs in gastric cancer: Functions and clinical applications. Mol Cancer. 20:992021. View Article : Google Scholar : PubMed/NCBI | |
Cosentino G, Romero-Cordoba S, Plantamura I, Cataldo A and Iorio MV: miR-9-mediated inhibition of EFEMP1 contributes to the acquisition of pro-tumoral properties in normal fibroblasts. Cells. 9:21432020. View Article : Google Scholar : PubMed/NCBI | |
Li J, Guan J, Long X, Wang Y and Xiang X: mir-1-mediated paracrine effect of cancer-associated fibroblasts on lung cancer cell proliferation and chemoresistance. Oncol Rep. 35:3523–3531. 2016. View Article : Google Scholar : PubMed/NCBI | |
Zhang L, Yao J, Li W and Zhang C: Micro-RNA-21 regulates cancer-associated fibroblast-mediated drug resistance in pancreatic cancer. Oncol Res. 26:827–835. 2018. View Article : Google Scholar | |
Xia B, Gu X, Xu T, Yan M, Huang L, Jiang C, Li M, Zhai G, Zhang G, Wu J, et al: Exosomes-mediated transfer of LINC00691 regulates the formation of CAFs and promotes the progression of gastric cancer. BMC Cancer. 23:9282023. View Article : Google Scholar | |
Pang W, Su J, Wang Y, Feng H, Dai X, Yuan Y, Chen X and Yao W: Pancreatic cancer-secreted miR-155 implicates in the conversion from normal fibroblasts to cancer-associated fibroblasts. Cancer Sci. 106:1362–1369. 2015. View Article : Google Scholar : PubMed/NCBI | |
Li F, Zhang F, Wang T, Xie Z, Luo H, Dong W, Zhang J, Ren C and Peng W: A self-amplifying loop of TP53INP1 and P53 drives oxidative stress-induced apoptosis of bone marrow mesenchymal stem cells. Apoptosis. 29:882–897. 2024. View Article : Google Scholar : PubMed/NCBI | |
Vennin C, Mélénec P, Rouet R, Nobis M, Cazet AS, Murphy KJ, Herrmann D, Reed DA, Lucas MC, Warren SC, et al: CAF hierarchy driven by pancreatic cancer cell p53-status creates a pro-metastatic and chemoresistant environment via perlecan. Nat Commun. 10:36372019. View Article : Google Scholar : PubMed/NCBI | |
Fang T, Lv H, Lv G, Li T, Wang C, Han Q, Yu L, Su B, Guo L, Huang S, et al: Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat Commun. 9:1912018. View Article : Google Scholar : PubMed/NCBI | |
Zhou Y, Ren H, Dai B, Li J, Shang L, Huang J and Shi X: Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. J Exp Clin Cancer Res. 37:3242018. View Article : Google Scholar : PubMed/NCBI | |
Ye B, Duan Y, Zhou M, Wang Y, Lai Q, Yue K, Cao J, Wu Y, Wang X and Jing C: Hypoxic tumor-derived exosomal miR-21 induces cancer-associated fibroblast activation to promote head and neck squamous cell carcinoma metastasis. Cell Signal. 108:1107252023. View Article : Google Scholar : PubMed/NCBI | |
Hu T and Hu J: Melanoma-derived exosomes induce reprogramming fibroblasts into cancer-associated fibroblasts via Gm26809 delivery. Cell Cycle. 18:3085–3094. 2019. View Article : Google Scholar : PubMed/NCBI | |
Dror S, Sander L, Schwartz H, Sheinboim D, Barzilai A, Dishon Y, Apcher S, Golan T, Greenberger S, Barshack I, et al: Melanoma miRNA trafficking controls tumour primary niche formation. Nat Cell Biol. 18:1006–1017. 2016. View Article : Google Scholar : PubMed/NCBI | |
Vu LT, Peng B, Zhang DX, Ma V, Mathey-Andrews CA, Lam CK, Kiomourtzis T, Jin J, McReynolds L, Huang L, et al: Tumor-secreted extracellular vesicles promote the activation of cancer-associated fibroblasts via the transfer of microRNA-125b. J Extracell Vesicles. 8:15996802019. 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 | |
Baroni S, Romero-Cordoba S, Plantamura I, Dugo M, D'Ippolito E, Cataldo A, Cosentino G, Angeloni V, Rossini A, Daidone MG and Iorio MV: Exosome-mediated delivery of miR-9 induces cancer-associated fibroblast-like properties in human breast fibroblasts. Cell Death Dis. 7:e23122016. View Article : Google Scholar : PubMed/NCBI | |
Tong Y, Yang L, Yu C, Zhu W, Zhou X, Xiong Y, Wang W, Ji F, He D and Cao X: Tumor-secreted exosomal lncRNA POU3F3 promotes cisplatin resistance in ESCC by inducing fibroblast differentiation into CAFs. Mol Ther Oncolytics. 18:1–13. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wang D, Wang X, Song Y, Si M, Sun Y, Liu X, Cui S, Qu X and Yu X: Exosomal miR-146a-5p and miR-155-5p promote CXCL12/CXCR7-induced metastasis of colorectal cancer by crosstalk with cancer-associated fibroblasts. Cell Death Dis. 13:3802022. View Article : Google Scholar : PubMed/NCBI | |
Becker LM, O'Connell JT, Vo AP, Cain MP, Tampe D, Bizarro L, Sugimoto H, McGow AK, Asara JM, Lovisa S, et al: Epigenetic reprogramming of cancer-associated fibroblasts deregulates glucose metabolism and facilitates progression of breast cancer. Cell Rep. 31:1077012020. View Article : Google Scholar : PubMed/NCBI | |
Pavlides S, Whitaker-Menezes D, Castello-Cros R, Flomenberg N, Witkiewicz AK, Frank PG, Casimiro MC, Wang C, Fortina P, Addya S, et al: The reverse Warburg effect: Aerobic glycolysis in cancer associated fibroblasts and the tumor stroma. Cell Cycle. 8:3984–4001. 2009. View Article : Google Scholar : PubMed/NCBI | |
Kitamura F, Semba T, Yasuda-Yoshihara N, Yamada K, Nishimura A, Yamasaki J, Nagano O, Yasuda T, Yonemura A, Tong Y, et al: Cancer-associated fibroblasts reuse cancer-derived lactate to maintain a fibrotic and immunosuppressive microenvironment in pancreatic cancer. JCI Insight. 8:e1630222023. View Article : Google Scholar : PubMed/NCBI | |
Zhang C, Zhou W, Xu H, Xu J, Li J, Liu X, Lu X, Dai J, Jiang Y, Wang W, et al: Cancer-associated fibroblasts promote EGFR-TKI resistance via the CTHRC1/glycolysis/H3K18la positive feedback loop. Oncogene. 44:1400–1414. 2025. View Article : Google Scholar : PubMed/NCBI | |
