Open Access

Role of metabolic reprogramming of cancer‑associated fibroblasts in tumor development and progression (Review)

  • Authors:
    • Ruyue Li
    • Yintao Li
  • View Affiliations

  • Published online on: August 25, 2025     https://doi.org/10.3892/ijo.2025.5796
  • Article Number: 90
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The occurrence and development of tumors is affected by tumor cells themselves and various components of the tumor microenvironment (TME). Among these, cancer‑associated fibroblasts (CAFs), the main stromal component, can differentiate from different cell types and play an important role in the TME. The present review summarized the role of the metabolic reprogramming of CAFs in tumor development and progression. As the rapid growth of tumors is a process inseparable from energy supply and the TME is characterized by hypoxia and nutrient deficiencies, metabolic reprogramming can reverse the effects of a lack of energy supply in the TME. Studies have found that CAFs can affect tumor proliferation, migration, invasion, metastasis and drug resistance by changing metabolic patterns. The present review promoted research on the metabolic reprogramming of CAFs and emphasized the importance of considering the heterogeneity and plasticity of CAFs in the TME, which will lead to the development of more effective therapeutic strategies that target specific metabolic pathways in CAFs, potentially improving the efficacy of cancer treatments and overcoming drug resistance.

Introduction

As a systemic disease, tumor occurrence and development are dictated primarily by tumor cells but also by other components. This concept originated from the 'seed and soil' principle proposed by Stephen Paget in 1889 (1). The 'seeds' are the tumor cells and the 'soil' is the other component of the tumor microenvironment (TME) that maintains the growth of tumor cells. The TME is composed of tumor and nonmalignant cells and an extracellular matrix (ECM). Nonmalignant cells include immune, stromal and vascular cells (2). Cancer-associated fibroblasts (CAFs) are the predominant cell type (3). Several previous studies have found that CAFs, in addition to supporting the TME, promote tumor development and progression by eliciting changes that include the secretion of cytokines, release of exosomes and alteration of metabolic patterns (4-7). As an important communication hub in the TME, CAFs can interact with tumor cells and other cellular components to participate in tumor development and progression (8). Therefore, it is important to understand the role of CAFs in these processes.

Tumor development and progression require a high nutrient supply to meet the energy requirements of the abnormal growth and proliferation of tumor cells and tumor and stromal cells change their metabolic patterns to facilitate these processes (9). Under aerobic conditions, normal cells produce ATP through oxidative phosphorylation via the tricarboxylic acid cycle (TCA) (10), whereas tumor cells obtain energy through aerobic glycolysis, which is known as the 'Warburg effect' (11). In the TME, tumor cells alter other metabolic patterns, such as the lipid (12) and amino acid (13) metabolism. Thus, metabolic reprogramming is a hallmark of cancer (14).

There is metabolic coupling between tumor cells and the surrounding CAFs (15), with anabolism and catabolism dominating tumor cells and CAFs, respectively, suggesting a symbiotic relationship in the TME (16). Contrary to the tumor metabolic model, CAFs feed tumor cells with high-energy 'fuels' such as lactic acid, generated glycolysis, via is the 'reverse Warburg effect' (17). Additionally, CAFs undergo various metabolic changes, such as lipid and amino acid metabolism reprogramming. Therefore, this review aimed to summarize the specific role of metabolic reprogramming of CAFs in tumor development and progression. To this end, a comprehensive search in the PubMed database was performed using the following query: ('Metabolic Reprogramming') AND ('Cancer-Associated Fibroblasts') AND ('Cancer' OR 'Tumor' OR 'Neoplasm') AND ('Cancer Progression' OR 'Tumor Progression' OR 'Neoplasm Invasiveness'). The selection of literature focused on effective research from the last decade that has substantially influenced the field.

Heterogeneity of CAFs

Source of CAFs

There are multiple sources of CAFs in the TME (Fig. 1). CAFs are mainly produced by activating normal fibroblasts (NFs) (18). Previous studies have found that various regulatory factors, such as cytokines and signaling pathways, can participate in tumor progression and development through transforming growth factor beta 1 (TGF-β1), which is involved in the activation of NFs (19) and the activation of various pro-inflammatory fibroblast-derived cytokines, such as interleukin (IL)-1, IL-6 and tumor necrosis factor (20-22). Furthermore, previous studies revealed that the Notch vascular endothelial growth factor-related signaling pathways can mediate CAF activation (23) and ECM remodeling, which is conducive to the generation and maintenance of CAFs (24). Physiological factors such as reactive oxygen species (ROS) and metabolic disorders stimulate the activation of CAFs (25). Additionally, unlike other types of cancer, NFs in pancreatic and liver cancers have a unique name, known as 'stellate cells.' Activated pancreatic and hepatic stellate cells are transformed into CAFs (26-28).

Other cell types can be converted into CAFs. Bone marrow mesenchymal stem cells differentiate into CAFs by recruitment through the TGF-β-Smad signaling pathway (29-31). Additionally, epithelial (32), endothelial (33) and macrophage (34,35) cells can be converted into CAFs through epithelial-mesenchymal transition (EMT), endothelial-mesenchymal transition and macrophage-myofibroblast transition, respectively. For example, Dulauroy et al (36) found that when acute tissue injury occurs, the primary sources of CAFs are perivascular cells expressing disintegrin and metalloproteinase 12(+). In breast cancer, human adipose tissue-derived stem cells can be transdifferentiated into CAFs via the TGF-β1-Smad3 signaling pathway (37). Androgen receptor-associated protein 55 expression in stromal smooth muscle cells in prostate cancer has been implicated in CAF formation (38). Thus, CAFs are heterogeneous.

Biomarkers of CAFs

In the TME, unlike tumor and immune cells, CAFs have various biomarkers that are not specific (39). Vimentin is a biomarker for resting fibroblasts (40). As CAFs are derived from various cell precursors, activated CAFs retain some of the functions of the cells or tissues of origin; therefore, there are differences in the molecular expression of CAFs. Common biomarkers of activated CAFs include fibroblast-activating protein, α-smooth muscle actin, fibroblast-specific protein 1 and platelet-derived growth factor receptors (41). CAFs should be identified by combining multiple biomarkers owing to the non-specificity of activated molecular markers of CAFs. For example, Hu et al (42) performed a functional analysis of CAF heterogeneity in tumor tissues of patients with non-small cell lung cancer and identified CAF subtypes using CAF biomarkers. The identified CAF subtypes were distinguished as follows: The first was mainly characterized by high expression of hepatocyte growth factor and fibroblast growth factor 7, low expression of phospho-Smad2 and a strong protective effect on tumors; the second was mainly characterized by high expression of fibroblast growth factor 7, low expression of hepatocyte growth factor and phospho-Smad2 and a moderate protective effect on tumors; and the third was mainly characterized by high expression of phospho-Smad2, low expression of hepatocyte growth factor and fibroblast growth factor 7 and a minimum protective effect on tumors.

Subtypes of CAFs

CAFs are characterized by phenotypic heterogeneity and plasticity (43) and single-cell (sc)RNA-seq can be used to distinguish between CAF subtypes by detecting cellular heterogeneity (44). Previous studies named the CAF subtypes according to the function of a unique gene set (45-50) (Fig. 2). Therefore, the present review summarized the common CAF subtypes and their functions in different types of cancer. A previous study found that pancreatic stellate cells could differentiate into two subtypes of CAFs: Inflammatory CAFs (iCAFs) and myofibroblast CAFs. Myofibroblast CAFs were observed near tumor cells and exhibited upregulated expression of α-smooth muscle actin, while iCAFs detached from tumor cells and mainly secreted IL-6, IL-11, platelet-derived growth factor receptor α and other inflammatory mediators, while α-smooth muscle actin expression was not increased (45). iCAFs and myofibroblast CAFs are the most common CAF subtypes among multiple types of cancer (51), with similar genetic characteristics but distinct functional properties (52). A previous study on pancreatic ductal adenocarcinoma (PDAC) explored the specific mechanism of subtype heterogeneity of CAFs. Biffi et al (53) used organoids and mouse models to reveal that CAFs generate iCAFs through IL-1-induced leukemia inhibitory factor expression and the Janus kinase/signal transducer and activator of transcription downstream signaling pathway, while TGF-β can antagonize this process by downregulating IL-1 receptor, type I expression and promoting the differentiation of CAFs into myofibroblast CAFs. Subsequently, Elyada et al (46) discovered a new subtype of CAFs in PDAC that express major histocompatibility complex class II and CD74 but not the classical costimulatory molecule, called 'antigen-presenting CAF.' The three subtypes are interchangeable. Wang et al (54) compared tumor heterogeneity among patients with PDAC with different degrees of fibroproliferation and found that there is a new subtype of CAFs a with highly activated metabolic state in patients with loose PDAC, which mainly plays a role in tumor development and progression by enhancing glycolytic metabolism. Additionally, Niu et al (55) found a lipid-rich subtype of CAFs in SET domain-containing 2-deficient PDAC that is involved in tumor progression by providing lipids for mitochondrial oxidative phosphorylation through the ATP-binding cassette, sub-family A, member 8a transporter, suggesting that PDAC is a tumor type with significant metabolic changes.

Other types of cancer have their own CAF subtypes. For example, Lambrechts et al (56) compared normal and tumor lung tissues to identify multiple stromal cell subtypes in the TME of patients with lung cancer. Subsequently, Cords et al (47) investigated the prognosis-relevant CAF subtypes in non-small cell lung cancer, including tumor-like CAFs, matrix CAFs, iCAFs and interferon-response CAFs. Among them, tumor-like CAFs and matrix CAFs were associated with poor prognosis, whereas iCAFs and interferon-response CAFs exhibited improved prognosis. Additionally, in gastrointestinal cancer, differences in the CAF subtypes were observed within tumor tissues at different sites. For example, Li et al (48) identified two CAF subtypes in gastric cancer, ECM CAFs and iCAFs, with ECM CAFs being a newly discovered subtype of CAF with high periostin expression. Roulis et al (49) identified a rare CAF subtype, 'pericryptal Ptgs2-expressing fibroblasts,' in the intestinal mesenchyme, which is involved in the progression of colorectal cancer (CRC). Rare CAF subtypes have been identified in breast cancer. Bartoschek et al (50) detected vascular, m, cycling and developmental CAF subtypes with different origins in mesenchymal cells in a mouse breast cancer model. A previous breast cancer study (57) analyzed the CAF subtypes using spatial transcriptomics and proteomics and found steady-state, mechanoresponsive and immunomodulatory CAF superclusters. The differences in the CAF subtypes between these two breast cancers may be due to differences in ethnicity and experimental techniques used. Notably, Ma et al (58) analyzed the CAF subtypes in six types of cancer and found that metabolic reprogramming occurs in subtypes of CAFs other than metabolic state CAFs. For example, matrix CAFs were enriched in fatty acid biosynthesis. Therefore, the role of CAFs in the TME may depend on metabolic reprogramming (Fig. 2).

Metabolic reprogramming of CAFs

In the TME, metabolic reprogramming is not limited to altering a single metabolic mode, but to the interaction of the entire metabolic network, including glucose, lipid and amino acid metabolism (59). CAFs are primary regulators of tumor metabolism (60); accordingly, the present study has summarized the mechanisms of metabolic reprogramming in CAFs (Fig. 3).

