Fibroblast Subsets in Colorectal cancer: Comparison
Please note this is a comparison between Version 2 by Catherine Yang and Version 1 by Hao Dang.

CAFs are defined as fibroblasts surrounding malignant tumor cells and are the most abundant cell type in the tumor microenvironment of colorectal cancer (CRC). CAFs consist of various functionally heterogeneous subsets which can promote or restrain cancer progression. Although most previous research has focused on the biology of epithelial cells, accumulating evidence shows that certain CAF subsets can also importantly contribute to tumor initiation and progression, thereby possibly providing avenues for improvement of clinical care for CRC patients. However, attention needs to be paid to comprehensive characterization and reporting of the subsets being investigated in CAF studies to enable more precise identification and targeting of “unfavorable” CAFs in advanced CRC.

  • colorectal cancer
  • tumor stage
  • adenoma–carcinoma sequence
  • cancer-associated fibroblast

1. Introduction

Colorectal cancer (CRC) is the most commonly diagnosed malignancy in the gastrointestinal tract, constituting almost two million new cases and one million deaths per year worldwide [1,2][1][2]. The majority of CRC cases develop via the adenoma–carcinoma sequence [3]. This process is accompanied by sequential accumulation of (epi) genetic alterations in epithelial cells that lead to the formation of benign precursor lesions called adenomas [4]. Some of these adenomas eventually progress into invasive carcinomas. Although most previous research has focused on the biology of epithelial cells, it has become clear that the tumor microenvironment (TME), also known as the tumor stroma, plays an important role in CRC initiation and progression as well [5,6][5][6]. For instance, a high stromal content at the invasive front of CRCs is strongly correlated to an increased risk of CRC-related death [7,8][7][8]. Moreover, several studies have demonstrated that the prognostic value of the recently developed Consensus Molecular Subtypes (CMS) classification system for CRC [9] [9] can be mainly attributed to genes expressed by stromal cells, rather than tumor cells [10,11,12][10][11][12]. Of all the CMS categories, the mesenchymal or stromal CRC subtype (CMS4) is associated with the worst survival outcomes [9], thereby underlining the significant involvement of the TME in tumor progression.

The tumor stroma of CRC consists of several cell types (e.g., immune cells and endothelial cells), of which the cancer-associated fibroblasts (CAFs) are most abundant. CAFs are defined as fibroblasts surrounding malignant tumor cells [13], and consist of various heterogeneous subsets which can exert both tumor-promoting and -suppressing functions [5,6,13,14][5][6][13][14]. Many studies have shown that CAFs can make an important contribution to CRC progression. However, most of these findings have not yet been translated into applications with clinically relevant efficacy, mainly due to the considerable heterogeneity in CAF subsets and phenotypes in advanced CRCs [98,99,100,101,102][15][16][17][18][19]. To tackle this issue, Li et al. performed scRNAseq on human primary CRCs and found that CAFs seem to cluster into two major CAF types termed CAF-A (marker genes: e.g., MMP-2, FAP, and decorin) and CAF-B (marker genes: e.g., α-SMA, transgelin and platelet-derived growth factor subunit A (PDGFA)) [103][20]. Although it remains unclear how these CAF clusters are functionally involved in CRC progression, findings from clinical trials suggest that they are not exclusively composed of tumor-promoting or tumor-suppressing CAFs. For instance, therapeutic targeting of FAP-expressing CAFs (i.e., the CAF-A subset) does not appear to significantly affect clinical outcomes of patients with advanced CRC [97,104,105][21][22][23]. These results suggest that a more detailed, function-based subclassification of CAFs is required.

2. CAF subsets and functions in advanced CRC

In contrast to the scarce literature on tumor-suppressing functions of CAFs, there are quite some studies which have related various tumor-promoting functions to specific markers expressed by certain CAFs found in human CRCs (Table 1). However, as most of these CAFs have not been extensively characterized, it often remains unclear how they relate to the two major CAF clusters and whether they represent phenotypically distinct subsets or overlap with other tumor-promoting or -suppressing CAFs. In this entry, we provided an overview on (the markers of) these possible CAF subsets, stratified per tumor-promoting function which they have been linked to.

Table 1.

Examples of tumor-related functions which have been linked to factors expressed or secreted by certain cancer-associated fibroblast (CAF) populations in human advanced colorectal cancers (CRCs).

1

Combined with high expression of α-SMA.

2

High expression levels of podoplanin in CAFs were associated with favorable clinical outcomes in CRC patients.