Yan W, Wu X, Zhou W, Fong MY, Cao M, Liu J, Liu X, Chen CH, Fadare O, Pizzo DP, et al: Cancer-cell-secreted exosomal miR-105 promotes tumour growth through the MYC-dependent metabolic reprogramming of stromal cells. Nat Cell Biol. 20:597–609. 2018. View Article : Google Scholar : PubMed/NCBI | |
He R, Hu C, Yuan Y, Li T, Tian Q, Huang T, Lin Q, Zheng S, Chen C, Fu Z and Chen R: Glycolysis reprogramming in CAFs promotes oxaliplatin resistance in pancreatic cancer through circABCC4 mediated PKM2 nuclear translocation. Cell Death Dis. 16:1262025. 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 | |
Long X, Xiong W, Zeng X, Qi L, Cai Y, Mo M, Jiang H, Zhu B, Chen Z and Li Y: Cancer-associated fibroblasts promote cisplatin resistance in bladder cancer cells by increasing IGF-1/ERβ/Bcl-2 signalling. Cell Death Dis. 10:3752019. View Article : Google Scholar | |
Zhang T, Zhang P and Li HX: CAFs-derived exosomal miRNA-130a confers cisplatin resistance of NSCLC cells through PUM2-dependent packaging. Int J Nanomedicine. 16:561–577. 2021. View Article : Google Scholar : PubMed/NCBI | |
Yip HYK and Papa A: Signaling pathways in cancer: Therapeutic targets, combinatorial treatments, and new developments. Cells. 10:6592021. View Article : Google Scholar : PubMed/NCBI | |
Zhang YE: Non-smad signaling pathways of the TGF-β family. Cold Spring Harb Perspect Biol. 9:a0221292017. View Article : Google Scholar | |
Moon JY, Manh Hung LV, Unno T and Cho SK: Nobiletin enhances chemosensitivity to adriamycin through modulation of the Akt/GSK3β/β-catenin/MYCN/MRP1 signaling pathway in A549 human non-small-cell lung cancer cells. Nutrients. 10:18292018. View Article : Google Scholar | |
Tomar VS, Patil V and Somasundaram K: Temozolomide induces activation of Wnt/β-catenin signaling in glioma cells via PI3K/Akt pathway: Implications in glioma therapy. Cell Biol Toxicol. 36:273–278. 2020. View Article : Google Scholar | |
Sun Z, Jiang Q, Gao B, Zhang X, Bu L, Wang L, Lin Y, Xie W, Li J and Guo J: AKT blocks SIK1-mediated repression of STAT3 to promote breast tumorigenesis. Cancer Res. 83:1264–1279. 2023. View Article : Google Scholar : PubMed/NCBI | |
Belarif L, Mary C, Jacquemont L, Mai HL, Danger R, Hervouet J, Minault D, Thepenier V, Nerrière-Daguin V, Nguyen E, et al: IL-7 receptor blockade blunts antigen-specific memory T cell responses and chronic inflammation in primates. Nat Commun. 9:44832018. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Jiang W, Du Y, Zhu D, Zhang J, Fang C, Yan F and Chen ZS: Targeting feedback activation of signaling transduction pathways to overcome drug resistance in cancer. Drug Resist Updat. 65:1008842022. View Article : Google Scholar : PubMed/NCBI | |
Fang Z, Meng Q, Xu J, Wang W, Zhang B, Liu J, Liang C, Hua J, Zhao Y, Yu X and Shi S: Signaling pathways in cancer-associated fibroblasts: Recent advances and future perspectives. Cancer Commun (Lond). 43:3–41. 2023. View Article : Google Scholar | |
Butti R, Khaladkar A, Bhardwaj P and Prakasam G: Heterotypic signaling of cancer-associated fibroblasts in shaping the cancer cell drug resistance. Cancer Drug Resist. 6:182–204. 2023. View Article : Google Scholar : PubMed/NCBI | |
Keresztes D, Kerestély M, Szarka L, Kovács BM, Schulc K, Veres DV and Csermely P: Cancer drug resistance as learning of signaling networks. Biomed Pharmacother. 183:1178802025. View Article : Google Scholar : PubMed/NCBI | |
Xue W, Yang L, Chen C, Ashrafizadeh M, Tian Y and Sun R: Wnt/β-catenin-driven EMT regulation in human cancers. Cell Mol Life Sci. 81:792024. View Article : Google Scholar | |
Zhao H, Ming T, Tang S, Ren S, Yang H, Liu M, Tao Q and Xu H: Wnt signaling in colorectal cancer: Pathogenic role and therapeutic target. Mol Cancer. 21:1442022. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Zheng L, Shang W, Yang Z, Li T, Liu F, Shao W, Lv L, Chai L, Qu L, et al: Wnt/beta-catenin signaling confers ferroptosis resistance by targeting GPX4 in gastric cancer. Cell Death Differ. 29:2190–2202. 2022. View Article : Google Scholar : PubMed/NCBI | |
Essex A, Pineda J, Acharya G, Xin H and Evans J; Reproducibility Project: Cancer Biology: Replication study: Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. ELife. 8:e454262019. View Article : Google Scholar | |
Ren J, Ding L, Zhang D, Shi G, Xu Q, Shen S, Wang Y, Wang T and Hou Y: Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 8:3932–3948. 2018. View Article : Google Scholar : PubMed/NCBI | |
Deng X, Ruan H, Zhang X, Xu X, Zhu Y, Peng H, Zhang X, Kong F and Guan M: Long noncoding RNA CCAL transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells. Int J Cancer. 146:1700–1716. 2020. View Article : Google Scholar | |
Shan G, Zhou X, Gu J, Zhou D, Cheng W, Wu H, Wang Y, Tang T and Wang X: Downregulated exosomal microRNA-148b-3p in cancer associated fibroblasts enhance chemosensitivity of bladder cancer cells by downregulating the Wnt/β-catenin pathway and upregulating PTEN. Cell Oncol (Dordr). 44:45–59. 2021. View Article : Google Scholar : PubMed/NCBI | |
Cao L, Weng K, Li L, Lin G, Zhao Y, Gao Y, Huang X, Chen Q, Wang J, Zheng C, et al: BATF2 inhibits the stem cell-like properties and chemoresistance of gastric cancer cells through PTEN/AKT/β-catenin pathway. Theranostics. 14:7007–7022. 2024. View Article : Google Scholar : | |