Reverse 'Warburg effect'

Even under normoxia, CAFs preferentially convert glucose to lactate via aerobic glycolysis, a phenotype driven by factors such as TGF-β and exacerbated by hypoxia. TGF-β1 enhances glucose uptake by upregulating glucose transporter 1 (GLUT1) via Smad2/3, p38 mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) activation. Under hypoxia, hypoxia-inducible factor-1α (HIF-1α) directly activates GLUT1 transcription (61) and inhibition of prolyl hydroxylase-mediated hydroxylation stabilizes HIF-1α (62). For example, hypoxic gastric cancer cells upregulate circ-NRIP1 acts as a sponge for miR-138-5p, relieving HIF-1α inhibition and enhancing glycolysis (63). HIF-1α can also be stabilized by TGF-β1 or platelet-derived growth factor, which downregulate isocitrate dehydrogenase 3α via miR-424, causing α-ketoglutarate depletion and prolyl hydroxylase 2 inhibition (64). Furthermore, TGF-β1 and HIF-1α augment glycolytic flux by upregulating key enzymes, hexokinase 2, 6-phosphofructo-2-kinase/fructose-2,6-bis-phosphatase 3 and pyruvate kinase M2 (65-67).

Mitochondrial dysfunction is a key driver of the metabolic shift from oxidative phosphorylation to aerobic glycolysis in CAFs, a process regulated by multiple mechanisms. ROS are critical mediators of mitochondrial retrograde signaling (68). External stimuli, such as radiation, damage mitochondria, elevating ROS levels that progressively harm both the fibroblast nucleus and mitochondria (69). Dysregulated mitochondrial calcium homeostasis also plays a role in ovarian cancer; specifically, overexpression of mitochondrial calcium uptake 1 inhibits pyruvate dehydrogenase, blocking pyruvate entry into the TCA cycle and enhancing glycolysis (70). Additionally, Sung et al (71) showed that triple-negative breast cancer cells overexpress integrin β4 and secrete it via exosomes, which CAFs internalize. This triggers c-Jun and AMP kinase phosphorylation, activates BNIP3L-mediated mitophagy and induces hyperglycolysis with excessive lactate secretion.

Lactate, the end product of CAF glycolysis, reinforces its own production through multiple positive feedback mechanisms, promoting a glycolytic metabolic network (72). Intracellularly, lactate catalyzes histone lysine lactylation to upregulate glycolytic genes (73) and stabilizes HIF-1α. For example, Kozlov et al (74) showed that lactate reduces prolyl hydroxylase phosphorylation and increases ROS, thereby stabilizing HIF-1α, boosting transcription and protein levels of glycolytic enzymes and driving the shift from oxidative phosphorylation to glycolysis in fibroblasts. Lactate also binds its G-protein coupled receptor GPR81, activating downstream signaling in an autocrine manner to further enhance glycolysis (75). Specifically, lactate, acting via GPR81, reduces intracellular cAMP levels, thereby inhibiting protein kinase A activation. This process suppresses protein kinase A-induced ubiquitination of HIF-1α, leading to its stabilization (76,77). Lactate can also activate the PI3K/Akt signaling pathway via GPR81, which promotes the translocation of GLUT1 to the plasma membrane and activates glycolytic enzymes (78). Extracellularly, lactate within the TME can also enhance glycolysis through various mechanisms. For instance, lactate efflux acidifies the surrounding microenvironment, activating matrix metalloproteinases 2 and 9 to degrade the ECM. This process releases latent TGF-β, which then activates the TGF-β receptor/PI3K signaling pathway to synergistically enhance glycolysis (79).

Amino acid metabolism

Glutamine (Gln) is central to amino acid metabolic reprogramming in CAFs. CAFs exhibit a pronounced dependency on Gln; Gln-sensing activates the TRAF6-p62 complex, driving Akt2-mediated polarization and invasion (80). In triple-negative breast cancer (81), estrogen activates the G-protein-coupled estrogen receptor in CAFs, initiating a cAMP/protein kinase A/CREB signaling axis that upregulates Gln synthetase, which increases Gln synthesis. CAFs then catabolize Gln into metabolites readily taken up by tumor cells. For example, in PDAC, tumor cells stimulate pancreatic stellate cells to undergo autophagy, degrading cellular proteins to produce non-essential amino acids such as alanine, which tumor cells take up as an alternative carbon source to fuel the TCA cycle (82).

ECM remodeling is critical for tumor invasion and CAFs are the main producers of ECM components. They facilitate ECM production by using Gln for de novo proline synthesis, enabled by high expression of the rate-limiting enzyme pyrroline-5-carboxylate reductase 1 (83,84). Proline also contributes to other aspects of tumorigenesis (85). Under stress conditions such as glucose deprivation, proline, glutamine and their derivatives serve as alternative energy sources. Mitochondrial proline dehydrogenase/proline oxidase degrades proline into glutamate, which is subsequently converted to α-ketoglutarate for TCA cycle entry. This pathway also generates pyrroline-5-carboxylate, ATP and ROS. Moreover, ornithine, glutamate and proline interconvert dynamically.

CAFs also highly express arginase, dominating arginine metabolism to create an immunosuppressive microenvironment. In PDAC, hypoxia induces arginase II in CAFs, converting arginine to ornithine. Local arginine depletion impairs tumor-infiltrating T-cell function and fosters immunosuppression (86).

Lipid metabolism

Lipid metabolic reprogramming in CAFs drives tumorigenesis. Upstream signaling factors, including TGF-β and lysophosphatidic acid, initiate this reprogramming and trigger downstream events (87). To meet the high lipid demands of both CAFs and tumor cells, CAFs acquire lipids via exogenous uptake and endogenous de novo synthesis. They overexpress transporters such as CD36, which mediates fatty acid and oxidized LDL uptake in triple-negative breast cancer (88) and hepatocellular carcinoma (89) and members of the fatty acid transport protein family (90). When exogenous lipids are scarce, CAFs upregulate de novo synthesis enzymes; for example, 17β-estradiol (E2)/G1 activates epidermal growth factor receptor-extracellular signal-regulated kinase 1/2 signaling to upregulate fatty acid synthase expression and lipid production (91). A key outcome is lipid droplet formation to nourish tumor cells. Under hypoxia, HIF-1α induces stearoyl-CoA desaturase 1, expanding lipid droplet stores (92). Lipid droplets release free fatty acids for mitochondrial β-oxidation and CAFs transfer lipids via exosomes to support tumor progression (93).

Various products generated from CAF lipid metabolic reprogramming can also act on tumor cells. Lipid signaling molecules, for instance, can bind to specific receptors on the surface of CAFs, activating them and promoting their conversion to a pro-tumorigenic phenotype. TGF-β stimulates sphingosine kinase 1 to produce sphingosine-1-phosphate, which then binds to sphingosine-1-phosphate receptors 2 and 3, leading to p38 MAPK phosphorylation. This drives differentiation of CAF cells and their secrete of factors that promote ovarian cancer cell invasion (94). In PDAC, activated pancreatic stellate cells secrete large quantities of lipids, particularly lysophosphatidylcholine that is hydrolyzed to lysophosphatidic acid by the autocrine enzyme autotaxin, which in turn promotes the proliferation and migration of PDAC cells by activating the Akt2 signaling pathway (95). Under persistent pathological stimulation, human pancreatic stellate cells upregulate cyclooxygenase-2 to produce prostaglandin E2, which modulates the pro-fibrotic activity of the cells via the EP4 subtype of the prostaglandin E2 receptor (96). Furthermore, products such as ketone bodies and fatty acids generated by CAFs serve as important energy sources for tumor cells. For example, under conditions of Cav-1 loss or serum starvation, CAFs activate ketogenic enzymes to secrete ketone bodies, which are then transported into cancer cells via MCT1 to drive mitochondrial biogenesis and promote tumor progression (97).

Role of metabolic reprogramming of CAFs in tumor development and progression

Metabolic reprogramming of CAFs promotes tumor proliferation

Tumor cell proliferation can be monitored using the Ki-67 proliferation index employing immunohistochemistry or the mitotic count using hematoxylin and eosin staining (98). Tumor cell proliferation is central to carcinogenesis; the faster its proliferation, the higher the degree of tumor malignancy (99). In the TME, tumor cells promote their own proliferation and other components, such as CAFs, are also involved in this process (100). Additionally, metabolic reprogramming may affect the survival of cancer stem cells (99). Therefore, the role of the metabolic reprogramming of CAFs in tumor cell proliferation are summarized in Table I. CAFs can participate in tumor cell proliferation by reprogramming glucose metabolism through various regulatory factors. A previous study found that in various types of cancer, such as breast and prostate cancers, there is a metabolic coupling between CAFs and tumor cells. The specific mechanism is that tumor cells drive glycolysis in CAFs through HIF1-α and nuclear factor-κ-B to produce mitochondrial fuels such as lactic acid, ketone bodies, fatty acids, glutamine and other amino acids, which provide nutrients for tumors and participate in tumor growth (101). This phenomenon has been confirmed in prostate cancer (102). A previous study on breast cancer (103) have found that isolated CAFs exhibit a glycolytic phenotype and produce lactate and pyruvate; when the consumption of lactate production is inhibited, the metabolic pattern of CAFs is altered, thereby hindering tumor growth. A previous study (104) explored the specific mechanism of the glycolytic phenotype expressed by CAFs in breast cancer; small nucleolar RNA host gene 3 in exosomes secreted by CAFs can increase pyruvate kinase M expression by positively regulating its expression, then transitioning to a pro-glycolytic phenotype. Additionally, in oral squamous cell carcinoma, the high expression of integrin subunit β 2 in CAFs can lead to the regulation of the PI3K-serine-threonine kinase-mammalian target of the rapamycin signaling pathway, enhancement of the glycolytic pathway, increase in the secretion of lactic acid and promotion of tumor cell proliferation (105).

Table I

Specific mechanisms by which metabolic reprogramming of CAFs promotes tumor proliferation.

Table I

Specific mechanisms by which metabolic reprogramming of CAFs promotes tumor proliferation.

First author/s, yearType of tumorMetabolic modeActivating factorsMechanism(Refs.)
Martinez-Outschoorn et al, 2014/Becker et al, 2020Breast, prostate, and head and neck cancersGlycolysisAnoxia/Inflammatory factorsROS/HIF-1α/ NF-κB Axis(101/103)
Fiaschi et al, 2012Prostate cancerAerobic glycolysisCancer cell-CAF contactHIF1-SIRT3 Axis(102)
Li et al, 2020Breast cancerGlycolysisExosomesSNHG3/miR-330-5p/ PKM Axis(104)
Zhang et al, 2020Oral squamous cell carcinomaGlycolysisITGB2PI3K/AKT/mTOR Pathway(105)
Yang et al, 2016Ovarian carcinomaAmino acid metabolismNutrient scarcity in the TMEGlutamine Anabolic Pathway(107)
Linares et al, 2017Prostate cancerAmino acid metabolismP62 deficiencyP62/ATF4 Axis(108)
Mishra et al, 2018Prostate cancerAmino acid metabolismAndrogen signaling deprivation therapyRASAL3/Ras/Axis Macropinocytosis(109)

[i] AFT4, activating transcription factor 4; CAFs, cancer-associated fibroblasts; HIF-α, hypoxia-inducible factor 1-α; mTOR, mammalian target of rapamycin; NFκB, nuclear factor-κB; PKM, pyruvate kinase M; RASAL3, RAS-activating protein-like 3; ROS, reactive oxygen species; SIRT3, sirtuin 3; SNHG3, small nucleolar RNA host gene 3.

Other metabolic patterns of CAFs change tumor cell proliferation. CAFs can synthesize amino acids through the TCA to provide carbon and nitrogen sources for tumor cells and participate in tumor cell proliferation (106). For example, in ovarian cancer, CAFs promote Gln production by upregulating the Gln anabolic pathway. Gln is absorbed by ovarian cancer cells and converted to glutamate by glutaminase, thereby maintaining tumor cell proliferation (107). P62-deficient CAFs can also produce asparagine by activating glucose carbon flux through the pyruvate carboxylase-asparagine synthase cascade, which is involved in tumor growth as a nitrogen source for stromal and tumor epithelial proliferation (108). Additionally, crosstalk exists between amino acid metabolism in tumor cells and CAFs that promote tumor proliferation. A prostate cancer study (109) demonstrated that epigenetic silencing of CAFs can drive macropinocytosis-mediated Gln synthesis. This promotes tumor cell proliferation by increasing the TCA flux after Gln is absorbed by tumor cells. Lipids are the basis of tumor proliferation. However, there is no study confirming the effect of lipid metabolism in CAFs on tumor cell proliferation and further exploration is needed.