3

Only in patients with SMAD4-deficient tumors. CAF: cancer-associated fibroblast, CRC: colorectal cancer, ECM: extracellular matrix, α-SMA: alpha smooth muscle actin, FAP: fibroblast activation protein, COX-2: cyclooxigenase-2, HIC-5: hydrogen peroxide-inducible clone-5, FGF: fibroblast growth factor, miRNA: microRNA, CD: cluster of differentiation, RAB31: Ras-related protein RAB-31, CLEC3B: C-type lectin domain family 3 member B, SPARC: secreted protein acidic and rich in cysteine, IGF-2: insulin-like growth factor 2, CCBE1: collagen and calcium-binding epidermal growth factor domain 1, lncRNA: long non-coding RNA, TGF- β2: transforming growth factor-β2, CXCL5: C-X-C motif chemokine 5, BMP2: bone morphogenetic protein 2, IL-11: interleukin 11, STC1: stanniocalcin-1. ↑ indicates a tumor-promoting role, ↓ indicates a tumor-suppressing role, = indicates a neutral role (i.e., neither tumor-promoting nor tumor-suppressing).

2.1. Tumor Growth, Invasion, and TME Remodeling

Numerous studies have shown that CAFs can importantly contribute to cancer cell proliferation and invasion [5,6,13,14][5][6][13][14]. In addition, they are also able to affect to tumor progression via remodeling of the ECM or regulation of tumor angiogenesis [106,107,108][57][58][59]. In advanced CRCs, tumor cell proliferation can be promoted by CAFs which express hydrogen peroxide-inducible clone-5 (HIC-5) [25] [109] or Snail-1 [110][28], or CAFs which secrete FGF-1, FGF-3 [111][30], or exosomal circular RNA SLC7A6 [112][24]. Interestingly, some of these CAFs were also found to promote tumor progression via other mechanisms such as enhancing angiogenesis [111][30] or CRC cell invasion [110,112,113][28][24][60]. Another example of a “multifunctional subset” includes Wnt2-expressing CAFs, which can promote CRC cell proliferation and migration [114,115][31][32], facilitate CRC invasion via ECM remodeling [115][32], and increase tumor angiogenesis by secreting several pro-angiogenic factors (e.g., angiopoietin-2 (ANG2), placental growth factor (PGF)) [116][33]. Besides, high levels of Wnt2-expressing CAFs were also associated with an increased risk of cancer metastasis and recurrence in advanced CRCs [114,115][31][32]. A comparable association with worse patient prognosis was found for high expression levels of microRNA-21 [117][36], a factor which is predominantly expressed in CAFs and marks a CAF population which could support CRC cell proliferation and invasion [118,119][34][35].

2.2. Therapeutic Resistance and Immune Regulation

It is well known that CAFs can also facilitate cancer progression by promoting tumor cell resistance to cytotoxic therapies [120,121][61][62]. Several studies have demonstrated that certain CAFs in human CRCs are also able to do so. For example, radiation-induced apoptosis in CRC cells can be reduced by CAFs which express microRNA-31 [122][45] or microRNA-93–5p [123][46]. Moreover, CAFs which express long non-coding RNA (lncRNA) H19 [48] [124] and colorectal cancer-associated lncRNA (CCAL) [125][47], can promote CRC cell stemness and chemoresistance via exosomal transfer of these lncRNAs to tumor cells. TGF-β2 secreting CAFs have also been shown to enhance chemoresistance in a paracrine manner by upregulating glioma-associated oncogene family zinc finger 2 (GLI2) expression in CRC stem cells [126][49]. These CAFs may serve as an important target for prognostic applications, since high expression levels of TGF-β2 were strongly associated with an increased risk of relapse in chemotherapy-treated CRC patients [126][49]. In addition, therapeutic targeting of these CAFs may also become feasible in the (near) future, with several TGF-β inhibitors already being tested in clinical trials [127,128,129,130][63][64][65][66]. Next to supporting therapy resistance, certain CAFs in advanced CRCs can actively contribute to immune evasion of tumor cells (e.g., via aberrant TGF-β signaling [131][67]) and inhibition of anti-tumor immune responses [132,133,134,135][68][69][70][71]. Li et al. reported that CAFs expressing C-X-C motif chemokine 5 (CXCL5) can promote expression of programmed death-ligand 1 (PD-L1) [136][50], an important suppressor of T-cell activity [137][72]. The fact that CAFs are able to produce a defense against T-cells could have important implications for immunotherapies against CRC [132,133,134,135][68][69][70][71]. Moreover, CD70-expressing CAFs were shown to increase the survival of naturally occurring regulatory T-cells [138][51], which are key mediators of immunosuppression [139][73]. CD73-expressing CAFs were also able to enhance immune suppression via adenosine receptor 2A signaling [140][52]. Notably, the abundance of CD70- and CD73-expressing CAFs was significantly associated with a worse prognosis in CRC patients [138,140[51][52][74],141], thereby emphasizing the involvement of immunomodulatory CAFs in CRC progression.

