The Role of Cancer-Associated Fibroblasts in Tumor Progression
Abstract
:Simple Summary
Abstract
1. Introduction
2. Cancer-Associated Fibroblast (CAF) Markers
3. The Origin of CAFs
4. CAF Promotion of Tumor Growth and Maintenance of Stemness
4.1. Lung Cancer
4.2. Breast Cancer
4.3. Gastrointestinal (GI) Cancer
4.4. Skin Cancer
4.5. Ovarian Cancer
4.6. Endometrial Cancer
4.7. Prostate Cancer
4.8. Renal Cell Carcinoma
4.9. Bladder Cancer
5. Pro-Angiogenic Effects of CAFs
5.1. Breast Cancer
5.2. Head and Neck Cancer
5.3. GI Cancer
5.4. Skin Cancer
5.5. Renal Cell Carcinoma
6. CAF Stimulation of Invasion and Metastasis
6.1. GI Cancer
6.2. Lung Cancer
6.3. Endocrine/Neuroendocrine Cancers
6.4. Breast Cancer
6.5. Prostate Cancer
6.6. Renal Cell Carcinoma
6.7. Head and Neck Cancer
6.8. Bladder Cancer
7. Effects of CAFs on Immune Cells in the Tumor Microenvironment
7.1. Skin Cancer
7.2. Head and Neck Cancer
7.3. Lung Cancer
7.4. GI Cancer
8. CAF Regulation of Cancer Cell Metabolism
9. CAF Effects on Therapeutic Resistance
10. CAF Subtypes
11. Utility of CAFs as Prognostic Markers
12. Potential Therapeutic Targets for CAFs
12.1. Lung Cancer
12.2. GI Cancer
12.3. Breast Cancer
12.4. Skin Cancer
12.5. Ovarian Cancer
12.6. Endometrial Cancer
12.7. Prostate Cancer
12.8. Bladder Cancer
13. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Kobayashi, H.; Enomoto, A.; Woods, S.L.; Burt, A.D.; Takahashi, M.; Worthley, D.L. Cancer-associated fibroblasts in gastrointestinal cancer. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 282–295. [Google Scholar] [CrossRef]
- Ansems, M.; Span, P.N. The tumor microenvironment and radiotherapy response; a central role for cancer-associated fibroblasts. Clin. Transl. Radiat. Oncol. 2020, 22, 90–97. [Google Scholar] [CrossRef] [PubMed]
- Guo, Y.; Zhang, X.; Zeng, W.; Zhang, J.; Cai, L.; Wu, Z.; Su, J.; Xiao, Y.; Liu, N.; Tang, L.; et al. TRAF6 Activates Fibroblasts to Cancer-Associated Fibroblasts through FGF19 in Tumor Microenvironment to Benefit the Malignant Phenotype of Melanoma Cells. J. Investig. Dermatol. 2020, 140, 2268–2279.e11. [Google Scholar] [CrossRef] [PubMed]
- Xiang, H.; Ramil, C.P.; Hai, J.; Zhang, C.; Wang, H.; Watkins, A.A.; Afshar, R.; Georgiev, P.; Sze, M.A.; Song, X.S.; et al. Cancer-Associated Fibroblasts Promote Immunosuppression by Inducing ROS-Generating Monocytic MDSCs in Lung Squamous Cell Carcinoma. Cancer Immunol. Res. 2020, 8, 436–450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Q.; Yang, Y.; Jiang, X.; Jin, Y.; Wu, J.; Qin, Y.; Qi, X.; Cheng, Y.; Mao, Y.; Hua, D. The combined expressions of B7H4 and ACOT4 in cancer-associated fibroblasts are related to poor prognosis in patients with gastric carcinoma. Int. J. Clin. Exp. Pathol. 2019, 12, 2672–2681. [Google Scholar]
- Zhao, X.; Ding, L.; Lu, Z.; Huang, X.; Jing, Y.; Yang, Y.; Chen, S.; Hu, Q.; Ni, Y. Diminished CD68+ Cancer-Associated Fibroblast Subset Induces Regulatory T-Cell (Treg) Infiltration and Predicts Poor Prognosis of Oral Squamous Cell Carcinoma Patients. Am. J. Pathol. 2020, 190, 886–899. [Google Scholar] [CrossRef]
- Ramos-Vega, V.; Rojas, B.V.; Torres, W.D. Immunohistochemical analysis of cancer-associated fibroblasts and podoplanin in head and neck cancer. Med. Oral Patol. Oral Cir. Bucal 2020, 25, e268–e276. [Google Scholar] [CrossRef]
- Raudenská, M.; Svobodová, M.; Gumulec, J.; Falk, M.; Masařík, M. The importance of cancer-associated fibroblasts in the pathogenesis of head and neck cancers. Klin. Onkol. 2020, 33, 39–48. [Google Scholar] [CrossRef]
- Awaji, M.; Singh, R.K. Cancer-associated fibroblasts’ functional heterogeneity in pancreatic ductal adenocarcinoma. Cancers 2019, 11, 290. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Y.; Shi, C.; Zeng, L.; Liu, G.; Jiang, W.; Zhang, X.; Chen, S.; Guo, J.; Jian, X.; Ouyang, J.; et al. High COX-2 expression in cancer-associated fibiroblasts contributes to poor survival and promotes migration and invasiveness in nasopharyngeal carcinoma. Mol. Carcinog. 2020, 59, 265–280. [Google Scholar] [CrossRef] [Green Version]
- Johansson, A.C.; Ansell, A.; Jerhammar, F.; Lindh, M.B.; Grénman, R.; Munck-Wikland, E.; Östman, A.; Roberg, K. Cancer-associated fibroblasts induce matrix metalloproteinase-mediated cetuximab resistance in head and neck squamous cell carcinoma cells. Mol. Cancer Res. 2012, 10, 1158–1168. [Google Scholar] [CrossRef] [Green Version]
- Mishra, P.J.; Mishra, P.J.; Humeniuk, R.; Medina, D.J.; Alexe, G.; Mesirov, J.P.; Ganesan, S.; Glod, J.W.; Banerjee, D. Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells. Cancer Res. 2008, 68, 4331–4339. [Google Scholar] [CrossRef] [Green Version]
- Farmer, P.; Bonnefoi, H.; Anderle, P.; Cameron, D.; Wirapati, P.; Becette, V.; André, S.; Piccart, M.; Campone, M.; Brain, E.; et al. A stroma-related gene signature predicts resistance to neoadjuvant chemotherapy in breast cancer. Nat. Med. 2009, 15, 68–74. [Google Scholar] [CrossRef]
- Wu, H.J.; Hao, M.; Yeo, S.K.; Guan, J.L. FAK signaling in cancer-associated fibroblasts promotes breast cancer cell migration and metastasis by exosomal miRNAs-mediated intercellular communication. Oncogene 2020, 39, 2539–2549. [Google Scholar] [CrossRef]
- Li, F.; Zhao, S.; Cui, Y.; Guo, T.; Qiang, J.; Xie, Q.; Yu, W.; Guo, W. cancer-promoting capacity of cancer-associated fibroblasts (CAFs) by modifying EGFR core fucosylation (CF) in non-small cell lung cancer (NSCLC). Am. J. Cancer Res. 2020, 10, 816–837. [Google Scholar] [PubMed]
