Next Article in Journal
Imaging Delay Following Liver-Directed Therapy Increases Progression Risk in Early- to Intermediate-Stage Hepatocellular Carcinoma
Previous Article in Journal
Diffuse C-Cells Hyperplasia Is the Source of False Positive Calcitonin Measurement in FNA Washout Fluids of Thyroid Nodules: A Rational Clinical Approach to Avoiding Unnecessary Surgery
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Cancer-Associated Fibroblasts: Major Co-Conspirators in Tumor Development

1
Department of International Studies (Global Health), College of Arts and Sciences, University of South Alabama, Mobile, AL 36688, USA
2
Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
3
Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36617, USA
4
Department of Biochemistry and Molecular Biology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36688, USA
5
Biomedical Sciences, College of Medicine, Roseman University of Health Sciences, Las Vegas, NV 89135, USA
*
Authors to whom correspondence should be addressed.
Cancers 2024, 16(1), 211; https://doi.org/10.3390/cancers16010211
Submission received: 21 December 2023 / Accepted: 25 December 2023 / Published: 2 January 2024
(This article belongs to the Section Cancer Pathophysiology)
The tumor microenvironment (TME) is a critical determinant of tumor progression, metastasis, and therapeutic outcomes. It consists of an extracellular matrix (ECM) and a variety of stromal cells, which remain in constant crosstalk with each other and the tumor cells present. Within this complex ecosystem, cancer-associated fibroblasts (CAFs) play a pivotal role, orchestrating the TME through intricate interactions with other cells, leading to the remodeling of the extracellular matrix (ECM) [1,2]. A recently published review article in the journal Cancers by Joshi et al. [2] marvelously captured the information related to the origin of CAFs and their multi-faceted roles in tumor development, metastasis, angiogenesis, the immune microenvironment, and therapeutic resistance. The authors also discuss the potential utility of CAFs as prognostic markers and therapeutic targets in types of different cancer.
The origin of CAFs is often resident quiescent fibroblasts; however, research also suggests that they can originate through the trans-differentiation of epithelial cells, endothelial cells, pericytes, smooth muscle cells, bone-marrow-derived mesenchymal cells, and adipocytes [3,4,5,6,7,8,9]. The proportion of CAFs derived from sources other than resident fibroblasts varies among different tumor types. For instance, in breast cancer, adipocytes contribute to CAFs, while in squamous cell carcinoma and pancreatic adenocarcinoma, pericytes and stellate cells, respectively, are the major contributors [6,8,9]. Interestingly, some reports also mention that CAFs can originate from cancer stem cells (CSCs) that gain myofibroblast-like features [10,11,12].
Characterizing CAFs has been challenging due to their considerable heterogeneity within tumors and across various cancer types [2,13]. Common markers used for CAF identification include alpha-smooth muscle actin (α-SMA), vimentin, fibroblast activation protein (FAP), fibroblast-specific protein 1 (FSP1), and platelet-derived growth factor receptor α/β (PDGFR-α/β) [2]. The diversity in these CAF markers often results from their trans-differentiation, influenced by growth factors, miRNAs, and exosomes secreted by cancer cells [9]. For example, factors such as CXCL12, Wnt7a, miR-9, and miR-125b from cancer cells, can prompt resident fibroblasts to transform into α-SMA-expressing CAFs [14]. Moreover, TGF-β and PDGF can induce resident fibroblasts to express FSP, while miR-370-3p can stimulate fibroblasts to secrete interleukin (IL)-1β, IL-6, and IL-8, contributing to their transition into CAFs. These secreted factors commonly activate pathways such as CXCR4/CXCL12, TGFβR/Smads, Wnt/β-catenin, and AKT/ERK during the transactivation process of CAFs. Elevated FAP and PDGFR-α/β levels in CAFs correlate with decreased survival rates, while heightened α-SMA expression is associated with intensified tumor growth and metastasis across multiple tumor types [15]. The diversity in CAF markers offers opportunities to utilize specific biomarkers and CAF density for predicting the nature of the disease and making prognostic assessments [16].
Once CAFs are established in the TME, they promote tumor growth through the secretion of growth factors, cytokines, and chemokines, as well as through the shedding of extracellular vesicles (EVs). CAF-secreted factors encompass IL6/8, IL1β, hepatocyte growth factor (HGF), TGF-β, stromal-derived factor-1 (SDF-1/CXCL12), and PDGF [2]. These factors activate distinct pathways to facilitate tumor growth across various tumor types. For instance, SDF-1/CXCL12 secreted by CAFs in pancreatic cancer promotes chemoresistance, making CXCR4 signaling an attractive target for therapeutic development [17,18]. CXCL12/CXCR4 signaling has also been shown to counteract docetaxel therapy in prostate cancer [19]. Similarly, osteopontin (OPN) and vascular cell adhesion molecule-1 (VCAM-1) secreted by CAFs in lung cancer activate AKT and MAPK signaling to promote tumor cell invasion and metastasis [20,21]. In breast cancer, α-SMA-positive CAFs induce EMT in tumor cells by secreting TGF-β and CXCL12 to activate their downstream signaling [22,23]. Similarly, CAFs from colorectal and ovarian cancers are shown to secrete FGF-1, which stimulates MAPK/ERK signaling and promotes aggressive tumor phenotypes [24,25].
