The role of cancer-associated fibroblasts in the tumour microenvironment of urinary system

Ri Hong , Puguang Yu , Xiaoli Zhang , Peng Su , Hongyuan Liang , Dan Dong , Xuesong Wang , Kefeng Wang

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (4) : e70299

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (4) : e70299 DOI: 10.1002/ctm2.70299
REVIEW

The role of cancer-associated fibroblasts in the tumour microenvironment of urinary system

Author information +
History +
PDF

Abstract

Urological tumours are a type of neoplasms that significantly jeopardise human life and wellbeing. Cancer-associated fibroblasts (CAFs), serving as the primary component of the stromal cellular milieu, form a diverse cellular cohort that exerts substantial influence on tumourigenesis and tumour progression. In this review, we summarised the literatures regarding the functions of CAFs in the urinary tumour microenvironment (TME). We primarily examined the multifaceted activities of CAFs in the TME of urological system tumours, including inhibiting tumour immunity, remodelling the extracellular matrix, promoting tumour growth, metastasis, drug resistance and their clinical applications. We also discussed potential future directions for leveraging artificial intelligence in CAFs research.

Keywords

cancer-associated fibroblasts / exosomes / extracellular matrix / immune system / tumour microenvironment / urological tumours

Cite this article

Download citation ▾
Ri Hong, Puguang Yu, Xiaoli Zhang, Peng Su, Hongyuan Liang, Dan Dong, Xuesong Wang, Kefeng Wang. The role of cancer-associated fibroblasts in the tumour microenvironment of urinary system. Clinical and Translational Medicine, 2025, 15(4): e70299 DOI:10.1002/ctm2.70299

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA: Cancer J Clin. 2024; 74(1): 12-49.

[2]

Ziaee S, Chu GCY, Huang JM, Sieh S, Chung LWK. Prostate cancer metastasis: roles of recruitment and reprogramming, cell signal network and three-dimensional growth characteristics. Transl Androl Urol. 2015; 4(4): 438-454.

[3]

Wu X, Peng M, Huang B, et al. Immune microenvironment profiles of tumor immune equilibrium and immune escape states of mouse sarcoma. Cancer Lett. 2013; 340(1): 124-133.

[4]

Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science. 2011; 331(6024): 1565-1570.

[5]

Glaviano A, Lau HSH, Carter LM, et al. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol. 2025; 18(1): 6.

[6]

Yin CL, Ma YJ. The regulatory mechanism of hypoxia-inducible factor 1 and its clinical significance. Curr Mol Pharmacol. 2024; 17: e18761429266116.

[7]

Manohar SM. At the crossroads of TNF α signaling and cancer. Curr Mol Pharmacol. 2024; 17(1): e060923220758.

[8]

Zhang D, Tang Z, Huang H, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019; 574(7779): 575-580.

[9]

Qiong L, Shuyao X, Shan X, et al. Recent advances in the glycolytic processes linked to tumor metastasis. Curr Mol Pharmacol. 2024; 17: e18761429308361.

[10]

Xiao Y, Yu D. Tumor microenvironment as a therapeutic target in cancer. Pharmacol Ther. 2021; 221: 107753.

[11]

Peng S, Lin A, Jiang A, et al. CTLs heterogeneity and plasticity: implications for cancer immunotherapy. Mol Cancer. 2024; 23(1): 58.

[12]

Gao Z, Bai Y, Lin A, et al. Gamma delta T-cell-based immune checkpoint therapy: attractive candidate for antitumor treatment. Mol Cancer. 2023; 22(1): 31.

[13]

Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016; 16(9): 582-598.

[14]

Harper J, Sainson RCA. Regulation of the anti-tumour immune response by cancer-associated fibroblasts. Semin Cancer Biol. 2014; 25: 69-77.

[15]

Sahai E, Astsaturov I, Cukierman E, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 2020; 20(3): 174-186.

[16]

Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res. 2007; 67(21): 10123-10128.

[17]

Petersen OW, Nielsen HL, Gudjonsson T, et al. Epithelial to mesenchymal transition in human breast cancer can provide a nonmalignant stroma. Am J Pathol. 2003; 162(2): 391-402.

[18]

Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009; 119(6): 1420-1428.

[19]

Mishra PJ, Mishra PJ, Humeniuk R, et al. Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells. Cancer Res. 2008; 68(11): 4331-4339.

[20]

Jung Y, Kim JK, Shiozawa Y, et al. Recruitment of mesenchymal stem cells into prostate tumours promotes metastasis. Nat Commun. 2013; 4: 1795.

[21]

Barcellos-de-Souza P, Comito G, Pons-Segura C, et al. Mesenchymal stem cells are recruited and activated into carcinoma-associated fibroblasts by prostate cancer microenvironment-derived TGF-β1. Stem Cells (Dayton, Ohio). 2016; 34(10): 2536-2547.

[22]

Mishra R, Haldar S, Placencio V, et al. Stromal epigenetic alterations drive metabolic and neuroendocrine prostate cancer reprogramming. J Clin Invest. 2018; 128(10): 4472-4484.

[23]

Augsten M. Cancer-associated fibroblasts as another polarized cell type of the tumor microenvironment. Front Oncol. 2014; 4: 62.