2.3. Metastasis

Currently, metastatic disease remains the major cause of death in CRC patients [153][75]. CAFs can play an important role in mediating CRC metastasis, as summarized by Tommelein et al. [154][76]. Recent work from our group identified several metastasis-promoting CAFs present at the invasive front of primary CRCs, which were also significantly correlated with poor metastasis-free patient survival. These include CAFs which express endoglin [53] [150] or BMP2 [151][54]. Interestingly, BMP2 expression in CAFs seemed to be regulated by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a cytokine which is overexpressed in CRC cells with a deficiency for mothers against decapentaplegic homolog 4 (SMAD4) [151][54]. These findings provide further support for the idea that the interaction between CRC cells (with a certain mutational status) and CAFs could eventually progress to a tumor-promoting reciprocal loop [155,156,157,158][77][78][79][80]. Other examples of metastasis-promoting CAF populations include IL-11 secreting CAFs, which could activate apoptosis-suppressing programs in metastatic tumor cells [63][55], and stanniocalcin-1 (STC1) expressing CAFs [152][56]. Lastly, next to CAFs in the primary tumor, several studies have suggested that CAFs at the metastatic site, which could originate from resident fibroblasts in remote organs [81][82][83] [159,160,161] or CAFs co-travelling with tumor cells [162[84][85],163], can also contribute to CRC metastasis. For instance, resident liver and lung fibroblasts can induce formation of the pre-metastatic niche via (primary) tumor cell-driven upregulation of pro-inflammatory cytokines such as IL-6 and IL-8 [159,160,161] [81][82][83]. Studies on circulating CAFs originating from primary CRCs have not been conducted yet, but evidence from other cancer types suggests that circulating CAFs may be importantly involved in tumor metastasis and, thus, may serve as useful prognosticators [164][86]. For breast and prostate cancer, it has been shown that circulating CAFs are predominantly found in patients with metastatic disease, and rarely in patients with localized cancer [165,166][87][88].

3. Conclusions

In brief, various tumor-promoting functions (e.g., enhancing tumor invasion, therapy resistance, or metastasis) have been linked to markers expressed by certain CAF populations in advanced CRC. Some of them also seem to serve as promising targets for prognostic or therapeutic applications. However, clinical translation of these findings should be cautiously considered, since it is yet unknown whether these CAF markers demarcate functionally distinct subsets or show overlap with other tumor-promoting or -suppressing CAFs. To enable more precise identification and targeting of “unfavorable” CAFs in advanced CRC, attention needs to be paid to comprehensive characterization and reporting of the subsets being investigated in CAF studies.

 