- Santos, A.M.; Jung, J.; Aziz, N.; Kissil, J.L.; Puré, E. Targeting fibroblast activation protein inhibits tumor stromagenesis and growth in mice. J. Clin. Investig. 2009, 119, 3613–3625. [Google Scholar] [CrossRef] [PubMed]
- Direkze, N.C.; Hodivala-Dilke, K.; Jeffery, R.; Hunt, T.; Poulsom, R.; Oukrif, D.; Alison, M.R.; Wright, N.A. Bone marrow contribution to tumor-associated myofibroblasts and fibroblasts. Cancer Res. 2004, 64, 8492–8495. [Google Scholar] [CrossRef] [Green Version]
- Huang, C.; Xu, J.; Li, Z. Research progress of cancer-associated fibroblasts in lung cancer. Chin. J. Lung Cancer 2020, 23, 267–273. [Google Scholar] [CrossRef]
- Samain, R.; Sanz-Moreno, V. Cancer-associated fibroblasts: Activin A adds another string to their bow. EMBO Mol. Med. 2020, 12. [Google Scholar] [CrossRef]
- Erez, N.; Truitt, M.; Olson, P.; Hanahan, D. Cancer-Associated Fibroblasts Are Activated in Incipient Neoplasia to Orchestrate Tumor-Promoting Inflammation in an NF-κB-Dependent Manner. Cancer Cell 2010, 17, 135–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vosseler, S.; Lederle, W.; Airola, K.; Obermueller, E.; Fusenig, N.E.; Mueller, M.M. Distinct progression-associated expression of tumor and stromal MMPs in HaCaT skin SCCs correlates with onset of invasion. Int. J. Cancer 2009, 125, 2296–2306. [Google Scholar] [CrossRef]
- Vaquero, J.; Aoudjehane, L.; Fouassier, L. Cancer-associated fibroblasts in cholangiocarcinoma. Curr. Opin. Gastroenterol. 2020, 36, 63–69. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Deng, T.; Liu, R.; Ning, T.; Yang, H.; Liu, D.; Zhang, Q.; Lin, D.; Ge, S.; Bai, M.; et al. CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Mol. Cancer 2020, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bai, Y.P.; Shang, K.; Chen, H.; Ding, F.; Wang, Z.; Liang, C.; Xu, Y.; Sun, M.H.; Li, Y.Y. FGF-1/-3/FGFR4 signaling in cancer-associated fibroblasts promotes tumor progression in colon cancer through Erk and MMP-7. Cancer Sci. 2015, 106, 1278–1287. [Google Scholar] [CrossRef] [PubMed]
- Colvin, E.K.; Howell, V.M.; Mok, S.C.; Samimi, G.; Vafaee, F. Expression of long noncoding RNAs in cancer-associated fibroblasts linked to patient survival in ovarian cancer. Cancer Sci. 2020, 111, 1805–1817. [Google Scholar] [CrossRef]
- Teng, F.; Tian, W.Y.; Wang, Y.M.; Zhang, Y.F.; Guo, F.; Zhao, J.; Gao, C.; Xue, F.X. Cancer-associated fibroblasts promote the progression of endometrial cancer via the SDF-1/CXCR4 axis. J. Hematol. Oncol. 2016, 9. [Google Scholar] [CrossRef] [Green Version]
- Rick, J.; Nguyen, A.; Chandra, A.; Wadhwa, H.; Shah, S.; Wang, L.; Lau, D.; Safaee, M.; Ordaz, A.; Yagnik, G.; et al. TMIC-22. IDENTIFICATION OF CANCER-ASSOCIATED FIBROBLASTS IN GLIOBLASTOMA and Defining Their Protumoral Effects. Neuro-Oncology 2019, 21 (Suppl. 6), vi252. [Google Scholar] [CrossRef]
- Serres, E.; Debarbieux, F.; Stanchi, F.; Maggiorella, L.; Grall, D.; Turchi, L.; Burel-Vandenbos, F.; Figarella-Branger, D.; Virolle, T.; Rougon, G.; et al. Fibronectin expression in glioblastomas promotes cell cohesion, collective invasion of basement membrane in vitro and orthotopic tumor growth in mice. Br. Dent. J. 2014, 217, 3451–3462. [Google Scholar] [CrossRef] [PubMed]
- Trylcova, J.; Busek, P.; Smetana, K.; Balaziova, E.; Dvorankova, B.; Mifkova, A.; Sedo, A. Effect of cancer-associated fibroblasts on the migration of glioma cells in vitro. Tumor Biol. 2015, 36, 5873–5879. [Google Scholar] [CrossRef]
- Louault, K.; Li, R.R.; Declerck, Y.A. Cancer-associated fibroblasts: Understanding their heterogeneity. Cancers 2020, 12, 3108. [Google Scholar] [CrossRef]
- Nurmik, M.; Ullmann, P.; Rodriguez, F.; Haan, S.; Letellier, E. In search of definitions: Cancer-associated fibroblasts and their markers. Int. J Cancer 2020, 146, 895–905. [Google Scholar] [CrossRef] [Green Version]
- Ortiz-Otero, N.; Clinch, A.B.; Hope, J.; Wang, W.; Reinhart-King, C.A.; King, M.R. Cancer associated fibroblasts confer shear resistance to circulating tumor cells during prostate cancer metastatic progression. Oncotarget 2020, 11, 1037–1050. [Google Scholar] [CrossRef] [Green Version]
- Liu, Q.; Yu, B.; Tian, Y.; Dan, J.; Luo, Y.; Wu, X. P53 mutant P53N236S regulates cancer-associated fibroblasts properties through stat3 pathway. OncoTargets Ther. 2020, 13, 1355–1363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shen, T.; Li, Y.; Zhu, S.; Yu, J.; Zhang, B.; Chen, X.; Zhang, Z.; Ma, Y.; Niu, Y.; Shang, Z. 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. 2020, 39. [Google Scholar] [CrossRef] [PubMed]
- Jung, Y.Y.; Lee, Y.K.; Koo, J.S. Expression of cancer-associated fibroblast-related proteins in adipose stroma of breast cancer. Tumor Biol. 2015, 36, 8685–8695. [Google Scholar] [CrossRef] [PubMed]
- Aboussekhra, A. Role of cancer-associated fibroblasts in breast cancer development and prognosis. Int. J. Dev. Biol. 2011, 55, 841–849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pelon, F.; Bourachot, B.; Kieffer, Y.; Magagna, I.; Mermet-Meillon, F.; Bonnet, I.; Costa, A.; Givel, A.M.; Attieh, Y.; Barbazan, J.; et al. Cancer-associated fibroblast heterogeneity in axillary lymph nodes drives metastases in breast cancer through complementary mechanisms. Nat. Commun. 2020, 11. [Google Scholar] [CrossRef] [Green Version]