[24]

Ishii G, Ochiai A, Neri S. Phenotypic and functional heterogeneity of cancer-associated fibroblast within the tumor microenvironment. Adv Drug Delivery Rev. 2016; 99(Pt B): 186-196.

[25]

Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature. 2003; 425(6958): 577-584.

[26]

Öhlund D, Handly-Santana A, Biffi G, et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med. 2017; 214(3): 579-596.

[27]

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

[28]

Erez N, Truitt M, Olson P, Arron ST, Hanahan D. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell. 2010; 17(2): 135-147.

[29]

Miyai Y, Esaki N, Takahashi M, Enomoto A. Cancer-associated fibroblasts that restrain cancer progression: hypotheses and perspectives. Cancer Sci. 2020; 111(4): 1047-1057.

[30]

Ando R, Sakai A, Iida T, Kataoka K, Mizutani Y, Enomoto A. Good and bad stroma in pancreatic cancer: relevance of functional states of cancer-associated fibroblasts. Cancers. 2022; 14(14): 3315.

[31]

Strell C, Paulsson J, Jin SB, et al. Impact of epithelial-stromal interactions on peritumoral fibroblasts in ductal carcinoma in situ. J Natl Cancer Inst. 2019; 111(9): 983-995.

[32]

Ma Z, Li X, Mao Y, et al. Interferon-dependent SLC14A1+ cancer-associated fibroblasts promote cancer stemness via WNT5A in bladder cancer. Cancer Cell. 2022; 40(12): 1550-1565.

[33]

Elyada E, Bolisetty M, Laise P, et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 2019; 9(8): 1102-1123.

[34]

Wang Y, Liang Y, Xu H, et al. Single-cell analysis of pancreatic ductal adenocarcinoma identifies a novel fibroblast subtype associated with poor prognosis but better immunotherapy response. Cell Discov. 2021; 7(1): 36.

[35]

Ge W, Yue M, Lin R, et al. PLA2G2A+ cancer-associated fibroblasts mediate pancreatic cancer immune escape via impeding antitumor immune response of CD8+ cytotoxic T cells. Cancer Lett. 2023; 558: 216095.

[36]

Ishii G, Ishii T. Review of cancer-associated fibroblasts and their microenvironment in post-chemotherapy recurrence. Hum Cell. 2020; 33(4): 938-945.

[37]

Clear AJ, Lee AM, Calaminici M, et al. Increased angiogenic sprouting in poor prognosis FL is associated with elevated numbers of CD163+ macrophages within the immediate sprouting microenvironment. Blood. 2010; 115(24): 5053-5056.

[38]

Im JH, Buzzelli JN, Jones K, et al. FGF2 alters macrophage polarization, tumour immunity and growth and can be targeted during radiotherapy. Nat Commun. 2020; 11(1): 4064.

[39]

Groth C, Hu X, Weber R, et al. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br J Cancer. 2019; 120(1): 16-25.

[40]

Yang Y, Li J, Lei W, et al. CXCL12-CXCR4/CXCR7 axis in cancer: from mechanisms to clinical applications. Int J Biol Sci. 2023; 19(11): 3341-3359.

[41]

Wei CY, Zhu MX, Lu NH, et al. Circular RNA circ_0020710 drives tumor progression and immune evasion by regulating the miR-370-3p/CXCL12 axis in melanoma. Mol Cancer. 2020; 19(1): 84.

[42]

Liu X, Xiao Q, Bai X, et al. Activation of STAT3 is involved in malignancy mediated by CXCL12-CXCR4 signaling in human breast cancer. Oncol Rep. 2014; 32(6): 2760-2768.

[43]

Zhang Z, Yu Y, Zhang Z, et al. Cancer-associated fibroblasts-derived CXCL12 enhances immune escape of bladder cancer through inhibiting P62-mediated autophagic degradation of PDL1. J Exp Clin Cancer Res. 2023; 42(1): 316.

[44]

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

[45]

Peng YL, Xiong LB, Zhou ZH, et al. Single-cell transcriptomics reveals a low CD8+ T cell infiltrating state mediated by fibroblasts in recurrent renal cell carcinoma. J Immunother Cancer. 2022; 10(2): e004206.

[46]

Kraxner A, Braun F, Cheng WY, et al. Investigating the complex interplay between fibroblast activation protein α-positive cancer associated fibroblasts and the tumor microenvironment in the context of cancer immunotherapy. Front Immunol. 2024; 15: 1352632.

[47]

Yang M, Wang B, Hou W, et al. NAD+ metabolism enzyme NNMT in cancer-associated fibroblasts drives tumor progression and resistance to immunotherapy by modulating macrophages in urothelial bladder cancer. J Immunother Cancer. 2024; 12(7): e009281.

[48]

Costa D, Venè R, Benelli R, et al. Targeting the epidermal growth factor receptor can counteract the inhibition of natural killer cell function exerted by colorectal tumor-associated fibroblasts. Front Immunol. 2018; 9: 1150.

[49]

Ammirante M, Shalapour S, Kang Y, Jamieson CAM, Karin M. Tissue injury and hypoxia promote malignant progression of prostate cancer by inducing CXCL13 expression in tumor myofibroblasts. Proc Natl Acad Sci USA. 2014; 111(41): 14776-14781.