References

  1. Arnold, M.; Abnet, C.C.; Neale, R.E.; Vignat, J.; Giovannucci, E.L.; McGlynn, K.A.; Bray, F. Global Burden of 5 Major Types of Gastrointestinal Cancer. Gastroenterology 2020, 159, 335–349.e15.
  2. Arnold, M.; Sierra, M.S.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global patterns and trends in colorectal cancer incidence and mortality. Gut 2017, 66, 683–691.
  3. Kuipers, E.J.; Grady, W.M.; Lieberman, D.; Seufferlein, T.; Sung, J.J.; Boelens, P.G.; van de Velde, C.J.; Watanabe, T. Colorectal cancer. Nat. Rev. Dis. Primers 2015, 1, 15065.
  4. Fearon, E.R.; Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 1990, 61, 759–767.
  5. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598.
  6. 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.
  7. Mesker, W.E.; Junggeburt, J.M.C.; Szuhai, K.; De Heer, P.; Morreau, H.; Tanke, H.J.; Tollenaar, R.A. The Carcinoma–Stromal Ratio of Colon Carcinoma Is an Independent Factor for Survival Compared to Lymph Node Status and Tumor Stage. Cell. Oncol. 2007, 29, 387–398.
  8. Huijbers, A.; Tollenaar, R.A.; Pelt, G.W.V.; Zeestraten, E.C.M.; Dutton, S.; McConkey, C.C.; Domingo, E.; Smit, V.T.H.B.M.; Midgley, R.; Warren, B.F.; et al. The proportion of tumor-stroma as a strong prognosticator for stage II and III colon cancer patients: Validation in the VICTOR trial. Ann. Oncol. 2013, 24, 179–185.
  9. Guinney, J.; Dienstmann, R.; Wang, X.; De Reyniès, A.; Schlicker, A.; Soneson, C.; Marisa, L.; Roepman, P.; Nyamundanda, G.; Angelino, P.; et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 2015, 21, 1350–1356.
  10. Calon, A.; Lonardo, E.; Berenguer-Llergo, A.; Espinet, E.; Hernando-Momblona, X.; Iglesias, M.; Sevillano, M.; Palomo-Ponce, S.; Tauriello, D.V.; Byrom, D.; et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat. Genet. 2015, 47, 320–329.
  11. Isella, C.; Terrasi, A.; Bellomo, S.E.; Petti, C.; Galatola, G.; Muratore, A.; Mellano, A.; Senetta, R.; Cassenti, A.; Sonetto, C.; et al. Stromal contribution to the colorectal cancer transcriptome. Nat. Genet. 2015, 47, 312–319.
  12. Dunne, P.D.; McArt, D.G.; Bradley, C.A.; O’Reilly, P.G.; Barrett, H.L.; Cummins, R.; O’Grady, A.; Arthur, K.; Loughrey, M.B.; Allen, W.L.; et al. Challenging the Cancer Molecular Stratification Dogma: Intratumoral Heterogeneity Undermines Consensus Molecular Subtypes and Potential Diagnostic Value in Colorectal Cancer. Clin. Cancer Res. 2016, 22, 4095–4104.
  13. Sahai, E.; Astsaturov, I.; Cukierman, E.; DeNardo, D.G.; Egeblad, M.; Evans, R.M.; Fearon, D.; Greten, F.R.; Hingorani, S.R.; Hunter, T.; et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 2020, 20, 174–186.
  14. Gieniec, K.A.; Butler, L.M.; Worthley, D.L.; Woods, S.L. Cancer-associated fibroblasts—heroes or villains? Br. J. Cancer 2019, 121, 293–302.
  15. Herrera, M.; Islam, A.B.; Herrera, A.; Martín, P.; García, V.; Silva, J.; Garcia, J.M.; Salas, C.; Casal, I.; De Herreros, A.G.; et al. Functional Heterogeneity of Cancer-Associated Fibroblasts from Human Colon Tumors Shows Specific Prognostic Gene Expression Signature. Clin. Cancer Res. 2013, 19, 5914–5926.
  16. Mrazek, A.A.; Carmical, J.R.; Wood, T.G.; Hellmich, M.R.; Eltorky, M.; Bohanon, F.J.; Chao, C. Colorectal Cancer-Associated Fibroblasts are Genotypically Distinct. Curr. Cancer Ther. Rev. 2014, 10, 97–218.
  17. Nishishita, R.; Morohashi, S.; Seino, H.; Wu, Y.; Yoshizawa, T.; Haga, T.; Saito, K.; Hakamada, K.; Fukuda, S.; Kijima, H. Expression of cancer-associated fibroblast markers in advanced colorectal cancer. Oncol. Lett. 2018, 15, 6195–6202.
  18. Son, G.M.; Kwon, M.-S.; Shin, D.-H.; Shin, N.; Ryu, D.; Kang, C.-D. Comparisons of cancer-associated fibroblasts in the intratumoral stroma and invasive front in colorectal cancer. Medicine 2019, 98, e15164.
  19. Berdiel-Acer, M.; Sanz-Pamplona, R.; Calon, A.; Cuadras, D.; Berenguer, A.; Sanjuan, X.; Paules, M.J.; Salazar, R.; Moreno, V.; Batlle, E.; et al. Differences between CAFs and their paired NCF from adjacent colonic mucosa reveal functional heterogeneity of CAFs, providing prognostic information. Mol. Oncol. 2014, 8, 1290–1305.