Metabolic reprogramming of CAFs promotes tumor migration and invasion

The directed migration or invasion of the tumor into adjacent tissues, which is the initial stage of tumor metastasis, is a hallmark of cancer (110). This process involves tumor cells gaining the ability to actively move and migrate directionally to adjacent noncancerous tissues, known as the 'invasion front (margin)', wherein they approach the edge of adjacent noncancerous tissues (111); tumor cells then cross the adjacent basement membrane and invade surrounding tissues through EMT (112). Previous studies have revealed that the metabolic reprogramming of CAFs can participate in tumor migration and invasion (Table II). During tumor formation, tumors act on tumor cells by generating various forces that cause dynamic changes in behavior (113,114). In this manner, ECM hardness can affect hepatocellular carcinoma migration. The specific mechanism involves a hardened ECM that upregulates aerobic glycolysis through the MAPK-Yes-associated protein signaling cascade (115). In nasopharyngeal carcinoma, the extracellular vesicles package Epstein-Barr virus-encoded latent membrane protein 1 that activates NFs, which transform into CAFs and activated CAFs undergo aerobic glycolysis and autophagy, promoting tumor migration and formation of a premetastatic microenvironment (116). This process alters the lipid metabolism patterns. For example, Gong et al (117) found that fatty acid synthase in CAFs in CRC was markedly increased compared with that in NFs and that small-molecule metabolites of CAFs could promote the migration of CRC cells. In the TME, tumor cells secrete cytokines that activate quiescent fibroblasts. Moreover, activated CAFs exhibit increased active lipid levels, increased lipid content and lipid phagocytosis and thereby promote tumor cell migration through CD36 lipid metabolism reprogramming (118).

Table II

Specific mechanisms by which metabolic reprogramming of CAFs promotes tumor migration.

Table II

Specific mechanisms by which metabolic reprogramming of CAFs promotes tumor migration.

First author/s, yearType of tumorMetabolic modeActivating factorsMechanism(Refs.)
Liu et al, 2020Hepatocellular carcinomaAerobic glycolysisECM StiffnessMAPK-YAP Pathway(115)
Wu et al, 2020Nasopharyngeal carcinomaAerobic glycolysisExtracellular vesicle-packaged LMP1NF-κB p65 Pathway(116)
Gong et al, 2020Colorectal cancerLipid metabolismElevated FASN ExpressionLipid Trafficking(117)
Wang et al, 2024Hepatocellular carcinomaLipid metabolismLipid metabolismCD36-dependent Signaling Hub for CAF Activation(118)
Sun et al, 2019Breast cancerGlycolysisAnoxiaOxidized ATM-Orchestrated Glycolytic Reprogramming(119)
Shan et al, 2017Pancreas cancerAerobic glycolysisAerobic glycolysisUnidirectional Lactate Shuttle from CAFs to Cancer Cells(120)
Mestre-Farrera et al, 2021Epithelial tumorAmino acid metabolismTGFβ-Snail1TRAF6-p62-Akt2 Axis(80)

[i] Akt2, protein kinase B 2; ATM, Ataxia-Telangiectasia Mutated; CAF, cancer-associated fibroblast; ECM, extracellular matrix; FASN, fatty acid synthase; LMP1, latent membrane protein 1; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor-κB; Snail1, Zinc finger protein SNAI1; TGFβ, transforming growth factor beta; TRAF6, tumor necrosis factor receptor associated factor 6; YAP, yes-associated protein.

Tumor invasion includes multiple processes, such as promotion of EMT occurrence, ECM remodeling and enhanced angiogenesis and lymphangiogenesis (Table II). For example, it has been found that CAFs in breast cancer can enhance glycolysis, which produces lactate. This involves oxidizing ataxia-telangiectasia mutated kinase, which further activates the TGFβ1/MAPK signaling pathway and affects the mitochondrial activity of tumor cells to promote tumor invasion (119). The levels of key enzymes mediating aerobic glycolysis, such as lactate dehydrogenase B and pyruvate kinase M2 in CAFs in pancreatic cancer are increased, enhancing aerobic glycolysis and promoting pancreatic cancer cell invasion through 'tumor-stromal' metabolic coupling (120). Additionally, CAFs are Gln-dependent and when Gln is deficient, serine-threonine kinases regulate the migration and invasion of CAFs into Gln-rich regions, thereby promoting tumor epithelial cell invasion into adjacent tissues (80). ECM sclerosis is linked to Gln metabolism and forms a metabolic network in which various fuel sources promote tumor invasion (106).

Metabolic reprogramming of CAFs promotes tumor neovascularization

In the TME, the proliferation and differentiation of tumor cells require adequate oxygen and nutrients from blood vessels (121). Therefore, tumor neovascularization is closely related to the occurrence and development of tumors. Tumor neovascularization is the creation of blood vessels based on the original capillaries or post-capillary venules (122,123). This process involves the interaction of multiple growth factors and signaling pathways (124). Metabolic reprogramming of CAFs is a key factor in angiogenesis (Table III), including changes in multiple metabolic pathways, such as glucose, lipid and amino acid metabolism (125). In breast cancer, caveolin-1-deficient CAFs can upregulate the expression of glycolytic enzymes, provide energy through paracrine means and promote tumor angiogenesis (126). Fructose-2,6-biphosphatase 3, a metabolic enzyme that regulates glucose metabolism, is involved in glucose metabolism in CAFs in oral cancer to promote the growth of tumor tissues (127) by enhancing the pro-angiogenic phenotype (128). Additionally, hepatic stellate cells, which are unique to HCC, can be transformed into CAFs under the influence of tumor cells, induce abnormal lipid metabolism and secrete angiogenic cytokines to participate in the occurrence and development of tumors (129). In CRC, the oncogenic Kirsten rat sarcoma viral oncogene homolog activates transcription factor CP2 to upregulate bone morphogenetic protein 4 and wingless-type mammary tumor virus integration-site family member 5B adipogenesis factors, converting CAFs into lipid-rich CAFs, producing vascular endothelial growth factor A and stimulating tumor angiogenesis (130). Verginadis et al (131) found a strong correlation between activating transcription factor 4 in melanoma and pancreatic tumors and glycine and proline levels, the main amino acids of collagen. Activating transcription factor 4-deficient CAFs has been found to inhibit angiogenesis.

Table III

Specific mechanisms by which metabolic reprogramming of CAFs promotes tumor vascularization.

Table III

Specific mechanisms by which metabolic reprogramming of CAFs promotes tumor vascularization.

First author/s, yearType of tumorMetabolic modeActivating factorsMechanism(Refs.)
Bonuccelli et al, 2010Breast cancerAerobic glycolysisCaveolin-1 deficiency Glycolysis-Angiogenesis Axis(126)
Li et al, 2022Oral squamous cell carcinomaGlycolysisGlycolysis PGC-1α/PFKFB3-Angiogenesis signaling axis(128)
Zhou et al, 2018Hepatocellular carcinomaLipid metabolismmiR-21-Enriched ExosomesPDK1/Akt pathway(129)
Hsu et al, 2023Colorectal cancerLipid metabolismKRASTFCP2-BMP4/WNT5B Axis(130)
Verginadis et al, 2022Melanoma and pancreatic tumorsAmino acid metabolismATF4ATF4-Col1a1 Axis(131)

[i] AFT4, activating transcription factor 4; Akt, protein kinase B; BMP4, bone morphogenetic protein 4; Cola1, Collagen, type I, alpha 1; KRAS, Kirsten rat sarcoma viral oncogene homologue; PDK1, 3-phosphoinositide-dependent protein kinase-1; PFKFB3, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3; PGC-1α, peroxisome proliferator-activated receptor-gamma coactivator-1alpha; TFCP2, transcription factor CP2; WNT5B, Wnt Family Member 5B.

Metabolic reprogramming of cancer-associated fibroblasts promote tumor metastasis

Tumor metastasis is the leading cause of antineoplastic therapy failure and mortality in most patients with cancer (132). Tumor cells leave their primary organs to colonize and grow in distant organs, including the brain, lungs, liver and bone (133). Previous studies have revealed that NFs undergo metabolic changes after differentiating into CAFs, which promotes tumor metastasis (134) (Table IV). For example, in ovarian cancer cell lines, CAFs stimulate glycogen mobilization, which supports glycolysis in tumor cells and is involved in tumor metastasis (135). CRC liver metastasis is achieved by TGF-β1-induced conversion of hematopoietic stem cells into pro-metastatic CAFs, resulting in increased glucose uptake and glycolysis. The specific mechanism involves the combination of steroid receptor coactivator SH3 domain and SH3 domain-binding protein that forms the steroid receptor coactivator/SH3 domain-binding protein/Rab11/GLUT1 complex, which activates Rab11-dependent plasma membrane GLUT1 transport under TGF-β1 stimulation (136). Additionally, the CRC cell-derived exosome HSPC111 reprograms the lipid metabolism of CAFs, promoting the formation of a premetastatic microenvironment and CRC liver metastasis (137). Li et al (138) screened for lysyl oxidase, a CAF-derived oxidase that promotes the formation of a microenvironment in liver-metastatic gastric cancer by enhancing the Warburg effect.

Table IV

Specific mechanisms by which metabolic reprogramming of CAFs promotes tumor metastasis.

Table IV

Specific mechanisms by which metabolic reprogramming of CAFs promotes tumor metastasis.

First author/s, yearType of tumorMetabolic modeActivating factorsMechanism(Refs.)
Curtis et al, 2019Ovarian cancerGlycolysisCAFsp38α MAPK pathway(135)
Wang et al, 2024Colorectal cancer liver metastasesGlycolysisTGFβ1Src-SH3BP5-Rab11 Axis(136)
Zhang et al, 2022Colorectal cancer liver metastasesLipid metabolismHSPC111-containing exosomesCXCL5-CXCR2 Axis(137)
Li et al, 2019Gastric cancer liver metastasesAerobic glycolysisTGF-β1A Positive Feedback Loop involving the TGF-β1-LOX Axis and Aerobic Glycolysis(138)

[i] CAF, cancer-associated fibroblast; CXCL, C-X-C motif chemokine receptor; LOX, lysyl oxidase; MAPK, mitogen-activated protein kinase; Rab11, Ras related proteins in brain type 11; SH3BP5, SH3 domain-binding protein 5; TGFβ1, transforming growth factor β 1.

Metabolic reprogramming of CAFs promotes tumor drug resistance

Tumor drug resistance is a challenge for patients with cancer receiving anti-tumor therapy (139). Tumor cells do not respond to anti-tumor therapy when they are insensitive to anti-tumor drugs (primary resistance) and when they initially respond well to anti-tumor drugs, but the latter cannot achieve long-term benefits (secondary drug resistance) (140). Additionally, various tumor resistance mechanisms have been identified (141), including overexpression of ATP-binding cassette transporter excretion of drugs, EMT, cancer stem cells, cytoprotective autophagy, apoptosis and gene mutations. Tumor drug resistance is caused by the tumor cells and other cells in the TME, including CAFs. Zaal and Berkers (142) summarized the metabolic alterations associated with resistance to different antineoplastic drugs and found that the metabolic state of cells largely influences their therapeutic efficacy. Tumor cells adjust their metabolism to antineoplastic drugs through different mechanisms, such as maintaining a high glycolysis rate, switching glycolytic metabolites to the biosynthetic pathway and rewiring glutamine metabolism in mitochondria. A previous study found that metabolic alterations were associated with resistance to treatment. Broekgaarden et al (143) used a 3D culture model of PDAC and CAFs and found that metabolic alterations in tumor-matrix interactions caused statistically significant resistance of CAFs to anti-tumor drugs shown using high-throughput imaging or image analysis tools. Studies have proposed that specific metabolic pathway changes, such as in aerobic glycolysis and amino acid metabolism, are related to treatment resistance (Table V). For example, aerobic glycolysis of activated CAFs promotes the development of radiotherapy resistance in nasopharyngeal carcinoma cells (116). Ko et al (144) found that autophagic fibroblasts in breast cancer may be a significant source of glutamine and stromal fibroblasts and that glutamine exerts a synergistic effect that renders breast cancer cells resistant to tamoxifen. Additionally, Mishra et al (109) found that antagonizing Gln uptake (which involves CAF-driven micropinocytosis) in prostate cancer, restores the sensitivity to androgen deprivation therapy.

Table V

Specific mechanisms by which metabolic reprogramming of CAFs promotes tumor drug resistance.

Table V

Specific mechanisms by which metabolic reprogramming of CAFs promotes tumor drug resistance.

First author/s, yearType of tumorMetabolic modeActivating factorsMechanism(Refs.)
Wu et al, 2020Nasopharyngeal carcinomaAerobic glycolysisLMP1-containing EBV exosomesNF-κB p65 pathway(116)
Gong et al, 2020Breast cancerAmino acid metabolismPDAC-CAF InteractionMitochondrial Oxidative Metabolism(117)
Mishra et al, 2018Prostate cancerAmino acid metabolismandrogen signaling deprivation therapyThe Ras-Macropinocytosis-Glutamine Metabolic Axis(109)

[i] CAF, cancer-associated fibroblast; EBV, Epstein-Barr virus; LMP1, latent membrane protein 1; NF-κB, nuclear factor-κB; PDAC pancreatic ductal adenocarcinoma.

Clinical trials targeting metabolic reprogramming in CAFs

CAFs possess intrinsic characteristics that make them more amenable to therapeutic targeting than tumor cells. Their unique metabolic programs supply essential nutrients to tumor cells and support multiple stages of tumorigenesis. Metabolic reprogramming is central to meeting tumor energetic demands and adapting to the malignant microenvironment, making it a key vulnerability. Disrupting core CAF metabolic pathways thus represents a promising strategy to prevent cancer dissemination and recurrence and several clinical trials are underway. In CAFs exhibiting hyperactive glycolysis, the inhibitor 2-deoxy-D-glucose competitively blocks GLUTs and hexokinase, suppressing glycolysis and lactate secretion and depriving tumor cells of a critical energy source. This approach has been validated in patients with glioma and advanced solid tumors, where it also delayed repair of radiation- and chemotherapy-induced damage (145,146). The sensitizing effect of lonidamine, a direct hexokinase inhibitor, has also been demonstrated in solid tumors, including advanced breast, ovarian and lung cancers (147). Furthermore, clinical trials targeting lipid metabolic reprogramming in CAFs have focused primarily on fatty acid synthesis and oxidation. Since recurrent high-grade astrocytomas overexpress fatty acid synthase to fuel lipid biosynthesis and proliferation, combining the fatty acid synthase inhibitor TVB-2640 with bevacizumab nearly doubled the 6-month progression-free survival rate vs. bevacizumab alone (31.4 vs. 16%) (148). Similar clinical trials are ongoing for other types of cancer, including non-small cell lung cancer, breast, ovarian, prostate, colon and pancreatic cancers (148). Preclinical studies have confirmed that inhibiting acetyl-CoA carboxylase, a key rate-limiting enzyme in de novo fatty acid synthesis, with ND-646 can also control non-small cell lung cancer growth (149). Additionally, fatty acid β-oxidation is a crucial energy source for numerous tumor cells. It has been discovered that carnitine palmitoyltransferase 1 (CPT1) is not only the rate-limiting enzyme of fatty acid oxidation (150) but also essential for maintaining mitochondrial structure and function (151). Gugiatti et al (152) reported that microenvironment-activated chronic lymphocytic leukemia cells rely on carnitine palmitoyltransferase1A-mediated fatty acid oxidation to fuel proliferation and that combining the carnitine palmitoyltransferase1A inhibitor ST1326 with venetoclax produces a synergistic anti-tumor effect. Therapeutic targeting also has a focus on Gln metabolism: Gln is converted to glutamate by glutaminase and glutamate then fuels the TCA cycle. A Phase I study demonstrated that the glutaminase inhibitor telaglenastat (CB-839) has a favorable safety profile, pharmacokinetics/pharmacodynamics and antitumor activity in patients with solid tumors (153). Subsequent studies confirmed the efficacy of CB-839 in melanoma, renal cell carcinoma and non-small cell lung cancer, where it sensitizes tumors to other therapies and reverses drug resistance (154,155).

Conclusions and outlook

The occurrence and development of tumors are inseparable from the energy supply (156). A previous study found that most of the energy requirements of tumor tissues are due to changes in metabolic pathways (157). In the TME, metabolic reprogramming occurs as a single metabolic pathway and interactions within the entire metabolic network (158). Based on the source heterogeneity and phenotypic plasticity of CAFs, the present review described the metabolic reprogramming of CAFs and its role in tumorigenesis and development, including tumor proliferation, migration and invasion, angiogenesis, metastasis and drug resistance. It aimed to clarify future research directions by understanding the relevant research on CAF metabolic reprogramming in oncology. CAFs are the primary regulators of the TME, which directly interact with tumor cells and regulate immune cells to affect the occurrence and development of tumors (159). Additionally, metabolic reprogramming can act on immune cells to assist tumor cells in immune escape (160). Therefore, future studies should explore whether metabolic reprogramming of CAFs may act on immune cells to affect the occurrence and development of tumors.

CAFs are genetically stable and have a low possibility of drug resistance (161). Therefore, CAFs may be a potentially effective therapeutic target. Previous studies have investigated the use of CAFs as therapeutic options, including determining biomarkers targeting CAFs (18), activating CAFs to reverse their resting state (162), targeting CAF-related signaling pathways (162), reducing interstitial pressure and improving drug delivery (163) and coating drugs targeting CAFs by nano-counting to improve drug delivery (164). However, none of these studies achieved good treatment results, which may be because of the different CAF subtypes in the different types of cancer. Dysregulation of energy metabolism has become a research hotspot, including the development of inhibitors that inhibit key metabolic enzymes such as mutant isocitrate dehydrogenase. Glutathione peroxidase 4 and nicotinamide phosphoribosyl transferase have exhibited sound anti-tumor effects. Different CAF subtypes can affect tumorigenesis and development through metabolic mode changes; therefore, it was concluded that tumor progression can be inhibited by interfering with CAF metabolic reprogramming.

Analysis of multiple studies reveals that the differences in the metabolic profiles of CAFs across various types of cancer are likely attributable to the heterogeneity of CAF subtypes. For instance, breast cancer-associated CAFs predominantly rely on a glycolytic metabolism, characterized by enhanced glycolysis and increased lactate secretion (73). The hypoxic microenvironment can induce epigenetic reprogramming of HIF-1α and glycolytic enzymes, which sustains a hyper-glycolytic phenotype in CAFs. This results in the production of excess lactate and pyruvate that are subsequently transported to and utilized by cancer cells (103). Similarly, CAFs in melanoma (165) and head and neck squamous cell carcinoma (166) also primarily exhibit a high-glycolysis phenotype. PDAC is characterized by an abundant deposition of ECM by CAFs, with the ECM being primarily composed of collagen (167), which is rich in glycine, proline and their derivatives. Netrin G1 has been identified as a promoter of PDAC development; specifically, Netrin G1-positive CAFs support PDAC survival through Netrin G1-mediated glutamate/Gln metabolism (167). This suggests that PDAC-associated CAFs are mainly dependent on an amino acid-based metabolic program. By contrast, prostate cancer cells display a metabolic signature dominated by de novo fatty acid synthesis. Balaban et al (168) discovered that one subpopulation of prostate cancer cells (such as C4-2B) enhances fatty acid oxidation, while another (such as PC-3) increases triglyceride synthesis via DGAT1. Together, these distinct metabolic strategies cooperatively support the overall survival of the cancer cell population. Therefore, for clinical applications, scRNA sequencing can be employed to characterize the specific CAF subtypes present in individual patients across different cancers. This information can then guide the selection of the most appropriate inhibitors targeting the dominant CAF metabolic pathways.

Preclinical studies and early-phase clinical trials targeting CAF metabolic reprogramming have yielded encouraging preliminary results. However, translating these findings into effective clinical strategies remains a formidable challenge. First, the tumor microenvironment operates as an integrated metabolic network, which may activate compensatory pathways that lead to therapeutic resistance. Second, given the functional heterogeneity among CAF subtypes, the efficacy of monotherapeutic and non-selective targeting of CAF metabolism is unpredictable. Finally, therapeutic strategies targeting CAF metabolic reprogramming currently lack validation from large-scale, prospective studies to confirm their safety and efficacy profiles. Therefore, it was hypothesized that combining interventions against CAF metabolism with other anti-tumor therapies may be a viable therapeutic strategy. Research has found that metabolites from CAFs create an immunosuppressive TME, leading to resistance to immune checkpoint inhibitors in cancer patients (169). Targeting CAF metabolism to reduce the production of immunosuppressive metabolites can restore T-cell activity and have a sensitizing effect. Specifically, in soft tissue sarcoma, glycolytic CAFs not only form a physical barrier to block T-cell infiltration but also highly express GLUT1 to secrete CXCL16, which binds to CXCR6 on T-cells and impedes their migration into the tumor parenchyma. Targeting this immune barrier could potentially increase the efficacy of immune checkpoint inhibitors (170).

While molecularly targeted therapies are advancing towards the goal of precision medicine, the metabolic reprogramming of CAFs can help tumor cells survive the cytotoxic effects of these drugs by providing energy or metabolites. Co-targeting both CAF metabolic reprogramming and key gene mutations may reverse or delay the onset of resistance. For example, in renal cell carcinoma, monotherapy with signaling inhibitors such as cabozantinib or everolimus can easily lead to compensatory metabolic resistance. Emberley et al (171) attempted to add telaglenstat (CB-839) to inhibit glutaminase, thereby blocking glutaminolysis to cut off the tumor's energy supply and delay resistance. Targeting CAF metabolism can also reduce tumor cell self-repair and combining it with conventional anti-tumor therapies such as chemotherapy (172) or radiotherapy (173) may have a sensitizing effect. Currently, this combination strategy is still in the early stages of investigation and requires further research.

Availability of data and materials

Not applicable.

Authors' contributions

RL and YL designed the review and wrote the manuscript. RL prepared the figures and tables. Data authentication is not applicable. Both authors read and approved the final 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.

Abbreviations:

Akt

protein kinase B

CAF

cancer-associated fibroblast

CRC

colorectal cancer

ECM

extracellular matrix

EMT

epithelial-mesenchymal transition

Gln

glutamine

HCC

hepatocellular carcinoma

iCAF

inflammatory CAF

IL

interleukin

MAPK

mitogen-activated protein kinase

NF

normal fibroblast

PDAC

pancreatic ductal adenocarcinoma

PI3K

phosphoinositide 3-kinase

TCA

tricarboxylic acid cycle

TGF-β1

transforming growth factor beta 1

TME

tumor microenvironment

Acknowledgments

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 82373044) and the Natural Science Foundation of Shandong Province (grant no. ZR2023LSW023).