[50]

Fiaschi T, Marini A, Giannoni E, et al. Reciprocal metabolic reprogramming through lactate shuttle coordinately influences tumor-stroma interplay. Cancer Res. 2012; 72(19): 5130-5140.

[51]

Liang T, Tao T, Wu K, et al. Cancer-associated fibroblast-induced remodeling of tumor microenvironment in recurrent bladder cancer. Adv Sci (Weinh). 2023; 10(31): e2303230.

[52]

Salachan PV, Rasmussen M, Ulhøi BP, Jensen JB, Borre M, Sørensen KD. Spatial whole transcriptome profiling of primary tumor from patients with metastatic prostate cancer. Int J Cancer. 2023; 153(12): 2055-2067.

[53]

Chen H, Chen G. Dissecting immunosuppressive cell communication patterns reveals JunB proto-oncogene (JUNB) shaping a non-inflamed tumor microenvironment. Front Genet. 2022; 13: 883583.

[54]

Palumbo C, Pecoraro A, Knipper S, et al. Contemporary age-adjusted incidence and mortality rates of renal cell carcinoma: analysis according to gender, race, stage, grade, and histology. Eur Urol Focus. 2021; 7(3): 644-652.

[55]

Baldewijns MM, van Vlodrop IJH, Vermeulen PB, Soetekouw PMMB, van Engeland M, de Bruïne AP. VHL and HIF signalling in renal cell carcinogenesis. J Pathol. 2010; 221(2): 125-138.

[56]

Ambrosetti D, Coutts M, Paoli C, et al. Cancer-associated fibroblasts in renal cell carcinoma: implication in prognosis and resistance to anti-angiogenic therapy. BJU Int. 2022; 129(1): 80-92.

[57]

Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol. 2017; 17(9): 559-572.

[58]

De Palma M, Biziato D, Petrova TV. Microenvironmental regulation of tumour angiogenesis. Nat Rev Cancer. 2017; 17(8): 457-474.

[59]

LaGory EL, Giaccia AJ. The ever-expanding role of HIF in tumour and stromal biology. Nat Cell Biol. 2016; 18(4): 356-365.

[60]

Chhabra Y, Weeraratna AT. Fibroblasts in cancer: unity in heterogeneity. Cell. 2023; 186(8): 1580-1609.

[61]

Jamadar A, Dwivedi N, Mathew S, Calvet JP, Thomas SM, Rao R. Vasopressin receptor type-2 mediated signaling in renal cell carcinoma stimulates stromal fibroblast activation. Int J Mol Sci. 2022; 23(14): 7601.

[62]

Chakiryan NH, Kimmel GJ, Kim Y, et al. Geospatial cellular distribution of cancer-associated fibroblasts significantly impacts clinical outcomes in metastatic clear cell renal cell carcinoma. Cancers. 2021; 13(15): 3743.

[63]

Davidson G, Helleux A, Vano YA, et al. Mesenchymal-like tumor cells and myofibroblastic cancer-associated fibroblasts are associated with progression and immunotherapy response of clear cell renal cell carcinoma. Cancer Res. 2023; 83(17): 2952-2969.

[64]

Ljungberg B, Albiges L, Abu-Ghanem Y, et al. European Association of Urology guidelines on renal cell carcinoma: the 2019 update. Eur Urol. 2019; 75(5): 799-810.

[65]

Heidegger I, Pircher A, Pichler R. Targeting the tumor microenvironment in renal cell cancer biology and therapy. Front Oncol. 2019; 9: 490.

[66]

Yang J, Butti R, Cohn S, et al. Unconventional mechanism of action and resistance to rapalogs in renal cancer. Proc Natl Acad Sci USA. 2024; 121(25): e2310793121.

[67]

Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: Cancer J Clin. 2021; 71(3): 209-249.

[68]

Chang SS, Bochner BH, Chou R, et al. Treatment of non-metastatic muscle-invasive bladder cancer: AUA/ASCO/ASTRO/SUO guideline. J Urol. 2017; 198(3): 552-559.

[69]

Lenis AT, Lec PM, Chamie K, Mshs MD. Bladder cancer: a review. JAMA. 2020; 324(19): 1980-1991.

[70]

Li Y, Zheng H, Luo Y, et al. An HGF-dependent positive feedback loop between bladder cancer cells and fibroblasts mediates lymphangiogenesis and lymphatic metastasis. Cancer Commun (Lond). 2023; 43(12): 1289-1311.

[71]

Diepenbruck M, Christofori G. Epithelial-mesenchymal transition (EMT) and metastasis: yes, no, maybe?. Curr Opin Cell Biol. 2016; 43: 7-13.

[72]

Ping Q, Wang C, Cheng X, et al. TGF-β1 dominates stromal fibroblast-mediated EMT via the FAP/VCAN axis in bladder cancer cells. J Transl Med. 2023; 21(1): 475.

[73]

Zhuang J, Lu Q, Shen B, et al. TGFβ1 secreted by cancer-associated fibroblasts induces epithelial-mesenchymal transition of bladder cancer cells through lncRNA-ZEB2NAT. Sci Rep. 2015; 5: 11924.