  20. Li, H.; Courtois, E.T.; Sengupta, D.; Tan, Y.; Chen, K.H.; Goh, J.J.L.; Kong, S.L.; Chua, C.; Hon, L.K.; Tan, W.S.; et al. Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors. Nat. Genet. 2017, 49, 708–718.
  21. Musa, M.; Ali, A. Cancer-associated fibroblasts of colorectal cancer and their markers: Updates, challenges and translational outlook. Futur. Oncol. 2020, 16, 2329–2344.
  22. Brennen, W.N.; Isaacs, J.T.; Denmeade, S.R. Rationale behind targeting fibroblast activation protein-expressing carcino-ma-associated fibroblasts as a novel chemotherapeutic strategy. Mol. Cancer Ther. 2012, 11, 257–266.
  23. Kakarla, S.; Song, X.-T.; Gottschalk, S. Cancer-associated fibroblasts as targets for immunotherapy. Immunotherapy 2012, 4, 1129–1138.
  24. Gu, C.; Lu, H.; Qian, Z. Matrine reduces the secretion of exosomal circSLC7A6 from cancer-associated fibroblast to inhibit tu-morigenesis of colorectal cancer by regulating CXCR5. Biochem. Biophys. Res. Commun. 2020, 527, 638–645.
  25. Omoto, T.; Kim-Kaneyama, J.R.; Lei, X.F.; Orimo, A.; Ohnishi, K.; Yoshihara, K.; Miyauchi, A.; Li, S.; Gao, L.; Umemoto, T.; et al. The im-pact of stromal Hic-5 on the tumorigenesis of colorectal cancer through lysyl oxidase induction and stromal remodeling. Oncogene 2018, 37, 1205–1219.
  26. Zhu, M.; Zhu, Y.; Lance, P. TNF α-activated stromal COX -2 signalling promotes proliferative and invasive potential of colon cancer epithelial cells. Cell Prolif. 2013, 46, 374–381.
  27. Zhu, Y.; Zhu, M.; Lance, P. Stromal COX-2 signaling activated by deoxycholic acid mediates proliferation and invasiveness of colorectal epithelial cancer cells. Biochem. Biophys. Res. Commun. 2012, 425, 607–612.
  28. Herrera, A.; Herrera, M.; Castellon, L.A.; Silva, J.; García, V.; Loubat-Casanovas, J.; Álvarez-Cienfuegos, A.; García, J.M.; Rodríguez, R.; Gil, B.; et al. Protumorigenic effects of Snail-expression fibroblasts on colon cancer cells. Int. J. Cancer 2014, 134, 2984–2990.
  29. Francí, C.; Gallén, M.; Alameda, F.; Baró, T.; Iglesias, M.; Virtanen, I.; De Herreros, A.G. Snail1 Protein in the Stroma as a New Putative Prognosis Marker for Colon Tumours. PLoS ONE 2009, 4, e5595.
  30. 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.
  31. Aizawa, T.; Karasawa, H.; Funayama, R.; Shirota, M.; Suzuki, T.; Maeda, S.; Suzuki, H.; Yamamura, A.; Naitoh, T.; Nakayama, K.; et al. Cancer-associated fibroblasts secrete Wnt2 to promote cancer progression in colorectal cancer. Cancer Med. 2019, 8, 6370–6382.
  32. Kramer, N.; Schmöllerl, J.; Unger, C.; Nivarthi, H.; Rudisch, A.; Unterleuthner, D.; Scherzer, M.; Riedl, A.; Artaker, M.; Crncec, I.; et al. Autocrine WNT2 signaling in fibroblasts promotes colorectal cancer progression. Oncogene 2017, 36, 5460–5472.
  33. Unterleuthner, D.; Neuhold, P.; Schwarz, K.; Janker, L.; Neuditschko, B.; Nivarthi, H.; Crncec, I.; Kramer, N.; Unger, C.; Hengstschläger, M.; et al. Cancer-associated fibroblast-derived WNT2 increases tumor angiogenesis in colon cancer. Angiogenesis 2020, 23, 159–177.
  34. Bhome, R.; Goh, R.W.; Bullock, M.D.; Pillar, N.; Thirdborough, S.M.; Mellone, M.; Mirnezami, R.; Galea, D.; Veselkov, K.; Gu, Q.; et al. Exosomal microRNAs derived from colorectal cancer-associated fibroblasts: Role in driving cancer progression. Aging 2017, 9, 2666–2694.
  35. Bullock, M.D.; Pickard, K.M.; Nielsen, B.S.; Sayan, A.E.; Jenei, V.; Mellone, M.; Mitter, R.; Primrose, J.N.; Thomas, G.J.; Packham, G.K.; et al. Pleiotropic actions of miR-21 highlight the critical role of deregulated stromal microRNAs during colorectal cancer progression. Cell Death Dis. 2013, 4, e684.
  36. Guraya, S.Y. Prognostic significance of circulating microRNA-21 expression in esophageal, pancreatic and colorectal cancers; a systematic review and meta-analysis. Int. J. Surg. 2018, 60, 41–47.