Fang F, Guo C, Zheng W, Wang Q and Zhou L: Exosome-mediated transfer of miR-1323 from cancer-associated fibroblasts confers radioresistance of C33A cells by targeting PABPN1 and activating Wnt/β-catenin signaling pathway in cervical cancer. Reprod Sci. 29:1809–1821. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zou S, Tong Q, Liu B, Huang W, Tian Y and Fu X: Targeting STAT3 in cancer immunotherapy. Mol Cancer. 19:1452020. View Article : Google Scholar : PubMed/NCBI | |
Hu Y, Dong Z and Liu K: Unraveling the complexity of STAT3 in cancer: Molecular understanding and drug discovery. J Exp Clin Cancer Res. 43:232024. View Article : Google Scholar : PubMed/NCBI | |
Pan MS, Wang H, Ansari KH, Li XP, Sun W and Fan YZ: Gallbladder cancer-associated fibroblasts promote vasculogenic mimicry formation and tumor growth in gallbladder cancer via upregulating the expression of NOX4, a poor prognosis factor, through IL-6-JAK-STAT3 signal pathway. J Exp Clin Cancer Res. 39:2342020. View Article : Google Scholar : PubMed/NCBI | |
Yu Z, Zhang Q, Wei S, Zhang Y, Zhou T, Zhang Q, Shi R, Zinovkin D, Pranjol ZI, Zhang J and Wang H: CD146+CAFs promote progression of endometrial cancer by inducing angiogenesis and vasculogenic mimicry via IL-10/JAK1/STAT3 pathway. Cell Commun Signal. 22:1702024. View Article : Google Scholar | |
Tao L, Huang G, Wang R, Pan Y, He Z, Chu X, Song H and Chen L: Cancer-associated fibroblasts treated with cisplatin facilitates chemoresistance of lung adenocarcinoma through IL-11/IL-11R/STAT3 signaling pathway. Sci Rep. 6:384082016. View Article : Google Scholar : PubMed/NCBI | |
Li J, Wang Y, Wang Z, Wei Y, Diao P, Wu Y, Wang D, Jiang H, Wang Y and Cheng J: Super-enhancer driven LIF/LIFR-STAT3-SOX2 regulatory feedback loop promotes cancer stemness in head and neck squamous cell carcinoma. Adv Sci (Weinh). 11:e24044762024. View Article : Google Scholar : PubMed/NCBI | |
Hu C, Xia R, Zhang X, Li T, Ye Y, Li G, He R, Li Z, Lin Q, Zheng S and Chen R: circFARP1 enables cancer-associated fibroblasts to promote gemcitabine resistance in pancreatic cancer via the LIF/STAT3 axis. Mol Cancer. 21:242022. View Article : Google Scholar : PubMed/NCBI | |
Sun L, Ke M, Yin M, Zeng Y, Ji Y, Hu Y, Fu S and Zhang C: Extracellular vesicle-encapsulated microRNA-296-3p from cancer-associated fibroblasts promotes ovarian cancer development through regulation of the PTEN/AKT and SOCS6/STAT3 pathways. Cancer Sci. 115:155–169. 2024. View Article : Google Scholar | |
Zhao Q, Huang L, Qin G, Qiao Y, Ren F, Shen C, Wang S, Liu S, Lian J, Wang D, et al: Cancer-associated fibroblasts induce monocytic myeloid-derived suppressor cell generation via IL-6/exosomal miR-21-activated STAT3 signaling to promote cisplatin resistance in esophageal squamous cell carcinoma. Cancer Lett. 518:35–48. 2021. View Article : Google Scholar : PubMed/NCBI | |
Cao LQ, Yang XW, Chen YB, Zhang DW, Jiang XF and Xue P: Exosomal miR-21 regulates the TETs/PTENp1/PTEN pathway to promote hepatocellular carcinoma growth. Mol Cancer. 18:1482019. View Article : Google Scholar : PubMed/NCBI | |
Gupta S, Silveira DA, Lorenzoni PR, Mombach JCM and Hashimoto RF: LncRNA PTENP1/miR-21/PTEN axis modulates EMT and drug resistance in cancer: Dynamic boolean modeling for cell fates in DNA damage response. Int J Mol Sci. 25:82642024. View Article : Google Scholar : PubMed/NCBI | |
He Y, Sun MM, Zhang GG, Yang J, Chen KS, Xu WW and Li B: Targeting PI3K/Akt signal transduction for cancer therapy. Signal Transduct Target Ther. 6:4252021. View Article : Google Scholar : PubMed/NCBI | |
Murugan AK: mTOR: Role in cancer, metastasis and drug resistance. Semin Cancer Biol. 59:92–111. 2019. View Article : Google Scholar : PubMed/NCBI | |
Haddadi N, Lin Y, Travis G, Simpson AM, Nassif NT and McGowan EM: PTEN/PTENP1: 'Regulating the regulator of RTK-dependent PI3K/Akt signalling', new targets for cancer therapy. Mol Cancer. 17:372018. View Article : Google Scholar : PubMed/NCBI | |
Shi L, Zhu W, Huang Y, Zhuo L, Wang S, Chen S, Zhang B and Ke B: Cancer-associated fibroblast-derived exosomal microRNA-20a suppresses the PTEN/PI3K-AKT pathway to promote the progression and chemoresistance of non-small cell lung cancer. Clin Transl Med. 12:e9892022. View Article : Google Scholar : PubMed/NCBI | |
Qu Z, Yang KD, Luo BH and Zhang F: CAFs-secreted exosomal cricN4BP2L2 promoted colorectal cancer stemness and chemoresistance by interacting with EIF4A3. Exp Cell Res. 418:1132662022. View Article : Google Scholar : PubMed/NCBI | |
Li L, Liu D, Chen T, Wei C, Qiao Y, Liu W, Liang Y, Liang Z, Chen C, Li D, et al: Hypoxia-enhanced YAP1-EIF4A3 interaction drives circ_0007386 circularization by competing with CRIM1 pre-mRNA linear splicing and promotes non-small cell lung cancer progression. J Exp Clin Cancer Res. 43:2002024. View Article : Google Scholar : PubMed/NCBI | |
Ju C, Zhou M, Du D, Wang C, Yao J, Li H, Luo Y, He F and He J: EIF4A3-mediated circ_0042881 activates the RAS pathway via miR-217/SOS1 axis to facilitate breast cancer progression. Cell Death Dis. 14:5592023. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Pan Q and Shao Z: Extracellular vesicles derived from cancer-associated fibroblasts carry tumor-promotive microRNA-1228-3p to enhance the resistance of hepatocellular carcinoma cells to sorafenib. Hum Cell. 36:296–311. 2023. View Article : Google Scholar | |
Deng K, Zou F, Xu J, Xu D and Luo Z: Cancer-associated fibroblasts promote stemness maintenance and gemcitabine resistance via HIF-1α/miR-21 axis under hypoxic conditions in pancreatic cancer. Mol Carcinog. 63:524–537. 2024. View Article : Google Scholar : PubMed/NCBI | |