[74]

Schulte J, Weidig M, Balzer P, et al. Expression of the E-cadherin repressors Snail, Slug and Zeb1 in urothelial carcinoma of the urinary bladder: relation to stromal fibroblast activation and invasive behaviour of carcinoma cells. Histochem Cell Biol. 2012; 138(6): 847-860.

[75]

Zhou Z, Cui D, Sun MH, et al. CAFs-derived MFAP5 promotes bladder cancer malignant behavior through NOTCH2/HEY1 signaling. FASEB J. 2020; 34(6): 7970-7988.

[76]

Zheng H, An M, Luo Y, et al. PDGFRα+ITGA11+ fibroblasts foster early-stage cancer lymphovascular invasion and lymphatic metastasis via ITGA11-SELE interplay. Cancer Cell. 2024; 42(4): 682-700.

[77]

Yi C, Chen L, Lin Z, et al. Lenvatinib targets FGF receptor 4 to enhance antitumor immune response of anti-programmed cell death-1 in HCC. Hepatology (Baltimore, Md). 2021; 74(5): 2544-2560.

[78]

Katoh M. Fibroblast growth factor receptors as treatment targets in clinical oncology. Nat Rev Clin Oncol. 2019; 16(2): 105-122.

[79]

Adachi Y, Kamiyama H, Ichikawa K, et al. Inhibition of FGFR reactivates IFNγ signaling in tumor cells to enhance the combined antitumor activity of lenvatinib with anti-PD-1 antibodies. Cancer Res. 2022; 82(2): 292-306.

[80]

Liao CP, Adisetiyo H, Liang M, Roy-Burman P. Cancer-associated fibroblasts enhance the gland-forming capability of prostate cancer stem cells. Cancer Res. 2010; 70(18): 7294-7303.

[81]

Balar AV, Castellano D, O'Donnell PH, et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol. 2017; 18(11): 1483-1492.

[82]

Song Y, Peng Y, Qin C, et al. Fibroblast growth factor receptor 3 mutation attenuates response to immune checkpoint blockade in metastatic urothelial carcinoma by driving immunosuppressive microenvironment. J Immunother Cancer. 2023; 11(9): e006643.

[83]

Mariathasan S, Turley SJ, Nickles D, et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018; 554(7693): 544-548.

[84]

Calvete J, Larrinaga G, Errarte P, et al. The coexpression of fibroblast activation protein (FAP) and basal-type markers (CK 5/6 and CD44) predicts prognosis in high-grade invasive urothelial carcinoma of the bladder. Hum Pathol. 2019; 91: 61-68.

[85]

Ritch CR, Cookson MS. Advances in the management of castration resistant prostate cancer. BMJ (Clin Res Ed). 2016; 355: i4405.

[86]

Sun DY, Wu JQ, He ZH, He MF, Sun HB. Cancer-associated fibroblast regulate proliferation and migration of prostate cancer cells through TGF-β signaling pathway. Life Sci. 2019; 235: 116791.

[87]

Tuxhorn JA, Ayala GE, Smith MJ, Smith VC, Dang TD, Rowley DR. Reactive stroma in human prostate cancer: induction of myofibroblast phenotype and extracellular matrix remodeling. Clin Cancer Res. 2002; 8(9): 2912-2923.

[88]

Wu T, Wang W, Shi G, et al. Targeting HIC1/TGF-β axis-shaped prostate cancer microenvironment restrains its progression. Cell Death Dis. 2022; 13(7): 624.

[89]

Placencio VR, Sharif-Afshar AR, Li X, et al. Stromal transforming growth factor-beta signaling mediates prostatic response to androgen ablation by paracrine Wnt activity. Cancer Res. 2008; 68(12): 4709-4718.

[90]

Li X, Sterling JA, Fan KH, et al. Loss of TGF-β responsiveness in prostate stromal cells alters chemokine levels and facilitates the development of mixed osteoblastic/osteolytic bone lesions. Mol Cancer Res: MCR. 2012; 10(4): 494-503.

[91]

Banerjee J, Mishra R, Li X, Jackson RS, Sharma A, Bhowmick NA. A reciprocal role of prostate cancer on stromal DNA damage. Oncogene. 2014; 33(41): 4924-4931.

[92]

Meng X, Vander Ark A, Daft P, et al. Loss of TGF-β signaling in osteoblasts increases basic-FGF and promotes prostate cancer bone metastasis. Cancer Lett. 2018; 418: 109-118.

[93]

Shen T, Li Y, Zhu S, et al. YAP1 plays a key role of the conversion of normal fibroblasts into cancer-associated fibroblasts that contribute to prostate cancer progression. J Exp Clin Cancer Res: CR. 2020; 39(1): 36.

[94]

Song H, Lu T, Han D, et al. YAP1 inhibition induces phenotype switching of cancer-associated fibroblasts to tumor suppressive in prostate cancer. Cancer Res. 2024; 84(22): 3728-3742.

[95]

Zhang B, Liu M, Mai F, et al. Interruption of KLF5 acetylation promotes PTEN-deficient prostate cancer progression by reprogramming cancer-associated fibroblasts. J Clin Invest. 2024; 134(14): e175949.

[96]

Yang S, Pham LK, Liao CP, Frenkel B, Reddi AH, Roy-Burman P. A novel bone morphogenetic protein signaling in heterotypic cell interactions in prostate cancer. Cancer Res. 2008; 68(1): 198-205.

[97]

Ippolito L, Morandi A, Taddei ML, et al. Cancer-associated fibroblasts promote prostate cancer malignancy via metabolic rewiring and mitochondrial transfer. Oncogene. 2019; 38(27): 5339-5355.

[98]

Ippolito L, Comito G, Parri M, et al. Lactate rewires lipid metabolism and sustains a metabolic-epigenetic axis in prostate cancer. Cancer Res. 2022; 82(7): 1267-1282.

[99]

Neuwirt H, Bouchal J, Kharaishvili G, et al. Cancer-associated fibroblasts promote prostate tumor growth and progression through upregulation of cholesterol and steroid biosynthesis. Cell Commun Signal: CCS. 2020; 18(1): 11.

[100]

Bosso D, Pagliuca M, Sonpavde G, et al. PSA declines and survival in patients with metastatic castration-resistant prostate cancer treated with enzalutamide: a retrospective case-report study. Medicine (Baltimore). 2017; 96(24): e6817.

[101]

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

[102]

Yang L, Wang L, Lin HK, et al. Interleukin-6 differentially regulates androgen receptor transactivation via PI3K-Akt, STAT3, and MAPK, three distinct signal pathways in prostate cancer cells. Biochem Biophys Res Commun. 2003; 305(3): 462-469.

[103]

Cui D, Li J, Zhu Z, et al. Cancer-associated fibroblast-secreted glucosamine alters the androgen biosynthesis program in prostate cancer via HSD3B1 upregulation. J Clin Invest. 2023; 133(7): e161913.

[104]

Zhang Z, Karthaus WR, Lee YS, et al. Tumor microenvironment-derived NRG1 promotes antiandrogen resistance in prostate cancer. Cancer Cell. 2020; 38(2): 279-296.

[105]

Smith BN, Mishra R, Billet S, et al. Antagonizing CD105 and androgen receptor to target stromal-epithelial interactions for clinical benefit. Mol Ther. 2023; 31(1): 78-89.

[106]

Haldar S, Mishra R, Billet S, et al. Cancer epithelia-derived mitochondrial DNA is a targetable initiator of a paracrine signaling loop that confers taxane resistance. Proc Natl Acad Sci USA. 2020; 117(15): 8515-8523.

[107]

Eigentler A, Handle F, Schanung S, et al. Glucocorticoid treatment influences prostate cancer cell growth and the tumor microenvironment via altered glucocorticoid receptor signaling in prostate fibroblasts. Oncogene. 2024; 43(4): 235-247.

[108]

Xie J, Lin X, Deng X, et al. Cancer-associated fibroblast-derived extracellular vesicles: regulators and therapeutic targets in the tumor microenvironment. Cancer Drug Resist. 2025; 8: 2.

[109]

Mao Y, Liu P, Wei J, et al. Exosomes derived from umbilical cord mesenchymal stem cell promote hair regrowth in C57BL6 mice through upregulation of the RAS/ERK signaling pathway. J Transl Int Med. 2024; 12(5): 478-494.

[110]

Yuan Y, Han X, Zhao X, et al. Circulating exosome long non-coding RNAs are associated with atrial structural remodeling by increasing systemic inflammation in atrial fibrillation patients. J Transl Int Med. 2024; 12(1): 106-118.

[111]

Li Q, He G, Yu Y, Li X, Peng X, Yang L. Exosome crosstalk between cancer stem cells and tumor microenvironment: cancer progression and therapeutic strategies. Stem Cell Res Ther. 2024; 15(1): 449.

[112]

Webber J, Steadman R, Mason MD, Tabi Z, Clayton A. Cancer exosomes trigger fibroblast to myofibroblast differentiation. Cancer Res. 2010; 70(23): 9621-9630.

[113]

He T, Zhang Q, Xu P, et al. Extracellular vesicle-circEHD2 promotes the progression of renal cell carcinoma by activating cancer-associated fibroblasts. Mol Cancer. 2023; 22(1): 117.

[114]

Ding M, Zhao X, Chen X, et al. Cancer-associated fibroblasts promote the stemness and progression of renal cell carcinoma via exosomal miR-181d-5p. Cell Death Discovery. 2022; 8(1): 439.

[115]

Zhang Y, Luo G, You S, Zhang L, Liang C, Chen X. Exosomal LINC00355 derived from cancer-associated fibroblasts promotes bladder cancer cell proliferation and invasion by regulating miR-15a-5p/HMGA2 axis. Acta Biochim Biophys Sin (Shanghai). 2021; 53(6): 673-682.

[116]

Luo G, Zhang Y, Wu Z, Zhang L, Liang C, Chen X. Exosomal LINC00355 derived from cancer-associated fibroblasts promotes bladder cancer cell resistance to cisplatin by regulating miR-34b-5p/ABCB1 axis. Acta Biochim Biophys Sin (Shanghai). 2021; 53(5): 558-566.

[117]

Long X, Xiong W, Zeng X, et al. Cancer-associated fibroblasts promote cisplatin resistance in bladder cancer cells by increasing IGF-1/ERβ/Bcl-2 signalling. Cell Death Dis. 2019; 10(5): 375.

[118]

Zhuang J, Shen L, Li M, et al. Cancer-associated fibroblast-derived miR-146a-5p generates a Niche that promotes bladder cancer stemness and chemoresistance. Cancer Res. 2023; 83(10): 1611-1627.

[119]

Zhao J, Shen J, Mao L, Yang T, Liu J, Hongbin S. Cancer associated fibroblast secreted miR-432-5p targets CHAC1 to inhibit ferroptosis and promote acquired chemoresistance in prostate cancer. Oncogene. 2024; 43(27): 2104-2114.

[120]

Shan G, Gu J, Zhou D, et al. Cancer-associated fibroblast-secreted exosomal miR-423-5p promotes chemotherapy resistance in prostate cancer by targeting GREM2 through the TGF-β signaling pathway. Exp Mol Med. 2020; 52(11): 1809-1822.

[121]

Zhang Y, Zhao J, Ding M, et al. Loss of exosomal miR-146a-5p from cancer-associated fibroblasts after androgen deprivation therapy contributes to prostate cancer metastasis. J Exp Clin Cancer Res: CR. 2020; 39(1): 282.

[122]

Ma WK, Voss DM, Scharner J, et al. ASO-based PKM splice-switching therapy inhibits hepatocellular carcinoma growth. Cancer Res. 2022; 82(5): 900-915.

[123]

Huang M, Yang J, Wang T, et al. Homogeneous, low-volume, efficient, and sensitive quantitation of circulating exosomal PD-L1 for cancer diagnosis and immunotherapy response prediction. Angew Chem (Int Ed Engl). 2020; 59(12): 4800-4805.

[124]

Pascucci L, Coccè V, Bonomi A, et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: a new approach for drug delivery. J Control Release. 2014; 192: 262-270.

[125]

Tian Y, Li S, Song J, et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials. 2014; 35(7): 2383-2390.

[126]

Zhang M, Luo C, Cui K, Xiong T, Chen Z. Chronic inflammation promotes proliferation in the prostatic stroma in rats with experimental autoimmune prostatitis: study for a novel method of inducing benign prostatic hyperplasia in a rat model. World J Urol. 2020; 38(11): 2933-2943.

[127]

Livingston MJ, Shu S, Fan Y, et al. Tubular cells produce FGF2 via autophagy after acute kidney injury leading to fibroblast activation and renal fibrosis. Autophagy. 2023; 19(1): 256-277.

[128]

Li L, Lu M, Peng Y, et al. Oxidatively stressed extracellular microenvironment drives fibroblast activation and kidney fibrosis. Redox Biol. 2023; 67: 102868.

[129]

Nguyen EV, Pereira BA, Lawrence MG, et al. Proteomic profiling of human prostate cancer-associated fibroblasts (CAF) reveals LOXL2-dependent regulation of the tumor microenvironment. Mol Cell Proteomics: MCP. 2019; 18(7): 1410-1427.

[130]

Nagaraja AS, Dood RL, Armaiz-Pena G, et al. Adrenergic-mediated increases in INHBA drive CAF phenotype and collagens. JCI Insight. 2017; 2(16): e93076.

[131]

Lee YC, Kurtova AV, Xiao J, et al. Collagen-rich airway smooth muscle cells are a metastatic niche for tumor colonization in the lung. Nat Commun. 2019; 10(1): 2131.

[132]

Wan F, Wang H, Shen Y, et al. Upregulation of COL6A1 is predictive of poor prognosis in clear cell renal cell carcinoma patients. Oncotarget. 2015; 6(29): 27378-27387.

[133]

Chong IW, Chang MY, Chang HC, et al. Great potential of a panel of multiple hMTH1, SPD, ITGA11 and COL11A1 markers for diagnosis of patients with non-small cell lung cancer. Oncol Rep. 2006; 16(5): 981-988.

[134]

Ellsworth RE, Seebach J, Field LA, et al. A gene expression signature that defines breast cancer metastases. Clin Exp Metastasis. 2009; 26(3): 205-213.

[135]

Ewald JA, Downs TM, Cetnar JP, Ricke WA. Expression microarray meta-analysis identifies genes associated with Ras/MAPK and related pathways in progression of muscle-invasive bladder transition cell carcinoma. PLoS One. 2013; 8(2): e55414.

[136]

Adams LC, Ralla B, Jurmeister P, et al. Native T1 mapping as an in vivo biomarker for the identification of higher-grade renal cell carcinoma: correlation with histopathological findings. Invest Radiol. 2019; 54(2): 118-128.

[137]

Kesch C, Yirga L, Dendl K, et al. High fibroblast-activation-protein expression in castration-resistant prostate cancer supports the use of FAPI-molecular theranostics. Eur J Nucl Med Mol Imaging. 2021; 49(1): 385-389.

[138]

Penet MF, Kakkad S, Pathak AP, et al. Structure and function of a prostate cancer dissemination-permissive extracellular matrix. Clin Cancer Res. 2017; 23(9): 2245-2254.

[139]

Kakkad SM, Solaiyappan M, O'Rourke B, et al. Hypoxic tumor microenvironments reduce collagen I fiber density. Neoplasia. 2010; 12(8): 608-617.