  37. Cui, L.; Ohuchida, K.; Mizumoto, K.; Moriyama, T.; Onimaru, M.; Nakata, K.; Nabae, T.; Ueki, T.; Sato, N.; Tominaga, Y.; et al. Prospec-tively isolated cancer-associated CD10(+) fibroblasts have stronger interactions with CD133(+) colon cancer cells than with CD133(-) cancer cells. PLoS ONE 2010, 5, e12121.
  38. Zhu, H.F.; Zhang, X.H.; Gu, C.S.; Zhong, Y.; Long, T.; Ma, Y.D.; Hu, Z.Y.; Li, Z.G.; Wang, X.Y. Cancer-associated fibroblasts promote colo-rectal cancer progression by secreting CLEC3B. Cancer Biol. Ther. 2019, 20, 967–978.
  39. Yamanashi, T.; Nakanishi, Y.; Fujii, G.; Akishima-Fukasawa, Y.; Moriya, Y.; Kanai, Y.; Watanabe, M.; Hirohashi, S. Podoplanin expres-sion identified in stromal fibroblasts as a favorable prognostic marker in patients with colorectal carcinoma. Oncology 2009, 77, 53–62.
  40. Choi, S.-Y.; Sung, R.; Lee, S.-J.; Lee, T.-G.; Kim, N.; Yoon, S.M.; Lee, E.J.; Chae, H.B.; Youn, S.J.; Park, S.M. Podoplanin, α-Smooth Muscle Actin or S100A4 Expressing Cancer-Associated Fibroblasts Are Associated with Different Prognosis in Colorectal Cancers. J. Korean Med. Sci. 2013, 28, 1293–1301.
  41. Drev, D.; Harpain, F.; Beer, A.; Stift, A.; Gruber, E.S.; Klimpfinger, M.; Thalhammer, S.; Reti, A.; Kenner, L.; Bergmann, M.; et al. Impact of Fibroblast-Derived SPARC on Invasiveness of Colorectal Cancer Cells. Cancers 2019, 11, 1421.
  42. Unger, C.; Kramer, N.; Unterleuthner, D.; Scherzer, M.; Burian, A.; Rudisch, A.; Stadler, M.; Schlederer, M.; Lenhardt, D.; Riedl, A.; et al. Stromal-derived IGF2 promotes colon cancer progression via paracrine and autocrine mechanisms. Oncogene 2017, 36, 5341–5355.
  43. Yang, T.; Zhiheng, H.; Zhanhuai, W.; Qian, X.; Yue, L.; Xiaoxu, G.; Jingsun, W.; Shu, Z.; Ding, K. Increased RAB31 Expression in Cancer-Associated Fibroblasts Promotes Colon Cancer Progression through HGF-MET Signaling. Front. Oncol. 2020, 10, 1747.
  44. Song, J.; Chen, W.; Cui, X.; Huang, Z.; Wen, D.; Yang, Y.; Yu, W.; Cui, L.; Liu, C.Y. CCBE1 promotes tumor lymphangiogenesis and is negatively regulated by TGFbeta signaling in colorectal cancer. Theranostics 2020, 10, 2327–2341.
  45. Yang, X.; Xu, X.; Zhu, J.; Zhang, S.; Wu, Y.; Wu, Y.; Zhao, K.; Xing, C.; Cao, J.; Zhu, H.; et al. miR-31 affects colorectal cancer cells by inhibiting autophagy in cancer-associated fibroblasts. Oncotarget 2016, 7, 79617–79628.
  46. 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 can-cer-associated fibroblasts confer radioresistance in colorectal cancer cells by downregulating FOXA1 and upregulating TGFB3. J. Exp. Clin. Cancer Res. 2020, 39, 65.
  47. Deng, X.; Ruan, H.; Zhang, X.; Xu, X.; Zhu, Y.; Peng, H.; Zhang, X.; Kong, F.; Guan, M. Long noncoding RNA CCAL transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells. Int. J. Cancer 2020, 146, 1700–1716.
  48. Ren, J.; Ding, L.; Zhang, D.; Shi, G.; Xu, Q.; Shen, S.; Wang, Y.; Wang, T.; Hou, Y. Carcinoma-associated fibroblasts promote the stem-ness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics 2018, 8, 3932–3948.
  49. Tang, Y.A.; Chen, Y.F.; Bao, Y.; Mahara, S.; Yatim, S.; Oguz, G.; Lee, P.L.; Feng, M.; Cai, Y.; Tan, E.Y.; et al. Hypoxic tumor microenviron-ment activates GLI2 via HIF-1alpha and TGF-beta2 to promote chemoresistance in colorectal cancer. Proc. Natl. Acad. Sci. USA 2018, 115, E5990–E5999.
  50. Li, Z.; Zhou, J.; Zhang, J.; Li, S.; Wang, H.; Du, J. Cancer-associated fibroblasts promote PD-L1 expression in mice cancer cells via secreting CXCL5. Int. J. Cancer 2019, 145, 1946–1957.
  51. Jacobs, J.; Deschoolmeester, V.; Zwaenepoel, K.; Flieswasser, T.; Deben, C.; Bossche, J.V.D.; Hermans, C.; Rolfo, C.; Peeters, M.; De Wever, O.; et al. Unveiling a CD70-positive subset of cancer-associated fibroblasts marked by pro-migratory activity and thriving regulatory T cell accumulation. OncoImmunology 2018, 7, e1440167.
  52. Yu, M.; Guo, G.; Huang, L.; Deng, L.; Chang, C.S.; Achyut, B.R.; Canning, M.; Xu, N.; Arbab, A.S.; Bollag, R.J.; et al. CD73 on can-cer-associated fibroblasts enhanced by the A2B-mediated feedforward circuit enforces an immune checkpoint. Nat. Commun. 2020, 11, 515.