Xin X, Cheng X, Zeng F, Xu Q and Hou L: The role of TGF-β/SMAD signaling in hepatocellular carcinoma: From mechanism to therapy and prognosis. Int J Biol Sci. 20:1436–1451. 2024. View Article : Google Scholar : | |
Huang X, Jie S, Li W, Li H, Ni J and Liu C: miR-122-5p targets GREM2 to protect against glucocorticoid-induced endothelial damage through the BMP signaling pathway. Mol Cell Endocrinol. 544:1115412022. View Article : Google Scholar : PubMed/NCBI | |
Shan G, Gu J, Zhou D, Li L, Cheng W, Wang Y, Tang T and Wang X: Cancer-associated fibroblast-secreted exosomal miR-423-5p promotes chemotherapy resistance in prostate cancer by targeting GREM2 through the TGF-β signaling pathway. Exp Mol Med. 52:1809–1822. 2020. View Article : Google Scholar : PubMed/NCBI | |
Leng Z, Li Y, Zhou G, Lv X, Ai W, Li J and Hou L: Krüppel-like factor 4 regulates stemness and mesenchymal properties of colorectal cancer stem cells through the TGF-β1/Smad/snail pathway. J Cell Mol Med. 24:1866–1877. 2020. View Article : Google Scholar | |
Liu T, Jiang L, Bai Q, Wu S, Yu X, Wu T, Wang J, Zhang X, Li H, Zhao K and Wang L: CLDN6 suppresses migration and invasion of MCF-7 and SKBR-3 breast cancer cells by blocking the SMAD/Snail/MMP-2/9 axis. Bull Exp Biol Med. 175:376–381. 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 : | |
Sun J, Du R, Li X, Liu C, Wang D, He X, Li G, Zhang K, Wang S, Hao Q, et al: CD63+ cancer-associated fibroblasts confer CDK4/6 inhibitor resistance to breast cancer cells by exosomal miR-20. Cancer Lett. 588:2167472024. View Article : Google Scholar | |
Fang Y, Zhou W, Rong Y, Kuang T, Xu X, Wu W, Wang D and Lou W: Exosomal miRNA-106b from cancer-associated fibroblast promotes gemcitabine resistance in pancreatic cancer. Exp Cell Res. 383:1115432019. View Article : Google Scholar : PubMed/NCBI | |
Pan S, Deng Y, Fu J, Zhang Y, Zhang Z and Qin X: N6-methyladenosine upregulates miR-181d-5p in exosomes derived from cancer-associated fibroblasts to inhibit 5-FU sensitivity by targeting NCALD in colorectal cancer. Int J Oncol. 60:142022. View Article : Google Scholar : | |
Yang C, Zhang Y, Yan M, Wang J, Wang J, Wang M, Xuan Y, Cheng H, Ma J, Chai C, et al: Exosomes derived from cancer-associated fibroblasts promote tumorigenesis, metastasis and chemoresistance of colorectal cancer by upregulating circ_0067557 to target Lin28. BMC Cancer. 24:642024. View Article : Google Scholar : PubMed/NCBI | |
Luo D, Liang Y, Wang Y, Ye F, Jin Y, Li Y, Han D, Wang Z, Chen B, Zhao W, et al: Long non-coding RNA MIDEAS-AS1 inhibits growth and metastasis of triple-negative breast cancer via transcriptionally activating NCALD. Breast Cancer Res. 25:1092023. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Ratanasirintrawoot S, Chandrasekaran S, Wu Z, Ficarro SB, Yu C, Ross CA, Cacchiarelli D, Xia Q, Seligson M, et al: LIN28 regulates stem cell metabolism and conversion to primed pluripotency. Cell Stem Cell. 19:66–80. 2016. View Article : Google Scholar : PubMed/NCBI | |
Gong W, Guo Y, Yuan H, Chai R, Wan Z, Zheng B, Hu X, Chen B, Gao S, Dai Q, et al: Loss of exosomal miR-200b-3p from hypoxia cancer-associated fibroblasts promotes tumorigenesis and reduces sensitivity to 5-flourouracil in colorectal cancer via upregulation of ZEB1 and E2F3. Cancer Gene Ther. 30:905–916. 2023. View Article : Google Scholar : PubMed/NCBI | |
Obeng E: Apoptosis (programmed cell death) and its signals-a review. Braz J Biol. 81:1133–1143. 2021. View Article : Google Scholar | |
Xu X, Lai Y and Hua ZC: Apoptosis and apoptotic body: Disease message and therapeutic target potentials. Biosci Rep. 39:BSR201809922019. View Article : Google Scholar : | |
Moyer A, Tanaka K and Cheng EH: Apoptosis in cancer biology and therapy. Annu Rev Pathol. 20:303–328. 2025. View Article : Google Scholar : PubMed/NCBI | |
Sahoo G, Samal D, Khandayataray P and Murthy MK: A review on caspases: Key regulators of biological activities and apoptosis. Mol Neurobiol. 60:5805–5837. 2023. View Article : Google Scholar : PubMed/NCBI | |
Singh P and Lim B: Targeting apoptosis in cancer. Curr Oncol Rep. 24:273–284. 2022. View Article : Google Scholar : PubMed/NCBI | |
Au Yeung CL, Co NN, Tsuruga T, Yeung TL, Kwan SY, Leung CS, Li Y, Lu ES, Kwan K, Wong KK, et al: Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat Commun. 7:111502016. View Article : Google Scholar : PubMed/NCBI | |
Zhou Z, Arroum T, Luo X, Kang R, Lee YJ, Tang D, Hüttemann M and Song X: Diverse functions of cytochrome c in cell death and disease. Cell Death Differ. 31:387–404. 2024. View Article : Google Scholar : PubMed/NCBI | |
Sun LH, Tian D, Yang ZC and Li JL: Exosomal miR-21 promotes proliferation, invasion and therapy resistance of colon adenocarcinoma cells through its target PDCD4. Sci Rep. 10:82712020. View Article : Google Scholar : PubMed/NCBI | |
Zhao MY, Wang LM, Liu J, Huang X, Liu J and Zhang YF: MiR-21 suppresses anoikis through targeting PDCD4 and PTEN in human esophageal adenocarcinoma. Curr Med Sci. 38:245–251. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wei X, Wang W, Wang L, Zhang Y, Zhang X, Chen M, Wang F, Yu J, Ma Y and Sun G: MicroRNA-21 induces 5-fluorouracil resistance in human pancreatic cancer cells by regulating PTEN and PDCD4. Cancer Med. 5:693–702. 2016. View Article : Google Scholar : PubMed/NCBI | |
King LE, Hohorst L and García-Sáez AJ: Expanding roles of BCL-2 proteins in apoptosis execution and beyond. J Cell Sci. 136:jcs2607902023. View Article : Google Scholar : PubMed/NCBI | |