[140]

Lin D, Luo C, Wei P, et al. YAP1 recognizes inflammatory and mechanical cues to exacerbate benign prostatic hyperplasia via promoting cell survival and fibrosis. Adv Sci (Weinh). 2024; 11(5): e2304274.

[141]

Ou YC, Li JR, Wang JD, et al. Fibronectin promotes cell growth and migration in human renal cell carcinoma cells. Int J Mol Sci. 2019; 20(11): 2792.

[142]

Eiro N, Cid S, Fraile M, Cabrera JR, Gonzalez LO, Vizoso FJ. Analysis of the gene expression profile of stromal pro-tumor factors in cancer-associated fibroblasts from luminal breast carcinomas. Diagnostics (Basel, Switzerland). 2020; 10(11): 865.

[143]

Calvo F, Ege N, Grande-Garcia A, et al. Mechanotransduction and YAP-dependent matrix remodelling is required for the generation and maintenance of cancer-associated fibroblasts. Nat Cell Biol. 2013; 15(6): 637-646.

[144]

Olivares O, Mayers JR, Gouirand V, et al. Collagen-derived proline promotes pancreatic ductal adenocarcinoma cell survival under nutrient limited conditions. Nat Commun. 2017; 8: 16031.

[145]

Chen Y, Kim J, Yang S, et al. Type I collagen deletion in αSMA+ myofibroblasts augments immune suppression and accelerates progression of pancreatic cancer. Cancer Cell. 2021; 39(4): 548-565.

[146]

Erdogan B, Ao M, White LM, et al. Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin. J Cell Biol. 2017; 216(11): 3799-3816.

[147]

Levental KR, Yu H, Kass L, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009; 139(5): 891-906.

[148]

Heneberg P. Paracrine tumor signaling induces transdifferentiation of surrounding fibroblasts. Crit Rev Oncol Hematol. 2016; 97: 303-311.

[149]

Giannoni E, Bianchini F, Masieri L, et al. Reciprocal activation of prostate cancer cells and cancer-associated fibroblasts stimulates epithelial-mesenchymal transition and cancer stemness. Cancer Res. 2010; 70(17): 6945-6956.

[150]

Eiro N, Medina A, Gonzalez LO, et al. Evaluation of matrix metalloproteases by artificial intelligence techniques in negative biopsies as new diagnostic strategy in prostate cancer. Int J Mol Sci. 2023; 24(8): 7022.

[151]

Jia D, Zhou Z, Kwon OJ, et al. Stromal FOXF2 suppresses prostate cancer progression and metastasis by enhancing antitumor immunity. Nat Commun. 2022; 13(1): 6828.

[152]

Ippolito L, Duatti A, Iozzo M, et al. Lactate supports cell-autonomous ECM production to sustain metastatic behavior in prostate cancer. EMBO Rep. 2024; 25(8): 3506-3531.

[153]

Chen JL, Walton KL, Qian H, et al. Differential effects of IL6 and activin A in the development of cancer-associated cachexia. Cancer Res. 2016; 76(18): 5372-5382.

[154]

Togashi Y, Kogita A, Sakamoto H, et al. Activin signal promotes cancer progression and is involved in cachexia in a subset of pancreatic cancer. Cancer Lett. 2015; 356(Pt B): 819-827.

[155]

Boinapally S, Lisok A, Lofland G, et al. Hetero-bivalent agents targeting FAP and PSMA. Eur J Nucl Med Mol Imaging. 2022; 49(13): 4369-4381.

[156]

Mona CE, Benz MR, Hikmat F, et al. Correlation of 68Ga-FAPi-46 PET biodistribution with FAP expression by immunohistochemistry in patients with solid cancers: interim analysis of a prospective translational exploratory study. J Nucl Med. 2022; 63(7): 1021-1026.

[157]

Chen Y, McAndrews KM, Kalluri R. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat Rev Clin Oncol. 2021; 18(12): 792-804.

[158]

Menezes S, Okail MH, Jalil SMA, Kocher HM, Cameron AJM. Cancer-associated fibroblasts in pancreatic cancer: new subtypes, new markers, new targets. J Pathol. 2022; 257(4): 526-544.

[159]

Miao L, Liu Q, Lin CM, et al. Targeting tumor-associated fibroblasts for therapeutic delivery in desmoplastic tumors. Cancer Res. 2017; 77(3): 719-731.

[160]

Lang J, Zhao X, Qi Y, et al. Reshaping prostate tumor microenvironment to suppress metastasis via cancer-associated fibroblast inactivation with peptide-assembly-based nanosystem. ACS Nano. 2019; 13(11): 12357-12371.

[161]

Owaki T, Iida T, Miyai Y, et al. Synthetic retinoid-mediated preconditioning of cancer-associated fibroblasts and macrophages improves cancer response to immune checkpoint blockade. Br J Cancer. 2024; 131(2): 372-386.

[162]

Pietrobono S, Sabbadini F, Bertolini M, et al. Autotaxin secretion is a stromal mechanism of adaptive resistance to TGFβ inhibition in pancreatic ductal adenocarcinoma. Cancer Res. 2024; 84(1): 118-132.

[163]

Grünewald S, Politz O, Bender S, et al. Rogaratinib: a potent and selective pan-FGFR inhibitor with broad antitumor activity in FGFR-overexpressing preclinical cancer models. Int J Cancer. 2019; 145(5): 1346-1357.