  53. Paauwe, M.; Schoonderwoerd, M.J.A.; Helderman, R.; Harryvan, T.J.; Groenewoud, A.; van Pelt, G.W.; Bor, R.; Hemmer, D.M.; Versteeg, H.H.; Snaar-Jagalska, B.E.; et al. Endoglin Expression on Cancer-Associated Fibroblasts Regulates Invasion and Stimulates Col-orectal Cancer Metastasis. Clin. Cancer Res. 2018, 24, 6331–6344.
  54. Ouahoud, S.; Voorneveld, P.W.; van der Burg, L.R.A.; de Jonge-Muller, E.S.M.; Schoonderwoerd, M.J.A.; Paauwe, M.; de Vos, T.; de Wit, S.; van Pelt, G.W.; Mesker, W.E.; et al. Bidirectional tumor/stroma crosstalk promotes metastasis in mesenchymal colorectal cancer. Oncogene 2020, 39, 2453–2466.
  55. Calon, A.; Espinet, E.; Palomo-Ponce, S.; Tauriello, D.V.; Iglesias, M.; Cespedes, M.V.; Sevillano, M.; Nadal, C.; Jung, P.; Zhang, X.H.; et al. Dependency of colorectal cancer on a TGF-beta-driven program in stromal cells for metastasis initiation. Cancer Cell 2012, 22, 571–584.
  56. Peña, C.; Céspedes, M.V.; Lindh, M.B.; Kiflemariam, S.; Mezheyeuski, A.; Edqvist, P.-H.; Hägglöf, C.; Birgisson, H.; Bojmar, L.; Jirström, K.; et al. STC1 Expression By Cancer-Associated Fibroblasts Drives Metastasis of Colorectal Cancer. Cancer Res. 2013, 73, 1287–1297.
  57. 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.
  58. Santi, A.; Kugeratski, F.G.; Zanivan, S. Cancer Associated Fibroblasts: The Architects of Stroma Remodeling. Proteomics 2018, 18, e1700167.
  59. De Palma, M.; Biziato, D.; Petrova, T.V. Microenvironmental regulation of tumour angiogenesis. Nat. Rev. Cancer 2017, 17, 457–474.
  60. Henriksson, M.L.; Edin, S.; Dahlin, A.M.; Oldenborg, P.-A.; Öberg, Å.; Van Guelpen, B.; Rutegård, J.; Stenling, R.; Palmqvist, R. Colorectal Cancer Cells Activate Adjacent Fibroblasts Resulting in FGF1/FGFR3 Signaling and Increased Invasion. Am. J. Pathol. 2011, 178, 1387–1394.
  61. Bu, L.; Baba, H.; Yasuda, T.; Uchihara, T.; Ishimoto, T. Functional diversity of cancer-associated fibroblasts in modulating drug resistance. Cancer Sci. 2020, 111, 3468–3477.
  62. Goncalves-Ribeiro, S.; Diaz-Maroto, N.G.; Berdiel-Acer, M.; Soriano, A.; Guardiola, J.; Martinez-Villacampa, M.; Salazar, R.; Capella, G.; Villanueva, A.; Martinez-Balibrea, E.; et al. Carcinoma-associated fibroblasts affect sensitivity to oxaliplatin and 5FU in colo-rectal cancer cells. Oncotarget 2016, 7, 59766–59780.
  63. Huynh, L.K.; Hipolito, C.J.; Ten Dijke, P. A Perspective on the Development of TGF-beta Inhibitors for Cancer Treatment. Biomolecules 2019, 9, 743.
  64. Ungefroren, H. Blockade of TGF-beta signaling: A potential target for cancer immunotherapy? Expert Opin. Ther. Targets 2019, 23, 679–693.
  65. Villalba, M.; Evans, S.R.; Vidal-Vanaclocha, F.; Calvo, A. Role of TGF-beta in metastatic colon cancer: It is finally time for targeted therapy. Cell Tissue Res. 2017, 370, 29–39.
  66. Hawinkels, L.J.; Ten Dijke, P. Exploring anti-TGF-beta therapies in cancer and fibrosis. Growth Factors 2011, 29, 140–152.
  67. Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Canellas, A.; Hernando-Momblona, X.; et al. TGFbeta drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543.
  68. Barrett, R.; Puré, E. Cancer-associated fibroblasts: Key determinants of tumor immunity and immunotherapy. Curr. Opin. Immunol. 2020, 64, 80–87.
  69. Harryvan, T.J.; Verdegaal, E.M.E.; Hardwick, J.C.H.; Hawinkels, L.; van der Burg, S.H. Targeting of the Cancer-Associated Fibro-blast-T-Cell Axis in Solid Malignancies. J. Clin. Med. 2019, 8, 1989.
  70. Mhaidly, R.; Mechta-Grigoriou, F. Fibroblast heterogeneity in tumor micro-environment: Role in immunosuppression and new therapies. Semin. Immunol. 2020, 48, 101417.
  71. Monteran, L.; Erez, N. The Dark Side of Fibroblasts: Cancer-Associated Fibroblasts as Mediators of Immunosuppression in the Tumor Microenvironment. Front. Immunol. 2019, 10, 1835.
  72. Payandeh, Z.; Khalili, S.; Somi, M.H.; Mard-Soltani, M.; Baghbanzadeh, A.; Hajiasgharzadeh, K.; Samadi, N.; Baradaran, B. PD-1/PD-L1-dependent immune response in colorectal cancer. J. Cell. Physiol. 2020, 235, 5461–5475.