CAFs’ role in tumor invasion and metastasis involves orchestrating ECM remodeling and triggering EMT and actin-rich invadosome formation in tumor cells. In addition, through dysregulation of enzymes like lysyl oxidases (LOXs), matrix metalloproteases (MMPs), and transglutaminases, CAFs disrupt ECM stability, fostering invasion and dissemination of cancer cells [13,26]. Activation of TWIST, ZEB, SNAIL/SLUG transcription factors by CAFs induces EMT, leading to heightened mesenchymal markers (vimentin, fibronectin, N-cadherin) and suppression of epithelial junction proteins (E-cadherin, occludins, claudins) [27,28]. CAF-secreted TGF-β crucially promotes EMT, enhancing markers such as vimentin, SNAIL, and ZEB2 [29,30]. Moreover, CAFs contribute to EMT through the non-canonical Wnt pathway, often interacting with STAT-3 and survivin [31]. Other factors such as IL-6, IL-8, OPN, HGF, and CXCL12 also influence EMT through diverse pathways. For example, OPN affects the TWIST pathway, CXCL-12 triggers Wnt/β-catenin, HGF facilitates collagen degradation, and IL-6 activates the STAT3 and MEK/ERK pathways [32,33,34]. The variations in cytokine secretion among CAFs in different tumors add complexity to understanding their specific EMT-promoting mechanisms.
Emerging evidence suggests that CAFs frequently utilize exosomes to communicate with tumor cells. Additionally, these exosomes carry various microRNAs (miRNAs), facilitating migration, invasion, metastasis, and therapeutic resistance. Among the most prevalent miRNAs found in CAF exosomes across different tumors are miR-21, miR378-e, miR-148, and miR-92a-3p, known to be involved in EMT and therapeutic resistance mechanisms [35].
It is suggested that cancer cells rely predominantly on glycolysis and not on mitochondrial oxidative phosphorylation (OXPHOS) for their energy needs even in the presence of oxygen, a process known as aerobic glycolysis or the “Warburg effect”. However, in some cancer cells, a “Reverse Warburg effect” is noted which results from the metabolic coupling between cancer and stromal cells. Neoplastic cells trigger oxidative stress in nearby fibroblasts by releasing reactive oxygen species (ROS), prompting glycolysis and lactate production through NF-κB signaling. This reprograms CAFs, lowering caveolin 1 (CAV1) and increasing monocarboxylate transporter 4 (MCT4), leading to lactate release. Nearby cancer cells respond with heightened MCT1, enabling lactate uptake and T53-induced glycolysis and apoptosis regulator (TIGAR) which suppresses glycolysis and increases OXPHOS [36,37]. Similar observations have also been made between hypoxic and non-hypoxic cancer cells [38]. In other instances, CAFs are shown to reuse cancer-derived lactate to maintain fibrotic conditions and induce immunosuppression [39]. Thus, the metabolic partnership between cancer cells and CAFs enables the efficient use of available nutrient resources and oxygen to favor uninhabited tumor growth and metastasis.
The intricate involvement of CAFs in tumor growth, invasion, and metastasis positions them as potential therapeutic targets. Various markers expressed in CAFs and pathways crucial to tumor growth and metastasis have been the focus of targeted therapies in both pre-clinical and clinical trials. For instance, targeting the TGF-β pathway in hepatic, ovarian, pancreatic, and breast cancers has shown efficacy in preventing CAF activation and inhibiting metastasis [40,41,42,43]. However, clinical trials aimed at inhibiting MMPs did not yield the desired outcomes, although monoclonal antibody targeting demonstrated a reduction in tumor growth and metastasis [44]. Similarly, targeting FAP with antibodies in lung cancer has shown promise, but its non-specific expression in CAFs is associated with potential side effects [16,45]. Furthermore, targeting SDF-1, TGF-β, and IL-6 in CAFs has displayed the potential to reverse tumor immunosuppression in clinical trials [46]. In pancreatic and breast cancers, targeting the hedgehog pathway exhibited potential by reducing fibroblast accumulation [47] but was met with failure in a later clinical trial [48]. Similarly, in another study, the depletion of CAFs induced immunosuppression and promoted tumor aggressiveness [49]. Thus, CAFs’ involvement in cancer progression is more complex than thought and may vary along the course of cancer evolution. The complexity arising from diverse CAF subpopulations may underlie such contrasting observations and must be studied in detail in order to develop a clinically useful intervention approach. Thus, our comprehension of CAF markers, subpopulations, and signaling is still incomplete and requires extensive investigations to reap clinical benefits for cancer patients from their therapeutic targeting.