- Yang, S.S.; Ma, S.; Dou, H.; Liu, F.; Zhang, S.Y.; Jiang, C.; Xiao, M.; Huang, Y.X. Breast cancer-derived exosomes regulate cell invasion and metastasis in breast cancer via miR-146a to activate cancer associated fibroblasts in tumor microenvironment. Exp. Cell Res. 2020, 391. [Google Scholar] [CrossRef] [PubMed]
- Fan, J.; Xu, G.; Chang, Z.; Zhu, L.; Yao, J. MiR-210 transferred by lung cancer cell-derived exosomes may act as proangiogenic factor in cancer-associated fibroblasts by modulating JAK2/STAT3 pathway. Clin. Sci. 2020, 134, 807–825. [Google Scholar] [CrossRef]
- Neri, S.; Ishii, G.; Hashimoto, H.; Kuwata, T.; Nagai, K.; Date, H.; Ochiai, A. Podoplanin-expressing cancer-associated fibroblasts lead and enhance the local invasion of cancer cells in lung adenocarcinoma. Int. J. Cancer 2015, 137, 784–796. [Google Scholar] [CrossRef]
- Nie, S.; Wang, X.; Wang, H. NLRP3 inflammasome mediated interleukin-1β production in cancer-associated fibroblast contributes to ALA-PDT for cutaneous squamous cell carcinoma. Cancer Manag. Res. 2019, 11, 10257–10267. [Google Scholar] [CrossRef] [Green Version]
- Érsek, B.; Silló, P.; Cakir, U.; Molnár, V.; Bencsik, A.; Mayer, B.; Mezey, E.; Kárpáti, S.; Pós, Z.; Németh, K. Melanoma-associated fibroblasts impair CD8+ T cell function and modify expression of immune checkpoint regulators via increased arginase activity. Cell Mol. Life Sci. 2020. [Google Scholar] [CrossRef] [Green Version]
- Mezheyeuski, A.; Segersten, U.; Leiss, L.W.; Malmström, P.U.; Hatina, J.; Östman, A.; Strell, C. Fibroblasts in urothelial bladder cancer define stroma phenotypes that are associated with clinical outcome. Sci. Rep. 2020, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhuang, J.; Lu, Q.; Shen, B.; Huang, X.; Shen, L.; Zheng, X.; Huang, R.; Yan, J.; Guo, H. TGFβ1 secreted by cancer-associated fibroblasts induces epithelial-mesenchymal transition of bladder cancer cells through lncRNA-ZEB2NAT. Sci. Rep. 2015, 5. [Google Scholar] [CrossRef] [Green Version]
- Errarte, P.; Larrinaga, G.; López, J.I. The role of cancer-associated fibroblasts in renal cell carcinoma. An example of tumor modulation through tumor/non-tumor cell interactions. J. Adv. Res. 2020, 21, 103–108. [Google Scholar] [CrossRef]
- Zagzag, D.; Krishnamachary, B.; Yee, H.; Okuyama, H.; Chiriboga, L.; Ali, M.A.; Melamed, J.; Semenza, G.L. Stromal cell-derived factor-1α and CXCR4 expression in hemangioblastoma and clear cell-renal cell carcinoma: Von Hippel-Lindau loss-of-function induces expression of a ligand and its receptor. Cancer Res. 2005, 65, 6178–6188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, Y.; Fan, X.; Zhang, Q.; Shi, X.; Xu, G.; Zou, C. Cancer-associated fibroblasts secrete FGF-1 to promote ovarian proliferation, migration, and invasion through the activation of FGF-1/FGFR4 signaling. Tumor Biol. 2017, 39, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Guo, H.; Ha, C.; Dong, H.; Yang, Z.; Ma, Y.; Ding, Y. Cancer-associated fibroblast-derived exosomal microRNA-98-5p promotes cisplatin resistance in ovarian cancer by targeting CDKN1A. Cancer Cell Int. 2019, 19. [Google Scholar] [CrossRef] [PubMed]
- Ruiz-Herrera, A.; Smirnova, A.; Khoriauli, L.; Nergadze, S.G.; Mondello, C.; Giulotto, E. Gene amplification in human cells knocked down for RAD54. Genome Integr. 2011. [Google Scholar] [CrossRef] [Green Version]
- Zhao, G.; Li, H.; Guo, Q.; Zhou, A.; Wang, X.; Li, P.; Zhang, S. Exosomal Sonic Hedgehog derived from cancer-associated fibroblasts promotes proliferation and migration of esophageal squamous cell carcinoma. Cancer Med. 2020, 9, 2500–2513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, L.; Wang, Z.; Geng, X.; Zhang, Y.; Xue, Z. Exosomal miRNA-34 from cancer-associated fibroblasts inhibits growth and invasion of gastric cancer cells in vitro and in vivo. Aging 2020, 12, 8549–8564. [Google Scholar] [CrossRef]
- Ham, I.H.; Lee, D.; Hur, H. Role of cancer-associated fibroblast in gastric cancer progression and resistance to treatments. J. Oncol. 2019, 2019. [Google Scholar] [CrossRef]
- Wei, L.; Ye, H.; Li, G.; Lu, Y.; Zhou, Q.; Zheng, S.; Lin, Q.; Liu, Y.; Li, Z.; Chen, R. Cancer-associated fibroblasts promote progression and gemcitabine resistance via the SDF-1/SATB-1 pathway in pancreatic cancer. Cell Death Dis. 2018, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chuaysri, C.; Thuwajit, P.; Paupairoj, A.; Chau-In, S.; Suthiphongchai, T.; Thuwajit, C. Alpha-smooth muscle actin-positive fibroblasts promote biliary cell proliferation and correlate with poor survival in cholangiocarcinoma. Oncol. Rep. 2009, 21, 957–969. [Google Scholar] [CrossRef] [Green Version]
- Zou, B.; Liu, X.; Gong, Y.; Cai, C.; Li, P.; Xing, S.; Pokhrel, B.; Zhang, B.; Li, J. A novel 12-marker panel of cancer-associated fibroblasts involved in progression of hepatocellular carcinoma. Cancer Manag. Res. 2018, 10, 5303–5311. [Google Scholar] [CrossRef] [Green Version]
- Lau, E.Y.T.; Lo, J.; Cheng, B.Y.L.; Ma, M.K.F.; Lee, J.M.F.; Ng, J.K.Y.; Chai, S.; Lin, C.H.; Tsang, S.Y.; Ma, S.; et al. Cancer-Associated Fibroblasts Regulate Tumor-Initiating Cell Plasticity in Hepatocellular Carcinoma through c-Met/FRA1/HEY1 Signaling. Cell Rep. 2016, 15, 1175–1189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sha, M.; Jeong, S.; Qiu, B.J.; Tong, Y.; Xia, L.; Xu, N.; Zhang, J.J.; Xia, Q. Isolation of cancer-associated fibroblasts and its promotion to the progression of intrahepatic cholangiocarcinoma. Cancer Med. 2018, 7, 4665–4677. [Google Scholar] [CrossRef] [PubMed]