  73. Schmetterer, K.G.; Neunkirchner, A.; Pickl, W.F. Naturally occurring regulatory T cells: Markers, mechanisms, and manipulation. FASEB J. 2012, 26, 2253–2276.
  74. Inoue, S.; Ito, H.; Tsunoda, T.; Murakami, H.; Ebi, M.; Ogasawara, N.; Kasugai, K.; Kasai, K.; Ikeda, H.; Inaguma, S. CD70 expression in tumor-associated fibroblasts predicts worse survival in colorectal cancer patients. Virchows Arch. 2019, 475, 425–434.
  75. Dekker, E.; Tanis, P.J.; Vleugels, J.L.A.; Kasi, P.M.; Wallace, M.B. Colorectal cancer. Lancet 2019, 394, 1467–1480.
  76. Tommelein, J.; Verset, L.; Boterberg, T.; Demetter, P.; Bracke, M.; De Wever, O. Cancer-associated fibroblasts connect metasta-sis-promoting communication in colorectal cancer. Front. Oncol. 2015, 5, 63.
  77. Hawinkels, L.J.; Paauwe, M.; Verspaget, H.W.; Wiercinska, E.; van der Zon, J.M.; van der Ploeg, K.; Koelink, P.J.; Lindeman, J.H.; Mesker, W.; ten Dijke, P.; et al. Interaction with colon cancer cells hyperactivates TGF-beta signaling in cancer-associated fibroblasts. Oncogene 2014, 33, 97–107.
  78. Guillen Diaz-Maroto, N.; Sanz-Pamplona, R.; Berdiel-Acer, M.; Cimas, F.J.; Garcia, E.; Goncalves-Ribeiro, S.; Albert, N.; Garcia-Vicien, G.; Capella, G.; Moreno, V.; et al. Noncanonical TGFbeta Pathway Relieves the Blockade of IL1beta/TGFbeta-Mediated Crosstalk between Tumor and Stroma: TGFBR1 and TAK1 Inhibition in Colorectal Cancer. Clin. Cancer Res. 2019, 25, 4466–4479.
  79. Rai, A.; Greening, D.W.; Xu, R.; Suwakulsiri, W.; Simpson, R.J. Exosomes Derived from the Human Primary Colorectal Cancer Cell Line SW480 Orchestrate Fibroblast-Led Cancer Invasion. Proteomics 2020, 20, e2000016.
  80. Zhang, Y.; Wang, S.; Lai, Q.; Fang, Y.; Wu, C.; Liu, Y.; Li, Q.; Wang, X.; Gu, C.; Chen, J.; et al. Cancer-associated fibroblasts-derived exo-somal miR-17-5p promotes colorectal cancer aggressive phenotype by initiating a RUNX3/MYC/TGF-beta1 positive feedback loop. Cancer Lett. 2020, 491, 22–35.
  81. Ji, Q.; Zhou, L.; Sui, H.; Yang, L.; Wu, X.; Song, Q.; Jia, R.; Li, R.; Sun, J.; Wang, Z.; et al. Primary tumors release ITGBL1-rich extracellular vesicles to promote distal metastatic tumor growth through fibroblast-niche formation. Nat. Commun. 2020, 11, 1–18.
  82. Mueller, L.; Goumas, F.A.; Affeldt, M.; Sandtner, S.; Gehling, U.M.; Brilloff, S.; Walter, J.; Karnatz, N.; Lamszus, K.; Rogiers, X.; et al. Stromal fibroblasts in colorectal liver metastases originate from resident fibroblasts and generate an inflammatory microenvi-ronment. Am. J. Pathol. 2007, 171, 1608–1618.
  83. Mueller, L.; Von Seggern, L.; Schumacher, J.; Goumas, F.; Wilms, C.; Braun, F.; Broering, D.C. TNF-α similarly induces IL-6 and MCP-1 in fibroblasts from colorectal liver metastases and normal liver fibroblasts. Biochem. Biophys. Res. Commun. 2010, 397, 586–591.
  84. Gonzalez-Zubeldia, I.; Dotor, J.; Redrado, M.; Bleau, A.M.; Manrique, I.; de Aberasturi, A.L.; Villalba, M.; Calvo, A. Co-migration of colon cancer cells and CAFs induced by TGFbeta(1) enhances liver metastasis. Cell Tissue Res. 2015, 359, 829–839.
  85. Zhou, P.; Xiao, N.; Wang, J.; Wang, Z.; Zheng, S.; Shan, S.; Wang, J.; Du, J.; Wang, J. SMC1A recruits tumor-associated-fibroblasts (TAFs) and promotes colorectal cancer metastasis. Cancer Lett. 2017, 385, 39–45.
  86. Herrera, M.; Galindo-Pumariño, C.; García-Barberán, V.; Peña, C. A Snapshot of the Tumor Microenvironment in Colorectal Cancer: The Liquid Biopsy. Int. J. Mol. Sci. 2019, 20, 6016.
  87. Ao, Z.; Shah, S.H.; Machlin, L.M.; Parajuli, R.; Miller, P.C.; Rawal, S.; Williams, A.J.; Cote, R.J.; Lippman, M.E.; Datar, R.H.; et al. Identifica-tion of Cancer-Associated Fibroblasts in Circulating Blood from Patients with Metastatic Breast Cancer. Cancer Res. 2015, 75, 4681–4687.
  88. Jones, M.L.; Siddiqui, J.; Pienta, K.J.; Getzenberg, R.H. Circulating fibroblast-like cells in men with metastatic prostate cancer. Prostate 2013, 73, 176–181.
More
Video Production Service