Funding

The authors would like to acknowledge funding from the Department of Defense (W81XWH-22-1-0913 and HT94252310452) and the NCI/NIH (R01CA224306).

Conflicts of Interest

The authors have no conflicts of interest.

References

  1. Asif, P.J.; Longobardi, C.; Hahne, M.; Medema, J.P. The Role of Cancer-Associated Fibroblasts in Cancer Invasion and Metastasis. Cancers 2021, 13, 4720. [Google Scholar] [CrossRef] [PubMed]
  2. 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. [Google Scholar] [CrossRef]
  3. Arina, A.; Idel, C.; Hyjek, E.M.; Alegre, M.L.; Wang, Y.; Bindokas, V.P.; Weichselbaum, R.R.; Schreiber, H. Tumor-associated fibroblasts predominantly come from local and not circulating precursors. Proc. Natl. Acad. Sci. USA 2016, 113, 7551–7556. [Google Scholar] [CrossRef]
  4. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef] [PubMed]
  5. Shi, Y.; Du, L.; Lin, L.; Wang, Y. Tumour-associated mesenchymal stem/stromal cells: Emerging therapeutic targets. Nat. Rev. Drug Discov. 2017, 16, 35–52. [Google Scholar] [CrossRef] [PubMed]
  6. Hosaka, K.; Yang, Y.; Seki, T.; Fischer, C.; Dubey, O.; Fredlund, E.; Hartman, J.; Religa, P.; Morikawa, H.; Ishii, Y.; et al. Pericyte-fibroblast transition promotes tumor growth and metastasis. Proc. Natl. Acad. Sci. USA 2016, 113, E5618–E5627. [Google Scholar] [CrossRef] [PubMed]
  7. Fotsitzoudis, C.; Koulouridi, A.; Messaritakis, I.; Konstantinidis, T.; Gouvas, N.; Tsiaoussis, J.; Souglakos, J. Cancer-Associated Fibroblasts: The Origin, Biological Characteristics and Role in Cancer-A Glance on Colorectal Cancer. Cancers 2022, 14, 4394. [Google Scholar] [CrossRef]
  8. Manoukian, P.; Bijlsma, M.; van Laarhoven, H. The Cellular Origins of Cancer-Associated Fibroblasts and Their Opposing Contributions to Pancreatic Cancer Growth. Front. Cell Dev. Biol. 2021, 9, 743907. [Google Scholar] [CrossRef]
  9. 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. 2022, 42, 401–434. [Google Scholar] [CrossRef]
  10. Huang, Z.; Wu, T.; Liu, A.Y.; Ouyang, G. Differentiation and transdifferentiation potentials of cancer stem cells. Oncotarget 2015, 6, 39550–39563. [Google Scholar] [CrossRef]
  11. Osman, A.; Afify, S.M.; Hassan, G.; Fu, X.; Seno, A.; Seno, M. Revisiting Cancer Stem Cells as the Origin of Cancer-Associated Cells in the Tumor Microenvironment: A Hypothetical View from the Potential of iPSCs. Cancers 2020, 12, 879. [Google Scholar] [CrossRef] [PubMed]
  12. Nair, N.; Calle, A.S.; Zahra, M.H.; Prieto-Vila, M.; Oo, A.K.K.; Hurley, L.; Vaidyanath, A.; Seno, A.; Masuda, J.; Iwasaki, Y.; et al. A cancer stem cell model as the point of origin of cancer-associated fibroblasts in tumor microenvironment. Sci. Rep. 2017, 7, 6838. [Google Scholar] [CrossRef] [PubMed]