O'Neill KL, Huang K, Zhang J, Chen Y and Luo X: Inactivation of prosurvival Bcl-2 proteins activates Bax/Bak through the outer mitochondrial membrane. Genes Dev. 30:973–988. 2016. View Article : Google Scholar : PubMed/NCBI | |
Kang SH, Oh SY, Lee KY, Lee HJ, Kim MS, Kwon TG, Kim JW, Lee ST, Choi SY and Hong SH: Differential effect of cancer-associated fibroblast-derived extracellular vesicles on cisplatin resistance in oral squamous cell carcinoma via miR-876-3p. Theranostics. 14:460–479. 2024. View Article : Google Scholar : PubMed/NCBI | |
Price D, Muterspaugh R, Clegg B, Williams A, Stephens A, Guthrie J, Heyl D and Evans HG: IGFBP-3 blocks hyaluronan-CD44 signaling, leading to increased acetylcholinesterase levels in A549 cell media and apoptosis in a p53-dependent manner. Sci Rep. 10:50832020. View Article : Google Scholar : PubMed/NCBI | |
Wang H, Wang H, Li K, Li S and Sun B: IGFBP-3 Is the key target of sanguinarine in promoting apoptosis in hepatocellular carcinoma. Cancer Manag Res. 12:1007–1015. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wang H, Huang H, Wang L, Liu Y, Wang M, Zhao S, Lu G and Kang X: Cancer-associated fibroblasts secreted miR-103a-3p suppresses apoptosis and promotes cisplatin resistance in non-small cell lung cancer. Aging (Albany NY). 13:14456–14468. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhang HW, Shi Y, Liu JB, Wang HM, Wang PY, Wu ZJ, Li L, Gu LP, Cao PS, Wang GR, et al: Cancer-associated fibroblast-derived exosomal microRNA-24-3p enhances colon cancer cell resistance to MTX by down-regulating CDX2/HEPH axis. J Cell Mol Med. 25:3699–3713. 2021. View Article : Google Scholar : PubMed/NCBI | |
Qin X, Guo H, Wang X, Zhu X, Yan M, Wang X, Xu Q, Shi J, Lu E, Chen W and Zhang J: Exosomal miR-196a derived from cancer-associated fibroblasts confers cisplatin resistance in head and neck cancer through targeting CDKN1B and ING5. Genome Biol. 20:122019. View Article : Google Scholar : PubMed/NCBI | |
Yang H, Villani RM, Wang H, Simpson MJ, Roberts MS, Tang M and Liang X: The role of cellular reactive oxygen species in cancer chemotherapy. J Exp Clin Cancer Res. 37:2662018. View Article : Google Scholar : PubMed/NCBI | |
Zhou X, Tong Y, Yu C, Pu J, Zhu W, Zhou Y, Wang Y, Xiong Y and Sun X: FAP positive cancer-associated fibroblasts promote tumor progression and radioresistance in esophageal squamous cell carcinoma by transferring exosomal lncRNA AFAP1-AS1. Mol Carcinog. 63:1922–1937. 2024. View Article : Google Scholar : PubMed/NCBI | |
Zhang X, Zheng S, Hu C, Li G, Lin H, Xia R, Ye Y, He R, Li Z, Lin Q, et al: Cancer-associated fibroblast-induced lncRNA UPK1A-AS1 confers platinum resistance in pancreatic cancer via efficient double-strand break repair. Oncogene. 41:2372–2389. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zhang C, Liu X, Jin S, Chen Y and Guo R: Ferroptosis in cancer therapy: A novel approach to reversing drug resistance. Mol Cancer. 21:472022. View Article : Google Scholar : PubMed/NCBI | |
Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI | |
Tang L, Zhang Y, Qian Z and Shen X: The mechanism of Fe(2+)-initiated lipid peroxidation in liposomes: The dual function of ferrous ions, the roles of the pre-existing lipid peroxides and the lipid peroxyl radical. Biochem J. 352:27–36. 2000. View Article : Google Scholar : PubMed/NCBI | |
Dixon SJ and Stockwell BR: The role of iron and reactive oxygen species in cell death. Nat Chem Biol. 10:9–17. 2014. View Article : Google Scholar | |
Maiorino M, Conrad M and Ursini F: GPx4, lipid peroxidation, and cell death: Discoveries, rediscoveries, and open issues. Antioxid Redox Signal. 29:61–74. 2018. View Article : Google Scholar | |
Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB, et al: Regulation of ferroptotic cancer cell death by GPX4. Cell. 156:317–331. 2014. View Article : Google Scholar : PubMed/NCBI | |
Li J, Li Y, Wang D, Liao R and Wu Z: PLAG1 interacts with GPX4 to conquer vulnerability to sorafenib induced ferroptosis through a PVT1/miR-195-5p axis-dependent manner in hepatocellular carcinoma. J Exp Clin Cancer Res. 43:1432024. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Li J, Kang R, Klionsky DJ and Tang D: Ferroptosis: Machinery and regulation. Autophagy. 17:2054–2081. 2021. View Article : Google Scholar : | |
Zhao J, Yang S, Lv C and Liu Y: Cancer-associated fibroblasts suppressed ferroptosis in glioblastoma via upregulating lncRNA DLEU1. Am J Physiol Cell Physiol. 324:C1039–C1052. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Liu Y, Du T, Yang H, Lei L, Guo M, Ding HF, Zhang J, Wang H, Chen X and Yan C: ATF3 promotes erastin-induced ferroptosis by suppressing system Xc. Cell Death Differ. 27:662–675. 2020. View Article : Google Scholar | |
Qu X, Liu B, Wang L, Liu L, Zhao W, Liu C, Ding J, Zhao S, Xu B, Yu H, et al: Loss of cancer-associated fibroblast-derived exosomal DACT3-AS1 promotes malignant transformation and ferroptosis-mediated oxaliplatin resistance in gastric cancer. Drug Resist Updat. 68:1009362023. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Wang Z, Li C, Zhang Z, Lu S, Wang X, Liang Q, Zhu X, Pan C, Wang Q, et al: SIRT1 activated by AROS sensitizes glioma cells to ferroptosis via induction of NAD+ depletion-dependent activation of ATF3. Redox Biol. 69:1030302024. View Article : Google Scholar : PubMed/NCBI | |
Zhang T, Yao C, Zhou X, Liu S, Qi L, Zhu S, Zhao C, Hu D and Shen W: Glutathione-degrading enzymes in the complex landscape of tumors (review). Int J Oncol. 65:722024. View Article : Google Scholar : | |
Ma XH, Liu JH, Liu CY, Sun WY, Duan WJ, Wang G, Kurihara H, He RR, Li YF, Chen Y and Shang H: ALOX15-launched PUFA-phospholipids peroxidation increases the susceptibility of ferroptosis in ischemia-induced myocardial damage. Signal Transduct Target Ther. 7:2882022. View Article : Google Scholar : PubMed/NCBI | |
Ding K, Liu C, Li L, Yang M, Jiang N, Luo S and Sun L: Acyl-CoA synthase ACSL4: An essential target in ferroptosis and fatty acid metabolism. Chin Med J (Engl). 136:2521–2537. 2023. View Article : Google Scholar : PubMed/NCBI | |
Zhang H, Deng T, Liu R, Ning T, Yang H, Liu D, Zhang Q, Lin D, Ge S, Bai M, et al: CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Mol Cancer. 19:432020. View Article : Google Scholar : PubMed/NCBI | |
Pan G, Liu Y, Shang L, Zhou F and Yang S: EMT-associated microRNAs and their roles in cancer stemness and drug resistance. Cancer Commun (Lond). 41:199–217. 2021. View Article : Google Scholar : PubMed/NCBI | |
Du B and Shim JS: Targeting epithelial-mesenchymal transition (EMT) to overcome drug resistance in cancer. Molecules. 21:9652016. View Article : Google Scholar : PubMed/NCBI | |
Erin N, Grahovac J, Brozovic A and Efferth T: Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance. Drug Resist Updat. 53:1007152020. 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 | |
Li N, Babaei-Jadidi R, Lorenzi F, Spencer-Dene B, Clarke P, Domingo E, Tulchinsky E, Vries RGJ, Kerr D, Pan Y, et al: An FBXW7-ZEB2 axis links EMT and tumour microenvironment to promote colorectal cancer stem cells and chemoresistance. Oncogenesis. 8:132019. View Article : Google Scholar : PubMed/NCBI | |
Han G, Wu D, Yang Y, Li Z, Zhang J and Li C: CrkL meditates CCL20/CCR6-induced EMT in gastric cancer. Cytokine. 76:163–169. 2015. View Article : Google Scholar : PubMed/NCBI | |
Hu JH, Tang HN and Wang YH: Cancer-associated fibroblast exosome LINC00355 promotes epithelial-mesenchymal transition and chemoresistance in colorectal cancer through the miR-34b-5p/CRKL axis. Cancer Gene Ther. 31:259–272. 2024. View Article : Google Scholar | |
Zhang Y, Yin C, Wei C, Xia S, Qiao Z, Zhang XW, Yu B, Zhou J and Wang R: Exosomal miR-625-3p secreted by cancer-associated fibroblasts in colorectal cancer promotes EMT and chemotherapeutic resistance by blocking the CELF2/WWOX pathway. Pharmacol Res. 186:1065342022. View Article : Google Scholar : PubMed/NCBI | |
Khawaled S, Nigita G, Distefano R, Oster S, Suh SS, Smith Y, Khalaileh A, Peng Y, Croce CM, Geiger T, et al: Pleiotropic tumor suppressor functions of WWOX antagonize metastasis. Signal Transduct Target Ther. 5:432020. View Article : Google Scholar : PubMed/NCBI | |
Kadioglu O, Saeed MEM, Munder M, Spuller A, Greten HJ and Efferth T: Effect of ABC transporter expression and mutational status on survival rates of cancer patients. Biomed Pharmacother. 131:1107182020. View Article : Google Scholar : PubMed/NCBI | |
To KKW, Huang Z, Zhang H, Ashby CR Jr and Fu L: Utilizing non-coding RNA-mediated regulation of ATP binding cassette (ABC) transporters to overcome multidrug resistance to cancer chemotherapy. Drug Resist Updat. 73:1010582024. View Article : Google Scholar : PubMed/NCBI | |
Luo G, Zhang Y, Wu Z, Zhang L, Liang C and Chen X: Exosomal LINC00355 derived from cancer-associated fibroblasts promotes bladder cancer cell resistance to cisplatin by regulating miR-34b-5p/ABCB1 axis. Acta Biochim Biophys Sin (Shanghai). 53:558–566. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhang D, Ding L, Li Y, Ren J, Shi G, Wang Y, Zhao S, Ni Y and Hou Y: Midkine derived from cancer-associated fibroblasts promotes cisplatin-resistance via up-regulation of the expression of lncRNA ANRIL in tumour cells. Sci Rep. 7:162312017. View Article : Google Scholar : PubMed/NCBI | |
Kunou S, Shimada K, Takai M, Sakamoto A, Aoki T, Hikita T, Kagaya Y, Iwamoto E, Sanada M, Shimada S, et al: Exosomes secreted from cancer-associated fibroblasts elicit anti-pyrimidine drug resistance through modulation of its transporter in malignant lymphoma. Oncogene. 40:3989–4003. 2021. View Article : Google Scholar : PubMed/NCBI | |
Cheng SH, Chiou HC, Wang JW and Lin MH: Reciprocal regulation of cancer-associated fibroblasts and tumor microenvironment in gastrointestinal cancer: Implications for cancer dormancy. Cancers (Basel). 15:25132023. View Article : Google Scholar : PubMed/NCBI | |
Hulpke S and Tampé R: The MHC I loading complex: A multitasking machinery in adaptive immunity. Trends Biochem Sci. 38:412–420. 2013. View Article : Google Scholar : PubMed/NCBI | |
Yao H, Huang C, Zou J, Liang W, Zhao Y, Yang K, Zhong Z, Zhou S, Li J, Li Y, et al: Extracellular vesicle-packaged lncRNA from cancer-associated fibroblasts promotes immune evasion by downregulating HLA-A in pancreatic cancer. J Extracell Vesicles. 13:e124842024. View Article : Google Scholar : PubMed/NCBI | |
Jiang Y, Wang K, Lu X, Wang Y and Chen J: Cancer-associated fibroblasts-derived exosomes promote lung cancer progression by OIP5-AS1/miR-142-5p/ PD-L1 axis. Mol Immunol. 140:47–58. 2021. View Article : Google Scholar : PubMed/NCBI | |
Boussiotis VA: Molecular and biochemical aspects of the PD-1 checkpoint pathway. N Engl J Med. 375:1767–1778. 2016. View Article : Google Scholar : PubMed/NCBI | |
Shintani Y, Fujiwara A, Kimura T, Kawamura T, Funaki S, Minami M and Okumura M: IL-6 Secreted from cancer-associated fibroblasts mediates chemoresistance in NSCLC by increasing epithelial-mesenchymal transition signaling. J Thorac Oncol. 11:1482–1492. 2016. View Article : Google Scholar : PubMed/NCBI | |
Cheteh EH, Sarne V, Ceder S, Bianchi J, Augsten M, Rundqvist H, Egevad L, Östman A and Wiman KG: Interleukin-6 derived from cancer-associated fibroblasts attenuates the p53 response to doxorubicin in prostate cancer cells. Cell Death Discov. 6:422020. View Article : Google Scholar : PubMed/NCBI | |
Lau EY, Lo J, Cheng BY, Ma MK, Lee JM, Ng JK, Chai S, Lin CH, Tsang SY, Ma S, et al: Cancer-associated fibroblasts regulate tumor-initiating cell plasticity in hepatocellular carcinoma through c-Met/FRA1/HEY1 signaling. Cell Rep. 15:1175–1189. 2016. View Article : Google Scholar : PubMed/NCBI | |
Deying W, Feng G, Shumei L, Hui Z, Ming L and Hongqing W: CAF-derived HGF promotes cell proliferation and drug resistance by up-regulating the c-Met/PI3K/Akt and GRP78 signalling in ovarian cancer cells. Biosci Rep. 37:BSR201604702017. View Article : Google Scholar : PubMed/NCBI | |
Wang B, Liu W, Liu C, Du K, Guo Z, Zhang G, Huang Z, Lin S, Cen B, Tian Y, et al: Cancer-associated fibroblasts promote radioresistance of breast cancer cells via the HGF/c-met signaling pathway. Int J Radiat Oncol Biol Phys. 116:640–654. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wei L, Ye H, Li G, Lu Y, Zhou Q, Zheng S, Lin Q, Liu Y, Li Z and Chen R: Cancer-associated fibroblasts promote progression and gemcitabine resistance via the SDF-1/SATB-1 pathway in pancreatic cancer. Cell Death Dis. 9:10652018. View Article : Google Scholar : PubMed/NCBI | |
Zhang F, Cui JY, Gao HF, Yu H, Gao FF, Chen JL and Chen L: Cancer-associated fibroblasts induce epithelial-mesenchymal transition and cisplatin resistance in ovarian cancer via CXCL12/CXCR4 axis. Future Oncol. 16:2619–2633. 2020. View Article : Google Scholar : PubMed/NCBI | |
Jin Y, Bian S, Wang H, Mo J, Fei H, Li L, Chen T and Jiang H: CRMP2 derived from cancer associated fibroblasts facilitates progression of ovarian cancer via HIF-1α-glycolysis signaling pathway. Cell Death Dis. 13:6752022. View Article : Google Scholar | |
Luo M, Luo Y, Mao N, Huang G, Teng C, Wang H, Wu J, Liao X and Yang J: Cancer-associated fibroblasts accelerate malignant progression of non-small cell lung cancer via connexin 43-formed unidirectional gap junctional intercellular communication. Cell Physiol Biochem. 51:315–336. 2018. View Article : Google Scholar : PubMed/NCBI | |
Chelakkot C, Chelakkot VS, Shin Y and Song K: Modulating glycolysis to improve cancer therapy. Int J Mol Sci. 24:26062023. View Article : Google Scholar : PubMed/NCBI | |
Ippolito L, Comito G, Parri M, Iozzo M, Duatti A, Virgilio F, Lorito N, Bacci M, Pardella E, Sandrini G, et al: Lactate rewires lipid metabolism and sustains a metabolic-epigenetic axis in prostate cancer. Cancer Res. 82:1267–1282. 2022. View Article : Google Scholar : PubMed/NCBI | |
Cao Y: Adipocyte and lipid metabolism in cancer drug resistance. J Clin Invest. 129:3006–3017. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yang L, Achreja A, Yeung TL, Mangala LS, Jiang D, Han C, Baddour J, Marini JC, Ni J, Nakahara R, et al: Targeting stromal glutamine synthetase in tumors disrupts tumor microenvironment-regulated cancer cell growth. Cell Metab. 24:685–700. 2016. View Article : Google Scholar : PubMed/NCBI | |
Li J, Li Y, Fu L, Chen H, Du F, Wang Z, Zhang Y, Huang Y, Miao J and Xiao Y: Targeting ncRNAs to overcome metabolic reprogramming-mediated drug resistance in cancer (review). Int J Oncol. 66:352025. View Article : Google Scholar : | |
Yang J, Shi X, Yang M, Luo J, Gao Q, Wang X, Wu Y, Tian Y, Wu F and Zhou H: Glycolysis reprogramming in cancer-associated fibroblasts promotes the growth of oral cancer through the lncRNA H19/miR-675-5p/PFKFB3 signaling pathway. Int J Oral Sci. 13:122021. View Article : Google Scholar : PubMed/NCBI | |
Tao S, Gao Y, Wang X, Wu C, Zhang Y, Zhu H and Li J: CAF-derived exosomal LINC01711 promotes breast cancer progression by activating the miR-4510/NELFE axis and enhancing glycolysis. FASEB J. 39:e704712025. View Article : Google Scholar : PubMed/NCBI | |
Zhang C, Wang XY, Zhang P, He TC, Han JH, Zhang R, Lin J, Fan J, Lu L, Zhu WW, et al: Cancer-derived exosomal HSPC111 promotes colorectal cancer liver metastasis by reprogramming lipid metabolism in cancer-associated fibroblasts. Cell Death Dis. 13:572022. View Article : Google Scholar : PubMed/NCBI | |
Smith AG and Macleod KF: Autophagy, cancer stem cells and drug resistance. J Pathol. 247:708–718. 2019. View Article : Google Scholar : | |
Dikic I and Elazar Z: Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol. 19:349–364. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhu S, Mao J, Zhang X, Wang P, Zhou Y, Tong J, Peng H, Yang B and Fu Q: CAF-derived exosomal lncRNA FAL1 promotes chemoresistance to oxaliplatin by regulating autophagy in colorectal cancer. Dig Liver Dis. 56:330–342. 2024. View Article : Google Scholar | |
Towers CG, Wodetzki D and Thorburn A: Autophagy and cancer: Modulation of cell death pathways and cancer cell adaptations. J Cell Biol. 219:e2019090332020. | |
Liao JK, Zhou B, Zhuang XM, Zhuang PL, Zhang DM and Chen WL: Cancer-associated fibroblasts confer cisplatin resistance of tongue cancer via autophagy activation. Biomed Pharmacother. 97:1341–1348. 2018. View Article : Google Scholar | |
Gao Q, Fang X, Chen Y, Li Z and Wang M: Exosomal lncRNA UCA1 from cancer-associated fibroblasts enhances chemoresistance in vulvar squamous cell carcinoma cells. J Obstet Gynaecol Res. 47:73–87. 2021. View Article : Google Scholar | |
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 | |
Chen B, Dragomir MP, Yang C, Li Q, Horst D and Calin GA: Targeting non-coding RNAs to overcome cancer therapy resistance. Signal Transduct Target Ther. 7:1212022. View Article : Google Scholar : PubMed/NCBI | |
Ran FA, Hsu PD, Lin CY, Gootenberg JS, Konermann S, Trevino AE, Scott DA, Inoue A, Matoba S, Zhang Y and Zhang F: Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell. 154:1380–1389. 2013. View Article : Google Scholar : PubMed/NCBI | |
Shtam TA, Kovalev RA, Varfolomeeva EY, Makarov EM, Kil YV and Filatov MV: Exosomes are natural carriers of exogenous siRNA to human cells in vitro. Cell Commun Signal. 11:882013. View Article : Google Scholar : PubMed/NCBI | |
Kamali MJ, Salehi M, Fatemi S, Moradi F, Khoshghiafeh A and Ahmadifard M: Locked nucleic acid LNA): A modern approach to cancer diagnosis and treatment. Exp Cell Res. 423:1134422023. View Article : Google Scholar | |
Rupaimoole R and Slack FJ: MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 16:203–222. 2017. View Article : Google Scholar : PubMed/NCBI | |
Kwok A, Raulf N and Habib N: Developing small activating RNA as a therapeutic: Current challenges and promises. Ther Deliv. 10:151–164. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zheng B, Mai Q, Jiang J and Zhou Q: The therapeutic potential of small activating RNAs for colorectal carcinoma. Curr Gene Ther. 19:140–146. 2019. View Article : Google Scholar : PubMed/NCBI | |
Fu J, Dong H, Wu J and Jin Y: Emerging progress of RNA-based antitumor therapeutics. Int J Biol Sci. 19:3159–3183. 2023. View Article : Google Scholar : PubMed/NCBI | |
Guo W, Wu Z, Chen J, Guo S, You W, Wang S, Ma J, Wang H, Wang X, Wang H, et al: Nanoparticle delivery of miR-21-3p sensitizes melanoma to anti-PD-1 immunotherapy by promoting ferroptosis. J Immunother Cancer. 10:e0043812022. View Article : Google Scholar : PubMed/NCBI | |
Xin X, Kumar V, Lin F, Kumar V, Bhattarai R, Bhatt VR, Tan C and Mahato RI: Redox-responsive nanoplatform for codelivery of miR-519c and gemcitabine for pancreatic cancer therapy. Sci Adv. 6:eabd67642020. View Article : Google Scholar : PubMed/NCBI | |
Li B, Shao H, Gao L, Li H, Sheng H and Zhu L: Nano-drug co-delivery system of natural active ingredients and chemotherapy drugs for cancer treatment: A review. Drug Deliv. 29:2130–2161. 2022. View Article : Google Scholar : PubMed/NCBI | |
Tassone P, Di Martino MT, Arbitrio M, Fiorillo L, Staropoli N, Ciliberto D, Cordua A, Scionti F, Bertucci B, Salvino A, et al: Safety and activity of the first-in-class locked nucleic acid (LNA) miR-221 selective inhibitor in refractory advanced cancer patients: A first-in-human, phase 1, open-label, dose-escalation study. J Hematol Oncol. 16:682023. View Article : Google Scholar : PubMed/NCBI | |
Beg MS, Brenner AJ, Sachdev J, Borad M, Kang YK, Stoudemire J, Smith S, Bader AG, Kim S and Hong DS: Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Invest New Drugs. 35:180–188. 2017. View Article : Google Scholar | |
Rasmussen LJH, Schultz M, Gaardsting A, Ladelund S, Garred P, Iversen K, Eugen-Olsen J, Helms M, David KP, Kjaer A, et al: Inflammatory biomarkers and cancer: CRP and suPAR as markers of incident cancer in patients with serious nonspecific symptoms and signs of cancer. Int J Cancer. 141:191–199. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhu M, Gao Y, Zhu K, Yuan Y, Bai H and Meng L: Exosomal miRNA as biomarker in cancer diagnosis and prognosis: A review. Medicine (Baltimore). 103:e400822024. View Article : Google Scholar : PubMed/NCBI | |
Montani F and Bianchi F: Circulating cancer biomarkers: The macro-revolution of the Micro-RNA. EBioMedicine. 5:4–6. 2016. View Article : Google Scholar : PubMed/NCBI | |
Li C, Teixeira AF, Zhu HJ and Ten Dijke P: Cancer associated-fibroblast-derived exosomes in cancer progression. Mol Cancer. 20:1542021. View Article : Google Scholar : PubMed/NCBI | |
Xu W, Liu S, Ma L, Cheng L, Li Q, Qing L, Yang Y and Dong Z: Identification of miRNA signature in cancer-associated fibroblast to predict recurrent prostate cancer. Comput Biol Med. 180:1089892024. View Article : Google Scholar : PubMed/NCBI | |
Alzhrani R, Alsaab HO, Petrovici A, Bhise K, Vanamala K, Sau S, Krinock MJ and Iyer AK: Improving the therapeutic efficiency of noncoding RNAs in cancers using targeted drug delivery systems. Drug Discov Today. 25:718–730. 2020. View Article : Google Scholar : | |
Winkle M, El-Daly SM, Fabbri M and Calin GA: Noncoding RNA therapeutics-challenges and potential solutions. Nat Rev Drug Discov. 20:629–651. 2021. View Article : Google Scholar : PubMed/NCBI | |
Hong DS, Kang YK, Borad M, Sachdev J, Ejadi S, Lim HY, Brenner AJ, Park K, Lee JL, Kim TY, et al: Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br J Cancer. 122:1630–1637. 2020. View Article : Google Scholar : PubMed/NCBI | |
Patil S, Gao YG, Lin X, Li Y, Dang K, Tian Y, Zhang WJ, Jiang SF, Qadir A and Qian AR: The development of functional non-viral vectors for gene delivery. Int J Mol Sci. 20:54912019. View Article : Google Scholar : PubMed/NCBI | |
Yahya EB and Alqadhi AM: Recent trends in cancer therapy: A review on the current state of gene delivery. Life Sci. 269:1190872021. View Article : Google Scholar : PubMed/NCBI |