- Affo, S.; Yu, L.X.; Schwabe, R.F. The Role of Cancer-Associated Fibroblasts and Fibrosisin Liver Cancer. Annu. Rev. Pathol. Mech. Dis. 2017, 12, 153–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bello, I.O.; Vered, M.; Dayan, D.; Dobriyan, A.; Yahalom, R.; Alanen, K.; Nieminen, P.; Kantola, S.; Läärä, E.; Salo, T. Cancer-associated fibroblasts, a parameter of the tumor microenvironment, overcomes carcinoma-associated parameters in the prognosis of patients with mobile tongue cancer. Oral Oncol. 2011, 47, 33–38. [Google Scholar] [CrossRef] [PubMed]
- Takahashi, H.; Sakakura, K.; Kudo, T.; Toyoda, M.; Kaira, K.; Oyama, T.; Chikamatsu, K. Cancer-associated fibroblasts promote an immunosuppressive microenvironment through the induction and accumulation of protumoral macrophages. Oncotarget 2017, 8, 8633–8647. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Wu, Q.; Liu, Z.; Luo, X.; Fan, Y.; Liu, Y.; Zhang, Y.; Hua, S.; Fu, Q.; Zhao, M.; et al. Downregulation of FAP suppresses cell proliferation and metastasis through PTEN/PI3K/AKT and Ras-ERK signaling in oral squamous cell carcinoma. Cell Death Dis. 2014, 5. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Ma, N.; Kawanishi, S.; Hiraku, Y.; Oikawa, S.; Xie, Y.; Zhang, Z.; Huang, G.; Murata, M. Relationships of alpha-SMA-positive fibroblasts and SDF-1-positive tumor cells with neoangiogenesis in nasopharyngeal carcinoma. Biomed. Res. Int. 2014, 2014. [Google Scholar] [CrossRef] [PubMed]
- Hashimoto, O.; Yoshida, M.; Koma, Y.I.; Yanai, T.; Hasegawa, D.; Kosaka, Y.; Nishimura, N.; Yokozaki, H. Collaboration of cancer-associated fibroblasts and tumour-associated macrophages for neuroblastoma development. J. Pathol. 2016, 240, 211–223. [Google Scholar] [CrossRef]
- Minna, E.; Brich, S.; Todoerti, K.; Pilotti, S.; Collini, P.; Bonaldi, E.; Romeo, P.; de Cecco, L.; Dugo, M.; Perrone, F.; et al. Cancer associated fibroblasts and senescent thyroid cells in the invasive front of thyroid carcinoma. Cancers 2020, 12, 112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Su, S.; Chen, J.; Yao, H.; Liu, J.; Yu, S.; Lao, L.; Wang, M.; Luo, M.; Xing, Y.; Chen, F.; et al. CD10+GPR77+ Cancer-Associated Fibroblasts Promote Cancer Formation and Chemoresistance by Sustaining Cancer Stemness. Cell 2018, 172, 841–856.e16. [Google Scholar] [CrossRef] [PubMed]
- Zoetemelk, M.; Rausch, M.; Colin, D.J.; Dormond, O.; Nowak-Sliwinska, P. Short-term 3D culture systems of various complexity for treatment optimization of colorectal carcinoma. Sci. Rep. 2019, 9. [Google Scholar] [CrossRef] [PubMed]
- Tilghman, R.W.; Blais, E.M.; Cowan, C.R.; Sherman, N.E.; Grigera, P.R.; Jeffery, E.D.; Fox, J.W.; Blackman, B.R.; Tschumperlin, D.J.; Papin, J.A.; et al. Matrix rigidity regulates cancer cell growth by modulating cellular metabolism and protein synthesis. PLoS ONE 2012, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nebuloni, M.; Albarello, L.; Andolfo, A.; Magagnotti, C.; Genovese, L.; Locatelli, I.; Tonon, G.; Longhi, E.; Zerbi, P.; Allevi, R.; et al. Insight on Colorectal Carcinoma Infiltration by Studying Perilesional Extracellular Matrix. Sci. Rep. 2016, 6. [Google Scholar] [CrossRef]
- Xing, F.; Saidou, J.; Watabe, K. Cancer associated fibroblasts (CAFs) in tumor microenvironment. Front. Biosci. 2010, 15, 166–179. [Google Scholar] [CrossRef] [Green Version]
- Mezawa, Y.; Orimo, A. The roles of tumor- and metastasis-promoting carcinoma-associated fibroblasts in human carcinomas. Cell Tissue Res. 2016, 365, 675–689. [Google Scholar] [CrossRef]
- Togo, S.; Polanska, U.M.; Horimoto, Y.; Orimo, A. Carcinoma-associated fibroblasts are a promising therapeutic target. Cancers 2013, 5, 149–169. [Google Scholar] [CrossRef] [Green Version]
- Rick, J.W.; Chandra, A.; Dalle-Ore, C.; Nguyen, A.T.; Yagnik, G.; Aghi, M.K. Fibronectin in malignancy: Cancer-specific alterations, protumoral effects, and therapeutic implications. Semin. Oncol. 2019, 46, 284–290. [Google Scholar] [CrossRef] [PubMed]
- Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef] [PubMed]
- Shi, Y.; Du, L.; Lin, L.; Wang, Y. Tumour-associated mesenchymal stem/stromal cells: Emerging therapeutic targets. Nat. Rev. Drug Discov. 2016, 16, 35–52. [Google Scholar] [CrossRef] [PubMed]
- Worthley, D.L.; Si, Y.; Quante, M.; Churchill, M.; Mukherjee, S.; Wang, T.C. Bone marrow cells as precursors of the tumor stroma. Exp. Cell Res. 2013, 319, 1650–1656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Forino, M.; Torregrossa, R.; Ceol, M.; Murer, L.; Vella, M.D.; Prete, D.D.; D’Angelo, A.; Anglani, F. TGFβ1 induces epithelial-mesenchymal transition, but not myofibroblast transdifferentiation of human kidney tubular epithelial cells in primary culture. Int. J. Exp. Pathol. 2006, 87, 197–208. [Google Scholar] [CrossRef]
- Oft, M.; Akhurst, R.J.; Balmain, A. Metastasis is driven by sequential elevation of H-ras and Smad2 levels. Nat. Cell Biol. 2002, 4, 487–494. [Google Scholar] [CrossRef]
- Russo, F.P.; Alison, M.R.; Bigger, B.W.; Amofah, E.; Florou, A.; Amin, F.; Bou-Gharios, G.; Jeffery, R.; Iredale, J.P.; Forbes, S.J. The Bone Marrow Functionally Contributes to Liver Fibrosis. Gastroenterology 2006, 130, 1807–1821. [Google Scholar] [CrossRef]
- Zeisberg, E.M.; Tarnavski, O.; Zeisberg, M.; Dorfman, A.L.; McMullen, J.R.; Gustafsson, E.; Chandraker, A.; Yuan, X.; Pu, W.T.; Roberts, A.B.; et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat. Med. 2007, 13, 952–961. [Google Scholar] [CrossRef]