  13. Friedman, G.; Levi-Galibov, O.; David, E.; Bornstein, C.; Giladi, A.; Dadiani, M.; Mayo, A.; Halperin, C.; Pevsner-Fischer, M.; Lavon, H.; et al. Cancer-associated fibroblast compositions change with breast cancer progression linking the ratio of S100A4(+) and PDPN(+) CAFs to clinical outcome. Nat. Cancer 2020, 1, 692–708. [Google Scholar] [CrossRef] [PubMed]
  14. Kojima, Y.; Acar, A.; Eaton, E.N.; Mellody, K.T.; Scheel, C.; Ben-Porath, I.; Onder, T.T.; Wang, Z.C.; Richardson, A.L.; Weinberg, R.A.; et al. Autocrine TGF-β and stromal cell-derived factor-1 (SDF-1) signaling drives the evolution of tumor-promoting mammary stromal myofibroblasts. Proc. Natl. Acad. Sci. USA 2010, 107, 20009–20014. [Google Scholar] [CrossRef] [PubMed]
  15. 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] [PubMed]
  16. 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, 152915. [Google Scholar] [CrossRef] [PubMed]
  17. Singh, S.; Srivastava, S.K.; Bhardwaj, A.; Owen, L.B.; Singh, A.P. CXCL12-CXCR4 signalling axis confers gemcitabine resistance to pancreatic cancer cells: A novel target for therapy. Br. J. Cancer 2010, 103, 1671–1679. [Google Scholar] [CrossRef] [PubMed]
  18. Khan, M.A.; Srivastava, S.K.; Zubair, H.; Patel, G.K.; Arora, S.; Khushman, M.; Carter, J.E.; Gorman, G.S.; Singh, S.; Singh, A.P. Co-targeting of CXCR4 and hedgehog pathways disrupts tumor-stromal crosstalk and improves chemotherapeutic efficacy in pancreatic cancer. J. Biol. Chem. 2020, 295, 8413–8424. [Google Scholar] [CrossRef]
  19. Bhardwaj, A.; Srivastava, S.K.; Singh, S.; Arora, S.; Tyagi, N.; Andrews, J.; McClellan, S.; Carter, J.E.; Singh, A.P. CXCL12/CXCR4 signaling counteracts docetaxel-induced microtubule stabilization via p21-activated kinase 4-dependent activation of LIM domain kinase 1. Oncotarget 2014, 5, 11490–11500. [Google Scholar] [CrossRef]
  20. 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]
  21. 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]
  22. 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, 404. [Google Scholar] [CrossRef] [PubMed]
  23. 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] [PubMed]
  24. 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]
  25. 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, 1010428317712592. [Google Scholar] [CrossRef] [PubMed]
  26. Attieh, Y.; Clark, A.G.; Grass, C.; Richon, S.; Pocard, M.; Mariani, P.; Elkhatib, N.; Betz, T.; Gurchenkov, B.; Vignjevic, D.M. Cancer-associated fibroblasts lead tumor invasion through integrin-β3-dependent fibronectin assembly. J. Cell Biol. 2017, 216, 3509–3520. [Google Scholar] [CrossRef]
  27. Hwang, W.L.; Yang, M.H.; Tsai, M.L.; Lan, H.Y.; Su, S.H.; Chang, S.C.; Teng, H.W.; Yang, S.H.; Lan, Y.T.; Chiou, S.H.; et al. SNAIL regulates interleukin-8 expression, stem cell-like activity, and tumorigenicity of human colorectal carcinoma cells. Gastroenterology 2011, 141, 279–291.e5. [Google Scholar] [CrossRef]