[164]

Sternberg CN, Petrylak DP, Bellmunt J, et al. FORT-1: phase II/III study of rogaratinib versus chemotherapy in patients with locally advanced or metastatic urothelial carcinoma selected based on FGFR1/3 mRNA expression. J Clin Oncol. 2023; 41(3): 629-639.

[165]

Mei J, Ma J, Xu Y, et al. Cinnamaldehyde treatment of prostate cancer-associated fibroblasts prevents their inhibitory effect on T cells through toll-like receptor 4. Drug Des Dev Ther. 2020; 14: 3363-3372.

[166]

Youn HS, Lee JK, Choi YJ, et al. Cinnamaldehyde suppresses toll-like receptor 4 activation mediated through the inhibition of receptor oligomerization. Biochem Pharmacol. 2008; 75(2): 494-502.

[167]

Liu X, Tang J, Peng L, Nie H, Zhang Y, Liu P. Cancer-associated fibroblasts promote malignant phenotypes of prostate cancer cells via autophagy: cancer-associated fibroblasts promote prostate cancer development. Apoptosis. 2023; 28(5-6): 881-891.

[168]

Doldi V, Tortoreto M, Colecchia M, et al. Repositioning of antiarrhythmics for prostate cancer treatment: a novel strategy to reprogram cancer-associated fibroblasts towards a tumor-suppressive phenotype. J Exp Clin Cancer Res: CR. 2024; 43(1): 161.

[169]

Mezheyeuski A, Segersten U, Leiss LW, et al. Fibroblasts in urothelial bladder cancer define stroma phenotypes that are associated with clinical outcome. Sci Rep. 2020; 10(1): 281.

[170]

Ma LJ, Wang XY, Duan M, et al. Telomere length variation in tumor cells and cancer-associated fibroblasts: potential biomarker for hepatocellular carcinoma. J Pathol. 2017; 243(4): 407-417.

[171]

Heaphy CM, Yoon GS, Peskoe SB, et al. Prostate cancer cell telomere length variability and stromal cell telomere length as prognostic markers for metastasis and death. Cancer Discov. 2013; 3(10): 1130-1141.

[172]

Matsuda Y, Ye J, Yamakawa K, et al. Association of longer telomere length in cancer cells and cancer-associated fibroblasts with worse prognosis. J Natl Cancer Inst. 2023; 115(2): 208-218.

[173]

Imran SAM, Yazid MD, Idrus RBH, et al. Is there an interconnection between epithelial-mesenchymal transition (EMT) and telomere shortening in aging?. Int J Mol Sci. 2021; 22(8): 3888.

[174]

Zhang R, Liu F. Cancer-associated fibroblast-derived gene signatures predict radiotherapeutic survival in prostate cancer patients. J Transl Med. 2022; 20(1): 453.

[175]

Vickman RE, Broman MM, Lanman NA, et al. Heterogeneity of human prostate carcinoma-associated fibroblasts implicates a role for subpopulations in myeloid cell recruitment. Prostate. 2020; 80(2): 173-185.

[176]

Xie J, Yang A, Liu Q, et al. Single-cell RNA sequencing elucidated the landscape of breast cancer brain metastases and identified ILF2 as a potential therapeutic target. Cell Prolif. 2024; 57(11): e13697.

[177]

Zheng S, Liang JY, Tang Y, et al. Dissecting the role of cancer-associated fibroblast-derived biglycan as a potential therapeutic target in immunotherapy resistance: a tumor bulk and single-cell transcriptomic study. Clin Transl Med. 2023; 13(2): e1189.

[178]

Liu L, Xie Y, Yang H, et al. HPVTIMER: a shiny web application for tumor immune estimation in human papillomavirus-associated cancers. Imeta. 2023; 2(3): e130.

[179]

Xu K, Huang Y, Wu M, Yin J, Wei P. 3D bioprinting of multi-cellular tumor microenvironment for prostate cancer metastasis. Biofabrication. 2023; 15(3).

[180]

López JI, Errarte P, Erramuzpe A, et al. Fibroblast activation protein predicts prognosis in clear cell renal cell carcinoma. Hum Pathol. 2016; 54: 100-105.

[181]

Errarte P, Guarch R, Pulido R, et al. The expression of fibroblast activation protein in clear cell renal cell carcinomas is associated with synchronous lymph node metastases. PLoS One. 2016; 11(12): e0169105.

[182]

Abd El-Azeem MA, Ali MA, El-Shorbagy SH. Expression of GLUT4 and FAP in urothelial bladder carcinoma: correlation with angiogenesis and clinicopathological characteristics. J Egypt Natl Canc Inst. 2022; 34(1): 40.

[183]

Brennen WN, Rosen DM, Wang H, Isaacs JT, Denmeade SR. Targeting carcinoma-associated fibroblasts within the tumor stroma with a fibroblast activation protein-activated prodrug. J Natl Cancer Inst. 2012; 104(17): 1320-1334.

[184]

Poplawski SE, Lai JH, Li Y, et al. Identification of selective and potent inhibitors of fibroblast activation protein and prolyl oligopeptidase. J Med Chem. 2013; 56(9): 3467-3477.

[185]

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

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

7

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/