- Jung, Y.; Kim, J.K.; Shiozawa, Y.; Wang, J.; Mishra, A.; Joseph, J.; Berry, J.E.; McGee, S.; Lee, E.; Sun, H.; et al. Recruitment of mesenchymal stem cells into prostate tumours promotes metastasis. Nat. Commun. 2013, 4. [Google Scholar] [CrossRef] [Green Version]
- Quante, M.; Tu, S.P.; Tomita, H.; Gonda, T.; Wang, S.S.W.; Takashi, S.; Baik, G.H.; Shibata, W.; di Prete, B.; Betz, K.S.; et al. Bone Marrow-Derived Myofibroblasts Contribute to the Mesenchymal Stem Cell Niche and Promote Tumor Growth. Cancer Cell 2011, 19, 257–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patel, A.K.; Singh, S. Cancer associated fibroblasts: Phenotypic and functional heterogeneity. Front. Biosci. Landmark 2020, 25, 961–978. [Google Scholar] [CrossRef]
- Pechkovsky, D.V.; Hackett, T.L.; An, S.S.; Shaheen, F.; Murray, L.A.; Knight, D.A. Human lung parenchyma but not proximal bronchi produces fibroblasts with enhanced TGF-β signaling and α-SMA expression. Am. J. Respir. Cell Mol. Biol. 2010, 43, 641–651. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Z.; Zhou, Q.; Wu, X.; Xu, S.; Hu, X.; Tao, X.; Li, B.; Peng, J.; Li, D.; Shen, L.; et al. VCAM-1 secreted from cancer-associated fibroblasts enhances the growth and invasion of lung cancer cells through AKT and MAPK signaling. Cancer Lett. 2020, 473, 62–73. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.J.; Ho, C.C.; Chang, Y.L.; Chen, H.Y.; Lin, C.A.; Ling, T.Y.; Yu, S.L.; Yuan, S.S.; Louisa-Chen, Y.J.; Lin, C.Y.; et al. Cancer-associated fibroblasts regulate the plasticity of lung cancer stemness via paracrine signalling. Nat. Commun. 2014, 5. [Google Scholar] [CrossRef]
- Salimifard, S.; Masjedi, A.; Hojjat-Farsangi, M.; Ghalamfarsa, G.; Irandoust, M.; Azizi, G.; Mohammadi, H.; Keramati, M.R.; Jadidi-Niaragh, F. Cancer associated fibroblasts as novel promising therapeutic targets in breast cancer. Pathol. Res. Pract. 2020, 216. [Google Scholar] [CrossRef]
- Jung, Y.Y.; Kim, H.M.; Koo, J.S. The role of cancer-associated fibroblasts in breast cancer pathobiology. Histol Histopathol. 2016, 31, 371–378. [Google Scholar] [CrossRef]
- Yu, Y.; Xiao, C.H.; Tan, L.D.; Wang, Q.S.; Li, X.Q.; Feng, Y.M. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of breast cancer cells through paracrine TGF-β signalling. Br. J. Cancer. 2014, 110, 724–732. [Google Scholar] [CrossRef] [Green Version]
- Sirica, A.E.; Campbell, D.J.; Dumur, C.I. Cancer-associated fibroblasts in intrahepatic cholangiocarcinoma. Curr. Opin. Gastroenterol. 2011, 27, 276–284. [Google Scholar] [CrossRef]
- Parikh, J.G.; Kulkarni, A.; Johns, C. A-Smooth Muscle Actin-Positive Fibroblasts Correlate With Poor Survival in Hepatocellular Carcinoma. Oncol. Lett. 2014, 7, 573–575. [Google Scholar] [CrossRef] [Green Version]
- Liu, L.; Zhang, Z.; Zhou, L.; Hu, L.; Yin, C.; Qing, D.; Huang, S.; Cai, X.; Chen, Y. Cancer associated fibroblasts-derived exosomes contribute to radioresistance through promoting colorectal cancer stem cells phenotype. Exp. Cell Res. 2020, 391. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Liu, J.; Zhang, Q.; Liu, B.; Cheng, Y.; Zhang, Y.; Sun, Y.; Ge, H.; Liu, Y. Exosome-mediated transfer of miR-93-5p from cancer-associated fibroblasts confer radioresistance in colorectal cancer cells by downregulating FOXA1 and upregulating TGFB3. J. Exp. Clin. Cancer Res. 2020, 39. [Google Scholar] [CrossRef]
- Cangkrama, M.; Wietecha, M.; Mathis, N.; Okumura, R.; Ferrarese, L.; Al-Nuaimi, D.; Antsiferova, M.; Dummer, R.; Innocenti, M.; Werner, S. A paracrine activin A–mDia2 axis promotes squamous carcinogenesis via fibroblast reprogramming. EMBO Mol. Med. 2020, 12. [Google Scholar] [CrossRef] [PubMed]
- Denton, A.E.; Roberts, E.W.; Fearon, D.T. Stromal cells in the tumor microenvironment. In Stromal Immunology, Advances in Experimental Medicine and Biology; Springer International Publishing: Cham, Switzerland, 2018; Volume 1060, pp. 99–114. [Google Scholar] [CrossRef]
- Boire, A.; Covic, L.; Agarwal, A.; Jacques, S.; Sherifi, S.; Kuliopulos, A. PAR1 is a matrix metalloprotease-1 receptor that promotes invasion and tumorigenesis of breast cancer cells. Cell 2005, 120, 303–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Orimo, A.; Gupta, P.B.; Sgroi, D.C.; Arenzana-Seisdedos, F.; Delaunay, T.; Naeem, R.; Carey, V.J.; Richardson, A.L.; Weinberg, R.A. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 2005, 121, 335–348. [Google Scholar] [CrossRef] [PubMed]
- Dvorak, K.M.; Pettee, K.M.; Rubinic-Minotti, K.; Su, R.; Nestor-Kalinoski, A.; Eisenmann, K.M. Carcinoma associated fibroblasts (CAFs) promote breast cancer motility by suppressing mammalian Diaphanous-related formin-2 (mDia2). PLoS ONE 2018, 13. [Google Scholar] [CrossRef]
- De Francesco, E.M.; Lappano, R.; Santolla, M.F.; Marsico, S.; Caruso, A.; Maggiolini, M. HIF-1α/GPER signaling mediates the expression of VEGF induced by hypoxia in breast cancer associated fibroblasts (CAFs). Breast Cancer Res. 2013, 15. [Google Scholar] [CrossRef] [Green Version]
- Liu, Z.; Chen, M.; Zhao, R.; Huang, Y.; Liu, F.; Li, B.; Qin, Y. CAF-induced placental growth factor facilitates neoangiogenesis in hepatocellular carcinoma. Acta Biochim. Biophys. Sin. 2020, 52, 18–25. [Google Scholar] [CrossRef]
- Lukashev, M.E.; Werb, Z. ECM signalling: Orchestrating cell behaviour and misbehaviour. Trends Cell Biol. 1998, 8, 437–441. [Google Scholar] [CrossRef]