  28. Xiong, H.; Hong, J.; Du, W.; Lin, Y.W.; Ren, L.L.; Wang, Y.C.; Su, W.Y.; Wang, J.L.; Cui, Y.; Wang, Z.H.; et al. Roles of STAT3 and ZEB1 proteins in E-cadherin down-regulation and human colorectal cancer epithelial-mesenchymal transition. J. Biol. Chem. 2012, 287, 5819–5832. [Google Scholar] [CrossRef]
  29. 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, 70. [Google Scholar] [CrossRef]
  30. Pohl, M.; Radacz, Y.; Pawlik, N.; Schoeneck, A.; Baldus, S.E.; Munding, J.; Schmiegel, W.; Schwarte-Waldhoff, I.; Reinacher-Schick, A. SMAD4 mediates mesenchymal-epithelial reversion in SW480 colon carcinoma cells. Anticancer Res. 2010, 30, 2603–2613. [Google Scholar]
  31. Lee, S.C.; Kim, O.H.; Lee, S.K.; Kim, S.J. IWR-1 inhibits epithelial-mesenchymal transition of colorectal cancer cells through suppressing Wnt/β-catenin signaling as well as survivin expression. Oncotarget 2015, 6, 27146–27159. [Google Scholar] [CrossRef] [PubMed]
  32. Kothari, A.N.; Arffa, M.L.; Chang, V.; Blackwell, R.H.; Syn, W.K.; Zhang, J.; Mi, Z.; Kuo, P.C. Osteopontin—A Master Regulator of Epithelial-Mesenchymal Transition. J. Clin. Med. 2016, 5, 39. [Google Scholar] [CrossRef] [PubMed]
  33. Ebbing, E.A.; van der Zalm, A.P.; Steins, A.; Creemers, A.; Hermsen, S.; Rentenaar, R.; Klein, M.; Waasdorp, C.; Hooijer, G.K.J.; Meijer, S.L.; et al. Stromal-derived interleukin 6 drives epithelial-to-mesenchymal transition and therapy resistance in esophageal adenocarcinoma. Proc. Natl. Acad. Sci. USA 2019, 116, 2237–2242. [Google Scholar] [CrossRef] [PubMed]
  34. Hu, T.H.; Yao, Y.; Yu, S.; Han, L.L.; Wang, W.J.; Guo, H.; Tian, T.; Ruan, Z.P.; Kang, X.M.; Wang, J.; et al. SDF-1/CXCR4 promotes epithelial-mesenchymal transition and progression of colorectal cancer by activation of the Wnt/β-catenin signaling pathway. Cancer Lett. 2014, 354, 417–426. [Google Scholar] [CrossRef] [PubMed]
  35. Hu, J.L.; Wang, W.; Lan, X.L.; Zeng, Z.C.; Liang, Y.S.; Yan, Y.R.; Song, F.Y.; Wang, F.F.; Zhu, X.H.; Liao, W.J.; et al. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Mol. Cancer 2019, 18, 91. [Google Scholar] [CrossRef] [PubMed]
  36. Wilde, L.; Roche, M.; Domingo-Vidal, M.; Tanson, K.; Philp, N.; Curry, J.; Martinez-Outschoorn, U. Metabolic coupling and the Reverse Warburg Effect in cancer: Implications for novel biomarker and anticancer agent development. Semin. Oncol. 2017, 44, 198–203. [Google Scholar] [CrossRef]
  37. 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]
  38. Wang, Z.H.; Peng, W.B.; Zhang, P.; Yang, X.P.; Zhou, Q. Lactate in the tumour microenvironment: From immune modulation to therapy. EBioMedicine 2021, 73, 103627. [Google Scholar] [CrossRef]