References

1 

Fidler IJ: The pathogenesis of cancer metastasis: The 'seed and soil' hypothesis revisited. Nat Rev Cancer. 3:453–458. 2003. View Article : Google Scholar : PubMed/NCBI

2 

Zhao Y, Shen M, Wu L, Yang H, Yao Y, Yang Q, Du J, Liu L, Li Y and Bai Y: Stromal cells in the tumor microenvironment: accomplices of tumor progression? Cell Death Dis. 14:5872023. View Article : Google Scholar : PubMed/NCBI

3 

Glabman RA, Choyke PL and Sato N: Cancer-associated fibroblasts: Tumorigenicity and targeting for cancer therapy. Cancers (Basel). 14:39062022. View Article : Google Scholar : PubMed/NCBI

4 

Sun H, Wang X, Wang X, Xu M and Sheng W: The role of cancer-associated fibroblasts in tumorigenesis of gastric cancer. Cell Death Dis. 13:8742022. View Article : Google Scholar : PubMed/NCBI

5 

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

6 

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 :

7 

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

8 

Arima Y, Matsueda S and Saya H: Significance of cancer-associated fibroblasts in the interactions of cancer cells with the tumor microenvironment of heterogeneous tumor tissue. Cancers (Basel). 15:25362023. View Article : Google Scholar

9 

Zhang S, Xiao X, Yi Y, Wang X, Zhu L, Shen Y, Lin D and Wu C: Tumor initiation and early tumorigenesis: Molecular mechanisms and interventional targets. Signal Transduct Target Ther. 9:1492024. View Article : Google Scholar : PubMed/NCBI

10 

Martínez-Reyes I and Chandel NS: Mitochondrial TCA cycle metabolites control physiology and disease. Nat Commun. 11:1022020. View Article : Google Scholar : PubMed/NCBI

11 

Akter R, Awais M, Boopathi V, Ahn JC, Yang DC, Kang SC, Yang DU and Jung SK: Inversion of the warburg effect: Unraveling the metabolic nexus between obesity and cancer. ACS Pharmacol Transl Sci. 7:560–569. 2024. View Article : Google Scholar : PubMed/NCBI

12 

Yang K, Wang X, Song C, He Z, Wang R, Xu Y, Jiang G, Wan Y, Mei J and Mao W: The role of lipid metabolic reprogramming in tumor microenvironment. Theranostics. 13:1774–1808. 2023. View Article : Google Scholar : PubMed/NCBI

13 

Chen J, Cui L, Lu S and Xu S: Amino acid metabolism in tumor biology and therapy. Cell Death Dis. 15:422024. View Article : Google Scholar : PubMed/NCBI

14 

Medina M: Metabolic reprogramming is a Hallmark of metabolism itself. Bioessays. 42:e20000582020. View Article : Google Scholar : PubMed/NCBI

15 

Wu D, Zhuo L and Wang X: Metabolic reprogramming of carcinoma-associated fibroblasts and its impact on metabolic heterogeneity of tumors. Semin Cell Dev Biol. 64:125–131. 2017. View Article : Google Scholar

16 

Avagliano A, Granato G, Ruocco MR, Romano V, Belviso I, Carfora A, Montagnani S and Arcucci A: Metabolic reprogramming of cancer associated fibroblasts: The slavery of stromal fibroblasts. Biomed Res Int. 2018:60754032018. View Article : Google Scholar : PubMed/NCBI

17 

Liang L, Li W, Li X, Jin X, Liao Q, Li Y and Zhou Y: 'Reverse Warburg effect' of cancer-associated fibroblasts (Review). Int J Oncol. 60:672022. View Article : Google Scholar

18 

Hu D, Li Z, Zheng B, Lin X, Pan Y, Gong P, Zhuo W, Hu Y, Chen C, Chen L, et al: Cancer-associated fibroblasts in breast cancer: Challenges and opportunities. Cancer Commun (Lond). 42:401–434. 2022. View Article : Google Scholar : PubMed/NCBI

19 

Yoon H, Tang CM, Banerjee S, Delgado AL, Yebra M, Davis J and Sicklick JK: TGF-β1-mediated transition of resident fibroblasts to cancer-associated fibroblasts promotes cancer metastasis in gastrointestinal stromal tumor. Oncogenesis. 10:132021. View Article : Google Scholar

20 

Hartupee J and Mann DL: Role of inflammatory cells in fibroblast activation. J Mol Cell Cardiol. 93:143–148. 2016. View Article : Google Scholar :

21 

Erez N, Truitt M, Olson P, Arron ST and Hanahan D: Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell. 17:135–147. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Sanz-Moreno V, Gaggioli C, Yeo M, Albrengues J, Wallberg F, Viros A, Hooper S, Mitter R, Féral CC, Cook M, et al: ROCK and JAK1 signaling cooperate to control actomyosin contractility in tumor cells and stroma. Cancer Cell. 20:229–245. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Shen H, Yu X, Yang F, Zhang Z, Shen J, Sun J, Choksi S, Jitkaew S and Shu Y: Reprogramming of normal fibroblasts into cancer-associated fibroblasts by miRNAs-Mediated CCL2/VEGFA signaling. PLoS Genet. 12:e10062442016. View Article : Google Scholar : PubMed/NCBI

24 

Calvo F, Ege N, Grande-Garcia A, Hooper S, Jenkins RP, Chaudhry SI, Harrington K, Williamson P, Moeendarbary E, Charras G and Sahai E: Mechanotransduction and YAP-dependent matrix remodelling is required for the generation and maintenance of cancer-associated fibroblasts. Nat Cell Biol. 15:637–646. 2013. View Article : Google Scholar : PubMed/NCBI

25 

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

26 

Yin C, Evason KJ, Asahina K and Stainier DY: Hepatic stellate cells in liver development, regeneration, and cancer. J Clin Invest. 123:1902–1910. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Omary MB, Lugea A, Lowe AW and Pandol SJ: The pancreatic stellate cell: A star on the rise in pancreatic diseases. J Clin Invest. 117:50–59. 2007. View Article : Google Scholar :

28 

Wang F, Li L, Piontek K, Sakaguchi M and Selaru FM: Exosome miR-335 as a novel therapeutic strategy in hepatocellular carcinoma. Hepatology. 67:940–954. 2018. View Article : Google Scholar

29 

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

30 

Shangguan L, Ti X, Krause U, Hai B, Zhao Y, Yang Z and Liu F: Inhibition of TGF-β/Smad signaling by BAMBI blocks differentiation of human mesenchymal stem cells to carcinoma-associated fibroblasts and abolishes their protumor effects. Stem Cells. 30:2810–2819. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Weber CE, Kothari AN, Wai PY, Li NY, Driver J, Zapf MA, Franzen CA, Gupta GN, Osipo C, Zlobin A, et al: Osteopontin mediates an MZF1-TGF-β1-dependent transformation of mesenchymal stem cells into cancer-associated fibroblasts in breast cancer. Oncogene. 34:4821–4833. 2015. View Article : Google Scholar

32 

Iwano M, Plieth D, Danoff TM, Xue C, Okada H and Neilson EG: Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest. 110:341–350. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Zeisberg EM, Potenta S, Xie L, Zeisberg M and Kalluri R: Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res. 67:10123–10128. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Tang PC, Chung JY, Xue VW, Xiao J, Meng XM, Huang XR, Zhou S, Chan AS, Tsang AC, Cheng AS, et al: Smad3 promotes cancer-associated fibroblasts generation via macrophage-myofibroblast transition. Adv Sci (Weinh). 9:e21012352022. View Article : Google Scholar

35 

Tang PM, Zhang YY, Xiao J, Tang PC, Chung JY, Li J, Xue VW, Huang XR, Chong CC, Ng CF, et al: Neural transcription factor Pou4f1 promotes renal fibrosis via macrophage-myofibroblast transition. Proc Natl Acad Sci USA. 117:20741–20752. 2020. View Article : Google Scholar

36 

Dulauroy S, Di Carlo SE, Langa F, Eberl G and Peduto L: Lineage tracing and genetic ablation of ADAM12(+) perivascular cells identify a major source of profibrotic cells during acute tissue injury. Nat Med. 18:1262–1270. 2012. View Article : Google Scholar : PubMed/NCBI

37 

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

38 

Wikström P, Marusic J, Stattin P and Bergh A: Low stroma androgen receptor level in normal and tumor prostate tissue is related to poor outcome in prostate cancer patients. Prostate. 69:799–809. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Gieniec KA, Butler LM, Worthley DL and Woods SL: Cancer-associated fibroblasts-heroes or villains? Br J Cancer. 121:293–302. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Kanzaki R and Pietras K: Heterogeneity of cancer-associated fibroblasts: Opportunities for precision medicine. Cancer Sci. 111:2708–2717. 2020. View Article : Google Scholar : PubMed/NCBI

41 

Nurmik M, Ullmann P, Rodriguez F, Haan S and Letellier E: In search of definitions: Cancer-associated fibroblasts and their markers. Int J Cancer. 146:895–905. 2020. View Article : Google Scholar

42 

Hu H, Piotrowska Z, Hare PJ, Chen H, Mulvey HE, Mayfield A, Noeen S, Kattermann K, Greenberg M, Williams A, et al: Three subtypes of lung cancer fibroblasts define distinct therapeutic paradigms. Cancer Cell. 39:1531–1547.e10. 2021. View Article : Google Scholar : PubMed/NCBI

43 

Zhang H, Yue X, Chen Z, Liu C, Wu W, Zhang N, Liu Z, Yang L, Jiang Q, Cheng Q, et al: Define cancer-associated fibroblasts (CAFs) in the tumor microenvironment: new opportunities in cancer immunotherapy and advances in clinical trials. Mol Cancer. 22:1592023. View Article : Google Scholar : PubMed/NCBI

44 

Luo H, Xia X, Huang LB, An H, Cao M, Kim GD, Chen HN, Zhang WH, Shu Y, Kong X, et al: Pan-cancer single-cell analysis reveals the heterogeneity and plasticity of cancer-associated fibroblasts in the tumor microenvironment. 13:66192022.PubMed/NCBI

45 

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

46 

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

47 

Cords L, Engler S, Haberecker M, Rüschoff JH, Moch H, de Souza N and Bodenmiller B: Cancer-associated fibroblast phenotypes are associated with patient outcome in non-small cell lung cancer. Cancer Cell. 42:396–412.e5. 2024. View Article : Google Scholar : PubMed/NCBI

48 

Li X, Sun Z, Peng G, Xiao Y, Guo J, Wu B, Li X, Zhou W, Li J, Li Z, et al: Single-cell RNA sequencing reveals a pro-invasive cancer-associated fibroblast subgroup associated with poor clinical outcomes in patients with gastric cancer. Theranostics. 12:620–638. 2022. View Article : Google Scholar :

49 

Roulis M, Kaklamanos A, Schernthanner M, Bielecki P, Zhao J, Kaffe E, Frommelt LS, Qu R, Knapp MS, Henriques A, et al: Paracrine orchestration of intestinal tumorigenesis by a mesenchymal niche. Nature. 580:524–529. 2020. View Article : Google Scholar :

50 

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 :

51 

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

52 

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

53 

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

54 

Wang Y, Liang Y, Xu H, Zhang X, Mao T, Cui J, Yao J, Wang Y, Jiao F, Xiao X, et al: Single-cell analysis of pancreatic ductal adenocarcinoma identifies a novel fibroblast subtype associated with poor prognosis but better immunotherapy response. Cell Discov. 7:362021. View Article : Google Scholar : PubMed/NCBI

55 

Niu N, Shen X, Wang Z, Chen Y, Weng Y, Yu F, Tang Y, Lu P, Liu M, Wang L, et al: Tumor cell-intrinsic epigenetic dysregulation shapes cancer-associated fibroblasts heterogeneity to metabolically support pancreatic cancer. Cancer Cell. 42:869–884.e9. 2024. View Article : Google Scholar

56 

Lambrechts D, Wauters E, Boeckx B, Aibar S, Nittner D, Burton O, Bassez A, Decaluwé H, Pircher A, Van den Eynde K, et al: Phenotype molding of stromal cells in the lung tumor microenvironment. Nat Med. 24:1277–1289. 2018. View Article : Google Scholar : PubMed/NCBI

57 

Foster DS, Januszyk M, Delitto D, Yost KE, Griffin M, Guo J, Guardino N, Delitto AE, Chinta M, Burcham AR, et al: Multiomic analysis reveals conservation of cancer-associated fibroblast phenotypes across species and tissue of origin. Cancer Cell. 40:1392–1406.e7. 2022. View Article : Google Scholar : PubMed/NCBI

58 

Ma C, Yang C, Peng A, Sun T, Ji X, Mi J, Wei L, Shen S and Feng Q: Pan-cancer spatially resolved single-cell analysis reveals the crosstalk between cancer-associated fibroblasts and tumor microenvironment. Mol Cancer. 22:1702023. View Article : Google Scholar : PubMed/NCBI

59 

Nong S, Han X, Xiang Y, Qian Y, Wei Y, Zhang T, Tian K, Shen K, Yang J and Ma X: Metabolic reprogramming in cancer: Mechanisms and therapeutics. MedComm (2020). 4:e2182023. View Article : Google Scholar

60 

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

61 

Mamun AA, Hayashi H, Yamamura A, Nayeem MJ and Sato M: Hypoxia induces the translocation of glucose transporter 1 to the plasma membrane in vascular endothelial cells. J Physiol Sci. 70:442020. View Article : Google Scholar : PubMed/NCBI

62 

Snell CE, Turley H, McIntyre A, Li D, Masiero M, Schofield CJ, Gatter KC, Harris AL and Pezzella F: Proline-hydroxylated hypoxia-inducible factor 1α (HIF-1α) upregulation in human tumours. PLoS One. 9:e889552014. View Article : Google Scholar

63 

Xu G, Li M, Wu J, Qin C, Tao Y and He H: Circular RNA circ-NRIP1 sponges microRNA-138-5p to maintain hypoxia-induced resistance to 5-fluorouracil through HIF-1α-dependent glucose metabolism in gastric carcinoma. Cancer Manag Res. 12:2789–2802. 2020. View Article : Google Scholar

64 

Zhang D, Wang Y, Shi Z, Liu J, Sun P, Hou X, Zhang J, Zhao S, Zhou BP and Mi J: Metabolic reprogramming of cancer-associated fibroblasts by IDH3α downregulation. Cell Rep. 10:1335–1348. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Rodríguez-García A, Samsó P, Fontova P, Simon-Molas H, Manzano A, Castaño E, Rosa JL, Martinez-Outshoorn U, Ventura F, Navarro-Sabaté À and Bartrons R: TGF-β1 targets Smad, p38 MAPK, and PI3K/Akt signaling pathways to induce PFKFB3 gene expression and glycolysis in glioblastoma cells. FEBS J. 284:3437–3454. 2017. View Article : Google Scholar

66 

Wei X, Hou Y, Long M, Jiang L and Du Y: Molecular mechanisms underlying the role of hypoxia-inducible factor-1 α in metabolic reprogramming in renal fibrosis. Front Endocrinol (Lausanne). 13:9273292022. View Article : Google Scholar

67 

Shi X, Yang J, Deng S, Xu H, Wu D, Zeng Q, Wang S, Hu T, Wu F and Zhou H: TGF-β signaling in the tumor metabolic microenvironment and targeted therapies. J Hematol Oncol. 15:1352022. View Article : Google Scholar

68 

Wang SF, Tseng LM and Lee HC: Role of mitochondrial alterations in human cancer progression and cancer immunity. J Biomed Sci. 30:612023. View Article : Google Scholar :

69 

Shimura T, Sasatani M, Kawai H, Kamiya K, Kobayashi J, Komatsu K and Kunugita N: Radiation-Induced myofibroblasts promote tumor growth via mitochondrial ROS-Activated TGFβ Signaling. Mol Cancer Res. 16:1676–1686. 2018. View Article : Google Scholar

70 

Chakraborty PK, Mustafi SB, Xiong X, Dwivedi SKD, Nesin V, Saha S, Zhang M, Dhanasekaran D, Jayaraman M, Mannel R, et al: MICU1 drives glycolysis and chemoresistance in ovarian cancer. Nat Commun. 8:146342017. View Article : Google Scholar : PubMed/NCBI

71 

Sung JS, Kang CW, Kang S, Jang Y, Chae YC, Kim BG and Cho NH: ITGB4-mediated metabolic reprogramming of cancer-associated fibroblasts. Oncogene. 39:664–676. 2020. View Article : Google Scholar

72 

Li X, Yang Y, Zhang B, Lin X, Fu X, An Y, Zou Y, Wang JX, Wang Z and Yu T: Lactate metabolism in human health and disease. Signal Transduct Target Ther. 7:3052022. View Article : Google Scholar : PubMed/NCBI

73 

Qiao Y, Liu Y, Ran R, Zhou Y, Gong J, Liu L, Zhang Y, Wang H, Fan Y, Fan Y, et al: Lactate metabolism and lactylation in breast cancer: mechanisms and implications. Cancer Metastasis Rev. 44:482025. View Article : Google Scholar :

74 

Kozlov AM, Lone A, Betts DH and Cumming RC: Lactate preconditioning promotes a HIF-1α-mediated metabolic shift from OXPHOS to glycolysis in normal human diploid fibroblasts. Sci Rep. 10:83882020. View Article : Google Scholar

75 

Ishihara S, Hata K, Hirose K, Okui T, Toyosawa S, Uzawa N, Nishimura R and Yoneda T: The lactate sensor GPR81 regulates glycolysis and tumor growth of breast cancer. Sci Rep. 12:62612022. View Article : Google Scholar : PubMed/NCBI

76 

Luo M, Zhu J, Ren J, Tong Y, Wang L, Ma S and Wang J: Lactate increases tumor malignancy by promoting tumor small extracellular vesicles production via the GPR81-cAMP-PKA-HIF-1α axis. Front Oncol. 12:10365432022. View Article : Google Scholar

77 

Yang L, Gilbertsen A, Xia H, Benyumov A, Smith K, Herrera J, Racila E, Bitterman PB and Henke CA: Hypoxia enhances IPF mesenchymal progenitor cell fibrogenicity via the lactate/GPR81/HIF1α pathway. JCI insight. 8:e1638202023. View Article : Google Scholar

78 

Fontana F, Giannitti G, Marchesi S and Limonta P: The PI3K/Akt pathway and glucose metabolism: A dangerous liaison in cancer. Int J Biol Sci. 20:3113–3125. 2024. View Article : Google Scholar : PubMed/NCBI

79 

Payen VL, Porporato PE, Baselet B and Sonveaux P: Metabolic changes associated with tumor metastasis, part 1: Tumor pH, glycolysis and the pentose phosphate pathway. Cell Mol Life Sci. 73:1333–1348. 2016. View Article : Google Scholar

80 

Mestre-Farrera A, Bruch-Oms M, Peña R, Rodríguez-Morató J, Alba-Castellón L, Comerma L, Quintela-Fandino M, Duñach M, Baulida J, Pozo ÓJ and García de Herreros A: Glutamine-directed migration of cancer-activated fibroblasts facilitates epithelial tumor invasion. Cancer Res. 81:438–451. 2021. View Article : Google Scholar

81 

He C, Peng M, Zeng X, Dong H, Sun Z, Xu J, Liu M, Liu L, Huang Y, Peng Z, et al: Microenvironmental G protein-coupled estrogen receptor-mediated glutamine metabolic coupling between cancer-associated fibroblasts and triple-negative breast cancer cells governs tumour progression. Clin Transl Med. 14:e701312024. View Article : Google Scholar : PubMed/NCBI

82 

Sousa CM, Biancur DE, Wang X, Halbrook CJ, Sherman MH, Zhang L, Kremer D, Hwang RF, Witkiewicz AK, Ying H, et al: Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature. 536:479–483. 2016. View Article : Google Scholar : PubMed/NCBI

83 

Kay EJ, Paterson K, Riera-Domingo C, Sumpton D, Däbritz JHM, Tardito S, Boldrini C, Hernandez-Fernaud JR, Athineos D, Dhayade S, et al: Cancer-associated fibroblasts require proline synthesis by PYCR1 for the deposition of pro-tumorigenic extracellular matrix. Nat Metab. 4:693–710. 2022. View Article : Google Scholar : PubMed/NCBI

84 

Kay EJ, Zanivan S and Rufini A: Proline metabolism shapes the tumor microenvironment: From collagen deposition to immune evasion. Curr Opin Biotechnol. 84:1030112023. View Article : Google Scholar : PubMed/NCBI

85 

Huynh TYL, Zareba I, Baszanowska W, Lewoniewska S and Palka J: Understanding the role of key amino acids in regulation of proline dehydrogenase/proline oxidase (prodh/pox)-dependent apoptosis/autophagy as an approach to targeted cancer therapy. Mol Cell Biochem. 466:35–44. 2020. View Article : Google Scholar

86 

Ino Y, Yamazaki-Itoh R, Oguro S, Shimada K, Kosuge T, Zavada J, Kanai Y and Hiraoka N: Arginase II expressed in cancer-associated fibroblasts indicates tissue hypoxia and predicts poor outcome in patients with pancreatic cancer. PLoS One. 8:e551462013. View Article : Google Scholar : PubMed/NCBI

87 

Jin HR, Wang J, Wang ZJ, Xi MJ, Xia BH, Deng K and Yang JL: Lipid metabolic reprogramming in tumor microenvironment: From mechanisms to therapeutics. J Hematol Oncol. 16:1032023. View Article : Google Scholar : PubMed/NCBI

88 

Jabbari K, Cheng Q, Winkelmaier G, Furuta S and Parvin B: CD36(+) fibroblasts secrete protein ligands that growth-suppress triple-negative breast cancer cells while elevating adipogenic markers for a model of cancer-associated fibroblast. Int J Mol Sci. 23:127442022. View Article : Google Scholar : PubMed/NCBI

89 

Zhu GQ, Tang Z, Huang R, Qu WF, Fang Y, Yang R, Tao CY, Gao J, Wu XL, Sun HX, et al: CD36(+) cancer-associated fibroblasts provide immunosuppressive microenvironment for hepatocellular carcinoma via secretion of macrophage migration inhibitory factor. Cell Discov. 9:252023. View Article : Google Scholar

90 

Lopes-Coelho F, André S, Félix A and Serpa J: Breast cancer metabolic cross-talk: Fibroblasts are hubs and breast cancer cells are gatherers of lipids. Mol Cell Endocrinol. 462(Pt B): 93–106. 2018. View Article : Google Scholar

91 

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

92 

Zhang Y, Gu Z, Wan J, Lou X, Liu S, Wang Y, Bian Y, Wang F, Li Z and Qin Z: Stearoyl-CoA Desaturase-1 dependent lipid droplets accumulation in cancer-associated fibroblasts facilitates the progression of lung cancer. Int J Biol Sci. 18:6114–6128. 2022. View Article : Google Scholar :

93 

Santi A, Caselli A, Ranaldi F, Paoli P, Mugnaioni C, Michelucci E and Cirri P: Cancer associated fibroblasts transfer lipids and proteins to cancer cells through cargo vesicles supporting tumor growth. Biochim Biophys Acta. 1853:3211–3223. 2015. View Article : Google Scholar

94 

Beach JA, Aspuria PJ, Cheon DJ, Lawrenson K, Agadjanian H, Walsh CS, Karlan BY and Orsulic S: Sphingosine kinase 1 is required for TGF-β mediated fibroblastto-myofibroblast differentiation in ovarian cancer. Oncotarget. 7:4167–4182. 2016. View Article : Google Scholar

95 

Auciello FR, Bulusu V, Oon C, Tait-Mulder J, Berry M, Bhattacharyya S, Tumanov S, Allen-Petersen BL, Link J, Kendsersky ND, et al: A stromal lysolipid-autotaxin signaling axis promotes pancreatic tumor progression. Cancer Discov. 9:617–627. 2019. View Article : Google Scholar :

96 

Charo C, Holla V, Arumugam T, Hwang R, Yang P, Dubois RN, Menter DG, Logsdon CD and Ramachandran V: Prostaglandin E2 regulates pancreatic stellate cell activity via the EP4 receptor. Pancreas. 42:467–474. 2013. View Article : Google Scholar :

97 

Martinez-Outschoorn UE, Lin Z, Whitaker-Menezes D, Howell A, Lisanti MP and Sotgia F: Ketone bodies and two-compartment tumor metabolism: Stromal ketone production fuels mitochondrial biogenesis in epithelial cancer cells. Cell Cycle. 11:3956–3963. 2012. View Article : Google Scholar :

98 

Chirieac LR: Tumor cell proliferation, proliferative index and mitotic count in lung cancer. Transl Lung Cancer Res. 5:554–556. 2016. View Article : Google Scholar : PubMed/NCBI

99 

Feitelson MA, Arzumanyan A, Kulathinal RJ, Blain SW, Holcombe RF, Mahajna J, Marino M, Martinez-Chantar ML, Nawroth R, Sanchez-Garcia I, et al: Sustained proliferation in cancer: Mechanisms and novel therapeutic targets. Semin Cancer Biol. 35(Suppl): S25–S54. 2015. View Article : Google Scholar

100 

Prieto-Fernández L, Montoro-Jiménez I, de Luxan-Delgado B, Otero-Rosales M, Rodrigo JP, Calvo F, García-Pedrero JM and Álvarez-Teijeiro S: Dissecting the functions of cancer-associated fibroblasts to therapeutically target head and neck cancer microenvironment. Biomed Pharmacother. 161:1145022023. View Article : Google Scholar : PubMed/NCBI

101 

Martinez-Outschoorn UE, Lisanti MP and Sotgia F: Catabolic cancer-associated fibroblasts transfer energy and biomass to anabolic cancer cells, fueling tumor growth. Semin Cancer Biol. 25:47–60. 2014. View Article : Google Scholar : PubMed/NCBI

102 

Fiaschi T, Marini A, Giannoni E, Taddei ML, Gandellini P, De Donatis A, Lanciotti M, Serni S, Cirri P and Chiarugi P: Reciprocal metabolic reprogramming through lactate shuttle coordinately influences tumor-stroma interplay. Cancer Res. 72:5130–5140. 2012. View Article : Google Scholar : PubMed/NCBI

103 

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

104 

Li Y, Zhao Z, Liu W and Li X: SNHG3 Functions as miRNA sponge to promote breast cancer cells growth through the metabolic reprogramming. Appl Biochem Biotechnol. 191:1084–1099. 2020. View Article : Google Scholar : PubMed/NCBI

105 

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

106 

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

107 

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

108 

Linares JF, Cordes T, Duran A, Reina-Campos M, Valencia T, Ahn CS, Castilla EA, Moscat J, Metallo CM and Diaz-Meco MT: ATF4-induced metabolic reprograming is a synthetic vulnerability of the p62-deficient tumor stroma. Cell Metab. 26:817–829.e6. 2017. View Article : Google Scholar : PubMed/NCBI

109 

Mishra R, Haldar S, Placencio V, Madhav A, Rohena-Rivera K, Agarwal P, Duong F, Angara B, Tripathi M, Liu Z, et al: Stromal epigenetic alterations drive metabolic and neuroendocrine prostate cancer reprogramming. J Clin Invest. 128:4472–4484. 2018. View Article : Google Scholar :

110 

Gerashchenko TS, Novikov NM, Krakhmal NV, Zolotaryova SY, Zavyalova MV, Cherdyntseva NV, Denisov EV and Perelmuter VM: Markers of cancer cell invasion: Are they good enough? J Clin Med. 8:10922019. View Article : Google Scholar : PubMed/NCBI

111 

Bànkfalvi A and Piffkò J: Prognostic and predictive factors in oral cancer: The role of the invasive tumour front. J Oral Pathol Med. 29:291–298. 2000. View Article : Google Scholar

112 

Sahai E: Mechanisms of cancer cell invasion. Curr Opin Genet Dev. 15:87–96. 2005. View Article : Google Scholar : PubMed/NCBI

113 

Wirtz D, Konstantopoulos K and Searson PC: The physics of cancer: The role of physical interactions and mechanical forces in metastasis. Nat Rev Cancer. 11:512–522. 2011. View Article : Google Scholar : PubMed/NCBI

114 

Nagelkerke A, Bussink J, Rowan AE and Span PN: The mechanical microenvironment in cancer: How physics affects tumours. Semin Cancer Biol. 35:62–70. 2015. View Article : Google Scholar : PubMed/NCBI

115 

Liu QP, Luo Q, Deng B, Ju Y and Song GB: Stiffer matrix accelerates migration of hepatocellular carcinoma cells through enhanced aerobic glycolysis via the MAPK-YAP signaling. Cancers (Basel). 12:4902020. View Article : Google Scholar : PubMed/NCBI

116 

Wu X, Zhou Z, Xu S, Liao C, Chen X, Li B, Peng J, Li D and Yang L: Extracellular vesicle packaged LMP1-activated fibroblasts promote tumor progression via autophagy and stroma-tumor metabolism coupling. Cancer Lett. 478:93–106. 2020. View Article : Google Scholar : PubMed/NCBI

117 

Gong J, Lin Y, Zhang H, Liu C, Cheng Z, Yang X, Zhang J, Xiao Y, Sang N, Qian X, et al: Reprogramming of lipid metabolism in cancer-associated fibroblasts potentiates migration of colorectal cancer cells. Cell Death Dis. 11:2672020. View Article : Google Scholar : PubMed/NCBI

118 

Wang H, Liu F, Wu X, Zhu G, Tang Z, Qu W, Zhao Q, Huang R, Tian M, Fang Y, et al: Cancer-associated fibroblasts contributed to hepatocellular carcinoma recurrence and metastasis via CD36-mediated fatty-acid metabolic reprogramming. Exp Cell Res. 435:1139472024. View Article : Google Scholar : PubMed/NCBI

119 

Sun K, Tang S, Hou Y, Xi L, Chen Y, Yin J, Peng M, Zhao M, Cui X and Liu M: Oxidized ATM-mediated glycolysis enhancement in breast cancer-associated fibroblasts contributes to tumor invasion through lactate as metabolic coupling. EBioMedicine. 41:370–383. 2019. View Article : Google Scholar : PubMed/NCBI

120 

Shan T, Chen S, Chen X, Lin WR, Li W, Ma J, Wu T, Cui X, Ji H, Li Y and Kang Y: Cancer-associated fibroblasts enhance pancreatic cancer cell invasion by remodeling the metabolic conversion mechanism. Oncol Rep. 37:1971–1979. 2017. View Article : Google Scholar : PubMed/NCBI

121 

Siemann DW and Horsman MR: Modulation of the tumor vasculature and oxygenation to improve therapy. Pharmacol Ther. 153:107–124. 2015. View Article : Google Scholar : PubMed/NCBI

122 

Ribatti D and Pezzella F: Overview on the different patterns of tumor vascularization. Cells. 10:6392021. View Article : Google Scholar : PubMed/NCBI

123 

Katayama Y, Uchino J, Chihara Y, Tamiya N, Kaneko Y, Yamada T and Takayama K: Tumor neovascularization and developments in therapeutics. Cancers (Basel). 11:3162019. View Article : Google Scholar : PubMed/NCBI

124 

Dudley AC and Griffioen AW: Pathological angiogenesis: Mechanisms and therapeutic strategies. Angiogenesis. 26:313–347. 2023. View Article : Google Scholar : PubMed/NCBI

125 

Zhu Y, Li X, Wang L, Hong X and Yang J: Metabolic reprogramming and crosstalk of cancer-related fibroblasts and immune cells in the tumor microenvironment. Front Endocrinol (Lausanne). 13:9882952022. View Article : Google Scholar : PubMed/NCBI

126 

Bonuccelli G, Whitaker-Menezes D, Castello-Cros R, Pavlides S, Pestell RG, Fatatis A, Witkiewicz AK, Vander Heiden MG, Migneco G, Chiavarina B, et al: The reverse Warburg effect: glycolysis inhibitors prevent the tumor promoting effects of caveolin-1 deficient cancer associated fibroblasts. Cell Cycle. 9:1960–1971. 2010. View Article : Google Scholar : PubMed/NCBI

127 

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

128 

Li X, Jiang E, Zhao H, Chen Y, Xu Y, Feng C, Li J and Shang Z: Glycometabolic reprogramming-mediated proangiogenic phenotype enhancement of cancer-associated fibroblasts in oral squamous cell carcinoma: role of PGC-1α/PFKFB3 axis. Br J Cancer. 127:449–461. 2022. View Article : Google Scholar : PubMed/NCBI

129 

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

130 

Hsu WH, LaBella KA, Lin Y, Xu P, Lee R, Hsieh CE, Yang L, Zhou A, Blecher JM, Wu CJ, et al: Oncogenic KRAS drives lipofibrogenesis to promote angiogenesis and colon cancer progression. Cancer Discov. 13:2652–2673. 2023. View Article : Google Scholar : PubMed/NCBI

131 

Verginadis II, Avgousti H, Monslow J, Skoufos G, Chinga F, Kim K, Leli NM, Karagounis IV, Bell BI, Velalopoulou A, et al: A stromal integrated stress response activates perivascular cancer-associated fibroblasts to drive angiogenesis and tumour progression. Nat Cell Biol. 24:940–953. 2022. View Article : Google Scholar : PubMed/NCBI

132 

Qian CN, Mei Y and Zhang J: Cancer metastasis: Issues and challenges. Chin J Cancer. 36:382017. View Article : Google Scholar : PubMed/NCBI

133 

Fares J, Fares MY, Khachfe HH, Salhab HA and Fares Y: Molecular principles of metastasis: A hallmark of cancer revisited. Signal Transduct Target Ther. 5:282020. View Article : Google Scholar : PubMed/NCBI

134 

Eble JA and Niland S: The extracellular matrix in tumor progression and metastasis. Clin Exp Metastasis. 36:171–198. 2019. View Article : Google Scholar : PubMed/NCBI

135 

Curtis M, Kenny HA, Ashcroft B, Mukherjee A, Johnson A, Zhang Y, Helou Y, Batlle R, Liu X, Gutierrez N, et al: Fibroblasts mobilize tumor cell glycogen to promote proliferation and metastasis. Cell Metab. 29:141–155.e9. 2019. View Article : Google Scholar :

136 

Wang Y, Wang X, Bai B, Shaha A, He X, He Y, Ye Z, Shah VH and Kang N: Targeting Src SH3 domain-mediated glycolysis of HSC suppresses transcriptome, myofibroblastic activation, and colorectal liver metastasis. Hepatology. 80:578–594. 2024. View Article : Google Scholar : PubMed/NCBI

137 

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

138 

Li Q, Zhu CC, Ni B, Zhang ZZ, Jiang SH, Hu LP, Wang X, Zhang XX, Huang PQ, Yang Q, et al: Lysyl oxidase promotes liver metastasis of gastric cancer via facilitating the reciprocal interactions between tumor cells and cancer associated fibroblasts. EBioMedicine. 49:157–171. 2019. View Article : Google Scholar : PubMed/NCBI

139 

Tian Y, Wang X, Wu C, Qiao J, Jin H and Li H: A protracted war against cancer drug resistance. Cancer Cell Int. 24:3262024. View Article : Google Scholar : PubMed/NCBI

140 

Lei ZN, Tian Q, Teng QX, Wurpel JND, Zeng L, Pan Y and Chen ZS: Understanding and targeting resistance mechanisms in cancer. MedComm (2020). 4:e2652023. View Article : Google Scholar : PubMed/NCBI

141 

Dhanyamraju PK: Drug resistance mechanisms in cancers: Execution of pro-survival strategies. J Biomed Res. 38:95–121. 2024. View Article : Google Scholar : PubMed/NCBI

142 

Zaal EA and Berkers CR: The influence of metabolism on drug response in cancer. Front Oncol. 8:5002018. View Article : Google Scholar : PubMed/NCBI

143 

Broekgaarden M, Anbil S, Bulin AL, Obaid G, Mai Z, Baglo Y, Rizvi I and Hasan T: Modulation of redox metabolism negates cancer-associated fibroblasts-induced treatment resistance in a heterotypic 3D culture platform of pancreatic cancer. Biomaterials. 222:1194212019. View Article : Google Scholar : PubMed/NCBI

144 

Ko YH, Lin Z, Flomenberg N, Pestell RG, Howell A, Sotgia F, Lisanti MP and Martinez-Outschoorn UE: Glutamine fuels a vicious cycle of autophagy in the tumor stroma and oxidative mitochondrial metabolism in epithelial cancer cells: Implications for preventing chemotherapy resistance. Cancer Biol Ther. 12:1085–1097. 2011. View Article : Google Scholar

145 

Raez LE, Papadopoulos K, Ricart AD, Chiorean EG, Dipaola RS, Stein MN, Rocha Lima CM, Schlesselman JJ, Tolba K, Langmuir VK, et al: A phase I dose-escalation trial of 2-deoxy-D-glucose alone or combined with docetaxel in patients with advanced solid tumors. Cancer Chemother Pharmacol. 71:523–530. 2013. View Article : Google Scholar

146 

Mohanti BK, Rath GK, Anantha N, Kannan V, Das BS, Chandramouli BA, Banerjee AK, Das S, Jena A, Ravichandran R, et al: Improving cancer radiotherapy with 2-deoxy-D-glucose: Phase I/II clinical trials on human cerebral gliomas. Int J Radiat Oncol Biol Phys. 35:103–111. 1996. View Article : Google Scholar : PubMed/NCBI

147 

Di Cosimo S, Ferretti G, Papaldo P, Carlini P, Fabi A and Cognetti F: Lonidamine: Efficacy and safety in clinical trials for the treatment of solid tumors. Drugs Today (Barc). 39:157–174. 2003. View Article : Google Scholar : PubMed/NCBI

148 

Kelly W, Diaz Duque AE, Michalek J, Konkel B, Caflisch L, Chen Y, Pathuri SC, Madhusudanannair-Kunnuparampil V, Floyd J and Brenner A: Phase II investigation of TVB-2640 (Denifanstat) with bevacizumab in patients with first relapse high-grade astrocytoma. Clin Cancer Res. 29:2419–2425. 2023. View Article : Google Scholar : PubMed/NCBI

149 

Svensson RU, Parker SJ, Eichner LJ, Kolar MJ, Wallace M, Brun SN, Lombardo PS, Van Nostrand JL, Hutchins A, Vera L, et al: Inhibition of acetyl-CoA carboxylase suppresses fatty acid synthesis and tumor growth of non-small-cell lung cancer in preclinical models. Nat Med. 22:1108–1119. 2016. View Article : Google Scholar : PubMed/NCBI

150 

Schlaepfer IR and Joshi M: CPT1A-mediated fat oxidation, mechanisms, and therapeutic potential. Endocrinology. 161:bqz0462020. View Article : Google Scholar : PubMed/NCBI

151 

Yao CH, Liu GY, Wang R, Moon SH, Gross RW and Patti GJ: Identifying off-target effects of etomoxir reveals that carnitine palmitoyltransferase I is essential for cancer cell proliferation independent of β-oxidation. PLoS Biol. 16:e20037822018. View Article : Google Scholar

152 

Gugiatti E, Tenca C, Ravera S, Fabbi M, Ghiotto F, Mazzarello AN, Bagnara D, Reverberi D, Zarcone D, Cutrona G, et al: A reversible carnitine palmitoyltransferase (CPT1) inhibitor offsets the proliferation of chronic lymphocytic leukemia cells. Haematologica. 103:e531–e536. 2018. View Article : Google Scholar : PubMed/NCBI

153 

Harding JJ, Telli M, Munster P, Voss MH, Infante JR, DeMichele A, Dunphy M, Le MH, Molineaux C, Orford K, et al: A Phase I Dose-Escalation and Expansion Study of Telaglenastat in Patients with Advanced or Metastatic Solid Tumors. Clin Cancer Res. 27:4994–5003. 2021. View Article : Google Scholar : PubMed/NCBI

154 

Yang WH, Qiu Y, Stamatatos O, Janowitz T and Lukey MJ: Enhancing the Efficacy of Glutamine Metabolism Inhibitors in Cancer Therapy. Trends Cancer. 7:790–804. 2021. View Article : Google Scholar : PubMed/NCBI

155 

Wicker CA, Hunt BG, Krishnan S, Aziz K, Parajuli S, Palackdharry S, Elaban WR, Wise-Draper TM, Mills GB, Waltz SE and Takiar V: Glutaminase inhibition with telaglenastat (CB-839) improves treatment response in combination with ionizing radiation in head and neck squamous cell carcinoma models. Cancer Lett. 502:180–188. 2021. View Article : Google Scholar : PubMed/NCBI

156 

Han J, Li Q, Chen Y and Yang Y: Recent metabolomics analysis in tumor metabolism reprogramming. Front Mol Biosci. 8:7639022021. View Article : Google Scholar : PubMed/NCBI

157 

Ciavardelli D, Bellomo M, Consalvo A, Crescimanno C and Vella V: Metabolic alterations of thyroid cancer as potential therapeutic targets. Biomed Res Int. 2017:25450312017. View Article : Google Scholar : PubMed/NCBI

158 

Wang YA, Li XL, Mo YZ, Fan CM, Tang L, Xiong F, Guo C, Xiang B, Zhou M, Ma J, et al: Effects of tumor metabolic microenvironment on regulatory T cells. Mol Cancer. 17:1682018. View Article : Google Scholar : PubMed/NCBI

159 

Wright K, Ly T, Kriet M, Czirok A and Thomas SM: Cancer-Associated Fibroblasts: Master tumor microenvironment modifiers. Cancers (Basel). 15:18992023. View Article : Google Scholar : PubMed/NCBI

160 

Sun L, Yang X, Yuan Z and Wang H: Metabolic reprogramming in immune response and tissue inflammation. Arterioscler Thromb Vasc Biol. 40:1990–2001. 2020. View Article : Google Scholar : PubMed/NCBI

161 

Lindner T, Loktev A, Giesel F, Kratochwil C, Altmann A and Haberkorn U: Targeting of activated fibroblasts for imaging and therapy. EJNMMI Radiopharm Chem. 4:162019. View Article : Google Scholar : PubMed/NCBI

162 

Yang D, Liu J, Qian H and Zhuang Q: Cancer-associated fibroblasts: From basic science to anticancer therapy. Exp Mol Med. 55:1322–1332. 2023. View Article : Google Scholar : PubMed/NCBI

163 

Zhao Z, Li T, Sun L, Yuan Y and Zhu Y: Potential mechanisms of cancer-associated fibroblasts in therapeutic resistance. Biomed Pharmacother. 166:1154252023. View Article : Google Scholar : PubMed/NCBI

164 

Wang M, Xue W, Yuan H, Wang Z and Yu L: Nano-Drug delivery systems targeting CAFs: A promising treatment for pancreatic cancer. Int J Nanomedicine. 19:2823–2849. 2024. View Article : Google Scholar : PubMed/NCBI

165 

Giuliani S, Accetta C, di Martino S, De Vitis C, Messina E, Pescarmona E, Fanciulli M, Ciliberto G, Mancini R and Falcone I: Metabolic reprogramming in melanoma: An epigenetic point of view. Pharmaceuticals (Basel). 18:8532025. View Article : Google Scholar : PubMed/NCBI

166 

Kumar D, New J, Vishwakarma V, Joshi R, Enders J, Lin F, Dasari S, Gutierrez WR, Leef G, Ponnurangam S, et al: Cancer-Associated fibroblasts drive glycolysis in a targetable signaling loop implicated in head and neck squamous cell carcinoma progression. Cancer Res. 78:3769–3782. 2018. View Article : Google Scholar : PubMed/NCBI

167 

Francescone R, Barbosa Vendramini-Costa D, Franco-Barraza J, Wagner J, Muir A, Lau AN, Gabitova L, Pazina T, Gupta S, Luong T, et al: Netrin G1 promotes pancreatic tumorigenesis through cancer-associated fibroblast-driven nutritional support and immunosuppression. Cancer Discov. 11:446–479. 2021. View Article : Google Scholar

168 

Balaban S, Nassar ZD, Zhang AY, Hosseini-Beheshti E, Centenera MM, Schreuder M, Lin HM, Aishah A, Varney B, Liu-Fu F, et al: Extracellular fatty acids are the major contributor to lipid synthesis in prostate cancer. Mol Cancer Res. 17:949–962. 2019. View Article : Google Scholar : PubMed/NCBI

169 

Yu M, Guo G, Huang L, Deng L, Chang CS, Achyut BR, Canning M, Xu N, Arbab AS, Bollag RJ, et al: CD73 on cancer-associated fibroblasts enhanced by the A2B-mediated feedforward circuit enforces an immune checkpoint. Nat Commun. 11:5152020. View Article : Google Scholar

170 

Broz MT, Ko EY, Ishaya K, Xiao J, De Simone M, Hoi XP, Piras R, Gala B, Tessaro FHG, Karlstaedt A, et al: Metabolic targeting of cancer associated fibroblasts overcomes T-cell exclusion and chemoresistance in soft-tissue sarcomas. Nat Commun. 15:24982024. View Article : Google Scholar : PubMed/NCBI

171 

Emberley E, Pan A, Chen J, Dang R, Gross M, Huang T, Li W, MacKinnon A, Singh D, Sotirovska N, et al: The glutaminase inhibitor telaglenastat enhances the antitumor activity of signal transduction inhibitors everolimus and cabozantinib in models of renal cell carcinoma. PLoS One. 16:e02592412021. View Article : Google Scholar : PubMed/NCBI

172 

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 :

173 

Wang Z, Tang Y, Tan Y, Wei Q and Yu W: Cancer-associated fibroblasts in radiotherapy: Challenges and new opportunities. Cell Commun Signal. 17:472019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2025
Volume 67 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li R and Li Y: Role of metabolic reprogramming of cancer‑associated fibroblasts in tumor development and progression (Review). Int J Oncol 67: 90, 2025.
APA
Li, R., & Li, Y. (2025). Role of metabolic reprogramming of cancer‑associated fibroblasts in tumor development and progression (Review). International Journal of Oncology, 67, 90. https://doi.org/10.3892/ijo.2025.5796
MLA
Li, R., Li, Y."Role of metabolic reprogramming of cancer‑associated fibroblasts in tumor development and progression (Review)". International Journal of Oncology 67.5 (2025): 90.
Chicago
Li, R., Li, Y."Role of metabolic reprogramming of cancer‑associated fibroblasts in tumor development and progression (Review)". International Journal of Oncology 67, no. 5 (2025): 90. https://doi.org/10.3892/ijo.2025.5796