- Gaggioli, C.; Hooper, S.; Hidalgo-Carcedo, C.; Grosse, R.; Marshall, J.F.; Harrington, K.; Sahai, E. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat. Cell Biol. 2007, 9, 1392–1400. [Google Scholar] [CrossRef]
- Malanchi, I.; Santamaria-Martínez, A.; Susanto, E.; Peng, H.; Lehr, H.A.; Delaloye, J.F.; Huelsken, J. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature 2012, 481, 85–91. [Google Scholar] [CrossRef] [PubMed]
- Oskarsson, T.; Massagué, J. Extracellular matrix players in metastatic niches. EMBO J. 2012, 31, 254–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shen, J.; Zhai, J.; You, Q.; Zhang, G.; He, M.; Yao, X.; Shen, L. Cancer-associated fibroblasts-derived VCAM1 induced by H. pylori infection facilitates tumor invasion in gastric cancer. Oncogene 2020, 39, 2961–2974. [Google Scholar] [CrossRef] [PubMed]
- Pazolli, E.; Luo, X.; Brehm, S.; Carbery, K.; Chung, J.J.; Prior, J.L.; Doherty, J.; Demehri, S.; Salavaggione, L.; Piwnica-Worms, D.; et al. Senescent stromal-derived osteopontin promotes preneoplastic cell growth. Cancer Res. 2009, 69, 1230–1239. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Yang, Y.; Du, J.; Lin, D.; Li, F. MiR-3613-3p from carcinoma-associated fibroblasts exosomes promoted breast cancer cell proliferation and metastasis by regulating SOCS2 expression. IUBMB Life 2020, 72, 1705–1714. [Google Scholar] [CrossRef]
- Xiang, L.; Song, Z.; Rong, G. Taxotere-induced WNT16 Expression in Carcinoma-Associated Fibroblasts Might Associate with Progression and Chemoresistance of Breast Cancer. Ann. Clin. Lab. Sci. 2020, 50, 205–212. [Google Scholar]
- Demircioglu, F.; Wang, J.; Candido, J.; Costa, A.S.H.; Casado, P.; de Luxan-Delgado, B.; Reynolds, L.E.; Gomez-Escudero, J.; Newport, E.; Rajeeve, V.; et al. Cancer associated fibroblast FAK regulates malignant cell metabolism. Nat. Commun. 2020, 11. [Google Scholar] [CrossRef] [Green Version]
- Bakhtyar, N.; Wong, N.; Kapoor, A.; Cutz, J.C.; Hill, B.; Ghert, M.; Tang, D. Clear cell renal cell carcinoma induces fibroblast-mediated production of stromal periostin. Eur. J. Cancer 2013, 49, 3537–3546. [Google Scholar] [CrossRef]
- Chen, J.; Yang, P.; Xiao, Y.; Zhang, Y.; Liu, J.; Xie, D.; Cai, M.; Zhang, X. Overexpression of α-sma-positive fibroblasts (CAFs) in nasopharyngeal Carcinoma predicts poor prognosis. J. Cancer 2017, 8, 3897–3902. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Z.; Cui, D.; Sun, M.H.; Huang, J.L.; Deng, Z.; Han, B.M.; Sun, X.W.; Xia, S.J.; Sun, F.; Shi, F. CAFs-derived MFAP5 promotes bladder cancer malignant behavior through NOTCH2/HEY1 signaling. FASEB J. 2020, 34, 7970–7988. [Google Scholar] [CrossRef] [Green Version]
- Flier, J.S.; Underhill, L.H.; Dvorak, H.F. Tumors: Wounds That Do Not Heal. N. Engl. J. Med. 1986, 315, 1650–1659. [Google Scholar] [CrossRef] [PubMed]
- Allavena, P.; Sica, A.; Garlanda, C.; Mantovani, A. The Yin-Yang of tumor-associated macrophages in neoplastic progression and immune surveillance. Immunol. Rev. 2008, 222, 155–161. [Google Scholar] [CrossRef] [PubMed]
- Chiarugi, P. Cancer-associated fibroblasts and macrophages friendly conspirators for malignancy. Oncoimmunology 2013, 2. [Google Scholar] [CrossRef] [Green Version]
- Yu, Y.; Ke, L.; Lv, X.; Ling, Y.H.; Lu, J.; Liang, H.; Qiu, W.; Huang, X.; Liu, G.; Li, W.; et al. The prognostic significance of carcinoma-associated fibroblasts and tumor-associated macrophages in nasopharyngeal carcinoma. Cancer Manag. Res. 2018, 10, 1935–1946. [Google Scholar] [CrossRef] [Green Version]
- Feig, C.; Jones, J.O.; Kraman, M.; Wells, R.J.B.; Deonarine, A.; Chan, D.S.; Connell, C.M.; Roberts, E.W.; Zhao, Q.; Caballero, O.L.; et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 20212–20217. [Google Scholar] [CrossRef] [Green Version]
- Kato, T.; Noma, K.; Ohara, T.; Kashima, H.; Katsura, Y.; Sato, H.; Komoto, S.; Katsube, R.; Ninomiya, T.; Tazawa, H.; et al. Cancer-associated fibroblasts affect intratumoral CD8þ and Foxp3þ T cells via IL6 in the tumor microenvironment. Clin. Cancer Res. 2018, 24, 4820–4833. [Google Scholar] [CrossRef] [Green Version]
- Warburg, O.; Wind, F.; Negelein, E. The metabolism of tumors in the body. J. Gen. Physiol. 1927, 8, 519–530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gentric, G.; Mechta-Grigoriou, F. Tumor cells and cancer-associated fibroblasts: An updated metabolic perspective. Cancers 2021, 13, 399. [Google Scholar] [CrossRef]
- Pavlides, S.; Whitaker-Menezes, D.; Castello-Cros, R.; Flomenberg, N.; Witkiewicz, A.K.; Frank, P.G.; Casimiro, M.C.; Wang, C.; Fortina, P.; Addya, S.; et al. The reverse Warburg effect: Aerobic glycolysis in cancer associated fibroblasts and the tumor stroma. Cell Cycle 2009, 8, 3984–4001. [Google Scholar] [CrossRef] [Green Version]
- Guido, C.; Whitaker-Menezes, D.; Capparelli, C.; Balliet, R.; Lin, Z.; Pestell, R.G.; Howell, A.; Aquila, S.; Ando, S.; Martinez-Outschoorn, U.; et al. Metabolic reprogramming of cancer-associated fibroblasts by TGF-β drives tumor growth: Connecting TGF-β signaling with “Warburg- like” cancer metabolism and L-lactate production. Cell Cycle 2012, 11, 3019–3035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, D.; Wang, Y.; Shi, Z.; Liu, J.; Sun, P.; Hou, X.; Zhang, J.; Zhao, S.; Zhou, B.P.; Mi, J. Metabolic Reprogramming of Cancer-Associated Fibroblasts by IDH3α Downregulation. Cell Rep. 2015, 10, 1335–1348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fiaschi, T.; Marini, A.; Giannoni, E.; Taddei, M.L.; Gandellini, P.; De Donatis, A.; Lanciotti, M.; Serni, S.; Cirri, P.; Chiarugi, P. Reciprocal metabolic reprogramming through lactate shuttle coordinately influences tumor-stroma interplay. Cancer Res. 2012, 72, 5130–5140. [Google Scholar] [CrossRef] [Green Version]
- Knudsen, E.S.; Balaji, U.; Freinkman, E.; McCue, P.; Witkiewicz, A.K. Unique metabolic features of pancreatic cancer stroma: Relevance to the tumor compartment, prognosis, and invasive potential. Oncotarget 2016, 7, 78396–78411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lemons, J.M.S.; Coller, H.A.; Feng, X.J.; Bennett, B.D.; Legesse-Miller, A.; Johnson, E.L.; Raitman, I.; Pollina, E.A.; Rabitz, H.A.; Rabinowitz, J.D. Quiescent fibroblasts exhibit high metabolic activity. PLoS Biol. 2010, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Givel, A.M.; Kieffer, Y.; Scholer-Dahirel, A.; Sirven, P.; Cardon, M.; Pelon, F.; Magagna, I.; Gentric, G.; Costa, A.; Bonneau, C.; et al. MiR200-regulated CXCL12β promotes fibroblast heterogeneity and immunosuppression in ovarian cancers. Nat. Commun. 2018, 9. [Google Scholar] [CrossRef] [PubMed]
- Xiao, Z.; Dai, Z.; Locasale, J.W. Metabolic landscape of the tumor microenvironment at single cell resolution. Nat. Commun. 2019, 10. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Achreja, A.; Yeung, T.L.; Mangala, L.S.; Jiang, D.; Han, C.; Baddour, J.; Marini, J.C.; Ni, J.; Nakahara, R.; et al. Targeting Stromal Glutamine Synthetase in Tumors Disrupts Tumor Microenvironment-Regulated Cancer Cell Growth. Cell Metab. 2016, 24, 685–700. [Google Scholar] [CrossRef] [Green Version]
- Straussman, R.; Morikawa, T.; Shee, K.; Barzily-Rokni, M.; Qian, Z.R.; Du, J.; Davis, A.; Mongare, M.M.; Gould, J.; Frederick, D.T.; et al. Tumor microenvironment induces innate RAF-inhibitor resistance through HGF secretion. Nature 2012, 487, 500–504. [Google Scholar] [CrossRef] [Green Version]
- Amornsupak, K.; Insawang, T.; Thuwajit, P.; O-Charoenrat, P.; Eccles, S.A.; Thuwajit, C. Cancer-associated fibroblasts induce high mobility group box 1 and contribute to resistance to doxorubicin in breast cancer cells. BMC Cancer 2014, 14. [Google Scholar] [CrossRef]
- Miyai, Y.; Esaki, N.; Takahashi, M.; Enomoto, A. Cancer-associated fibroblasts that restrain cancer progression: Hypotheses and perspectives. Cancer Sci. 2020, 111, 1047–1057. [Google Scholar] [CrossRef] [Green Version]
- Özdemir, B.C.; Pentcheva-Hoang, T.; Carstens, J.L.; Zheng, X.; Wu, C.C.; Simpson, T.R.; Laklai, H.; Sugimoto, H.; Kahlert, C.; Novitskiy, S.V.; et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 2014, 25, 719–734. [Google Scholar] [CrossRef] [Green Version]
- Costa, A.; Kieffer, Y.; Scholer-Dahirel, A.; Pelon, F.; Bourachot, B.; Cardon, M.; Sirven, P.; Magagna, I.; Fuhrmann, L.; Bernard, C.; et al. Fibroblast Heterogeneity and Immunosuppressive Environment in Human Breast Cancer. Cancer Cell 2018, 33, 463–479.e10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bartoschek, M.; Oskolkov, N.; Bocci, M.; Lövrot, J.; Larsson, C.; Sommarin, M.; Madsen, C.D.; 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. 2018, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, H.M.; Jung, W.H.; Koo, J.S. Expression of cancer-associated fibroblast related proteins in metastatic breast cancer: An immunohistochemical analysis. J. Transl. Med. 2015, 13. [Google Scholar] [CrossRef] [Green Version]
- Cong, X.; Zhang, Y.; Zhu, Z.; Li, S.; Yin, X.; Zhai, Z.; Zhang, Y.; Xue, Y. CD66b+ neutrophils and α-SMA+ fibroblasts predict clinical outcomes and benefits from postoperative chemotherapy in gastric adenocarcinoma. Cancer Med. 2020, 9, 2761–2773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, T.; Han, C.; Wang, S.; Fang, P.; Ma, Z.; Xu, L.; Yin, R. Cancer-associated fibroblasts: An emerging target of anti-cancer immunotherapy. J. Hematol. Oncol. 2019, 12. [Google Scholar] [CrossRef] [PubMed]
- Geng, F.; Guo, J.; Guo, Q.Q.; Xie, Y.; Dong, L.; Zhou, Y.; Liu, C.L.; Yu, B.; Wu, H.; Wu, J.X.; et al. A DNA vaccine expressing an optimized secreted FAPα induces enhanced anti-tumor activity by altering the tumor microenvironment in a murine model of breast cancer. Vaccine 2019, 37, 4382–4391. [Google Scholar] [CrossRef] [PubMed]
- Neuwirt, H.; Bouchal, J.; Kharaishvili, G.; Ploner, C.; Jöhrer, K.; Pitterl, F.; Weber, A.; Klocker, H.; Eder, I.E. Cancer-associated fibroblasts promote prostate tumor growth and progression through upregulation of cholesterol and steroid biosynthesis. Cell Commun. Signal. 2020, 18. [Google Scholar] [CrossRef] [Green Version]
- Fiori, M.E.; di Franco, S.; Villanova, L.; Bianca, P.; Stassi, G.; De Maria, R. Cancer-associated fibroblasts as abettors of tumor progression at the crossroads of EMT and therapy resistance. Mol. Cancer 2019, 18. [Google Scholar] [CrossRef] [Green Version]
- Yamanaka, T.; Harimoto, N.; Yokobori, T.; Muranushi, R.; Hoshino, K.; Hagiwara, K.; Gantumur, D.; Handa, T.; Ishii, N.; Tsukagoshi, M.; et al. Nintedanib inhibits intrahepatic cholangiocarcinoma aggressiveness via suppression of cytokines extracted from activated cancer-associated fibroblasts. Br. J. Cancer. 2020, 122, 986–994. [Google Scholar] [CrossRef]
- Fourniols, T.; Bastien, E.; Canevat, A.; Feron, O.; Préat, V. Inhibition of colorectal cancer-associated fibroblasts by lipid nanocapsules loaded with acriflavine or paclitaxel. Int. J. Pharm. 2020, 584. [Google Scholar] [CrossRef] [PubMed]
- Lappano, R.; Rigiracciolo, D.C.; Belfiore, A.; Maggiolini, M.; De Francesco, E.M. Cancer associated fibroblasts: Role in breast cancer and potential as therapeutic targets. Expert Opin. Ther. Targets 2020, 24, 559–572. [Google Scholar] [CrossRef] [PubMed]
- Ignatov, A.; Ignatov, T.; Weienborn, C.; Eggemann, H.; Bischoff, J.; Semczuk, A.; Roessner, A.; Costa, S.D.; Kalinski, T. G-protein-coupled estrogen receptor GPR30 and tamoxifen resistance in breast cancer. Breast Cancer Res. Treat. 2011, 128, 457–466. [Google Scholar] [CrossRef]
- Shu, C.; Zha, H.; Long, H.; Wang, X.; Yang, F.; Gao, J.; Hu, C.; Zhou, L.; Guo, B.; Zhu, B. C3a-C3aR signaling promotes breast cancer lung metastasis via modulating carcinoma associated fibroblasts. J. Exp. Clin. Cancer Res. 2020, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Cancer Type | Markers | Relevant Studies |
---|---|---|
Breast | α-SMA, FAP, PDGFRα, PDGFRβ, CD29, NG2, FSP1, vimentin, PDPN | Jung et al. (2015) [35], Aboussekhra et al. (2011) [36], Pelon et al. (2020) [37], Yang et al. (2020) [38] |
Lung | α-SMA, FAP, vimentin, PDGFRβ, CD90, PDPN | Fan et al. (2020) [39], Xiang et al. (2020) [4], Neri et al. (2015) [40] |
Skin | α-SMA, FAP, vimentin, PDGFRα | Nie et al. (2019) [41], Ersek et al. (2020) [42], Erez et al. (2010) [20] |
Genitourinary: Bladder | α-SMA, FAP, CD90, vimentin, PDGFRα, PDGFRβ, MFAP5, FSP1 | Mezheyeuski et al. (2020) [43], Zhuang et al. (2015) [44] |
Genitourinary: Prostate | α-SMA, vimentin, FAP, FSP1, PDGFR-α, PDGFRβ | Ortiz-Otero et al. (2020) [32], Liu et al. (2020) [33], Shen et al. (2020) [34] |
Genitourinary: Renal | α-SMA, FAP, POSTN | Errarte et al. (2020) [45], Zagzag et al. (2005) [46] |
Genitourinary: Ovarian | α-SMA, FAP, FSP1, FGF-1 | Sun et al. (2017) [47], Guo et al. (2019) [48] |
Genitourinary: Endometrial | α-SMA, FSP1, FAP, vimentin | Teng et al. (2016) [26] |
Gastrointestinal: Colorectal | FAP, α-SMA, vimentin, FSP1, PDGFR-α | Herrera et al. (2013) [49], Bai et al. (2015) [24] |
Gastrointestinal: Esophageal | vimentin, a-SMA | Zhao et al. (2020) [50] |
Gastrointestinal: Gastric | FAP, α-SMA, FSP-1, vimentin, PDGRFα, PDGFRβ | Zhang et al. (2020) [23], Shi et al. (2020) [51], Ham et al. (2019) [52] |
Gastrointestinal: Pancreatic | α-SMA, vimentin, FAP, PDGFRβ, FSP1, PDGFR-α | Zhang et al. (2020) [23], Wei et al. (2018) [53] |
Gastrointestinal: Liver and Biliary System | α-SMA, FAP, FSP1, PDGFR-β, periostin | Chuaysri et al. (2009) [54], Zou et al. (2018) [55], Lau et al. (2016) [56], Sha et al. (2018) [57], Affo et al. (2017) [58] |
Gastrointestinal: Oral | FAP, α-SMA, vimentin | Bello et al. (2011) [59], Takahashi et al. (2017) [60], Wang et al. (2014) [61] |
Head and Neck | α-SMA, PDPN, FAP, PDGFR-α, PDGFR-β, FSP1, NG2 | Ramos-Vega et al. (2020) [7], Zhu et al. (2020) [10], Wang et al. (2014) [62] |
Endocrine/Neuroendocrine | A-SMA, FAP | Hashimoto et al. (2016) [63], Minna et al. (2020) [64] |
Gene/Marker | Drug | Observed Effect |
---|---|---|
SDF-1/CXCR4 axis | AMD3100 (Plerixafor) “an anti-SDF-1 neutralizing antibody” (clinical trial) [26,137] | Reversed FAP-positive AF-mediated immunosuppression; decreased proliferation, migration, and invasion as well as in vivo tumorigenesis |
TGF-β | Tranilast (Rizaben) (clinical trial) [137] | Inhibited TGF-β signaling in CAFs, facilitating T cell penetration into the tumor nest, and promoting anti-tumor immunity and tumor regression |
IL-6 | ROCKs and STAT3 “inhibitors of IL-6” (clinical trial) [137] | Improved the anti-tumor immune response and treated myeloproliferative diseases and autoimmune disorders |
FAP | FAP5-DM “anti-FAP monoclonal antibody” (pre-clinical study) [16]; “FAPα-targeted DNA vaccine” (pre-clinical study) [86,138]; Sibrotuzumab “anti-FAP monoclonal antibody” (clinical trial) [69] | Obstructed tumor growth for an extended time period or completely inhibited the tumor causing tumor regression; diminished secretion of SDF-1 and CCL2 |
Tenascin-C | 81C6 “anti-tenascin monoclonal antibody” (clinical trial) [69] | Targeted Tenascin-C, decreasing progression of colon cancer metastasis by mitigating response to TGF-beta signaling |
Androgen receptor | Simvastatin and AKR1C3 inhibitor (pre-clinical study) [139] | Overcame resistance to androgen receptor-targeted therapies in prostate cancer; enhanced tumor regression with targeted treatment |
Hedgehog signaling | Smoothened (SMO) inhibitor (clinical trial) [140] | Blocked Hedgehog signaling pathway activated in CAFs that fuels the therapy resistant phenotype in tumor cells |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Joshi, R.S.; Kanugula, S.S.; Sudhir, S.; Pereira, M.P.; Jain, S.; Aghi, M.K. The Role of Cancer-Associated Fibroblasts in Tumor Progression. Cancers 2021, 13, 1399. https://doi.org/10.3390/cancers13061399
Joshi RS, Kanugula SS, Sudhir S, Pereira MP, Jain S, Aghi MK. The Role of Cancer-Associated Fibroblasts in Tumor Progression. Cancers. 2021; 13(6):1399. https://doi.org/10.3390/cancers13061399
Chicago/Turabian StyleJoshi, Rushikesh S., Samanvi S. Kanugula, Sweta Sudhir, Matheus P. Pereira, Saket Jain, and Manish K. Aghi. 2021. "The Role of Cancer-Associated Fibroblasts in Tumor Progression" Cancers 13, no. 6: 1399. https://doi.org/10.3390/cancers13061399
APA StyleJoshi, R. S., Kanugula, S. S., Sudhir, S., Pereira, M. P., Jain, S., & Aghi, M. K. (2021). The Role of Cancer-Associated Fibroblasts in Tumor Progression. Cancers, 13(6), 1399. https://doi.org/10.3390/cancers13061399