  39. Kitamura, F.; Semba, T.; Yasuda-Yoshihara, N.; Yamada, K.; Nishimura, A.; Yamasaki, J.; Nagano, O.; Yasuda, T.; Yonemura, A.; Tong, Y.; et al. Cancer-associated fibroblasts reuse cancer-derived lactate to maintain a fibrotic and immunosuppressive microenvironment in pancreatic cancer. JCI Insight 2023, 8, e163022. [Google Scholar] [CrossRef]
  40. Mazzocca, A.; Fransvea, E.; Dituri, F.; Lupo, L.; Antonaci, S.; Giannelli, G. Down-regulation of connective tissue growth factor by inhibition of transforming growth factor β blocks the tumor-stroma cross-talk and tumor progression in hepatocellular carcinoma. Hepatology 2010, 51, 523–534. [Google Scholar] [CrossRef]
  41. Zhang, Q.; Hou, X.; Evans, B.J.; VanBlaricom, J.L.; Weroha, S.J.; Cliby, W.A. LY2157299 Monohydrate, a TGF-βR1 Inhibitor, Suppresses Tumor Growth and Ascites Development in Ovarian Cancer. Cancers 2018, 10, 260. [Google Scholar] [CrossRef] [PubMed]
  42. Mardhian, D.F.; Storm, G.; Bansal, R.; Prakash, J. Nano-targeted relaxin impairs fibrosis and tumor growth in pancreatic cancer and improves the efficacy of gemcitabine in vivo. J. Control. Release 2018, 290, 1–10. [Google Scholar] [CrossRef] [PubMed]
  43. Yao, Y.; Guo, Q.; Cao, Y.; Qiu, Y.; Tan, R.; Yu, Z.; Zhou, Y.; Lu, N. Artemisinin derivatives inactivate cancer-associated fibroblasts through suppressing TGF-β signaling in breast cancer. J. Exp. Clin. Cancer Res. 2018, 37, 282. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, T.; Zhou, L.; Li, D.; Andl, T.; Zhang, Y. Cancer-Associated Fibroblasts Build and Secure the Tumor Microenvironment. Front. Cell Dev. Biol. 2019, 7, 60. [Google Scholar] [CrossRef] [PubMed]
  45. 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]
  46. 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, 86. [Google Scholar] [CrossRef] [PubMed]
  47. Olive, K.P.; Jacobetz, M.A.; Davidson, C.J.; Gopinathan, A.; McIntyre, D.; Honess, D.; Madhu, B.; Goldgraben, M.A.; Caldwell, M.E.; Allard, D.; et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 2009, 324, 1457–1461. [Google Scholar] [CrossRef]
  48. Allison, M. Hedgehog hopes lifted by approval… and stung by failure. Nat. Biotechnol. 2012, 30, 203. [Google Scholar] [CrossRef]
  49. Ozdemir, 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]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Singh, S.; Singh, A.P.; Mitra, R. Cancer-Associated Fibroblasts: Major Co-Conspirators in Tumor Development. Cancers 2024, 16, 211. https://doi.org/10.3390/cancers16010211

AMA Style

Singh S, Singh AP, Mitra R. Cancer-Associated Fibroblasts: Major Co-Conspirators in Tumor Development. Cancers. 2024; 16(1):211. https://doi.org/10.3390/cancers16010211

Chicago/Turabian Style

Singh, Shubhangi, Ajay P. Singh, and Ranjana Mitra. 2024. "Cancer-Associated Fibroblasts: Major Co-Conspirators in Tumor Development" Cancers 16, no. 1: 211. https://doi.org/10.3390/cancers16010211

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop