Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2981 word(s) 2981 2021-07-12 03:53:27 |
2 format correct -21 word(s) 2960 2021-07-19 10:10:43 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Jean, C. Extracellular Matrices and Cancer-Associated Fibroblasts. Encyclopedia. Available online: https://encyclopedia.pub/entry/12167 (accessed on 26 April 2024).
Jean C. Extracellular Matrices and Cancer-Associated Fibroblasts. Encyclopedia. Available at: https://encyclopedia.pub/entry/12167. Accessed April 26, 2024.
Jean, Christine. "Extracellular Matrices and Cancer-Associated Fibroblasts" Encyclopedia, https://encyclopedia.pub/entry/12167 (accessed April 26, 2024).
Jean, C. (2021, July 19). Extracellular Matrices and Cancer-Associated Fibroblasts. In Encyclopedia. https://encyclopedia.pub/entry/12167
Jean, Christine. "Extracellular Matrices and Cancer-Associated Fibroblasts." Encyclopedia. Web. 19 July, 2021.
Extracellular Matrices and Cancer-Associated Fibroblasts
Edit

Solid cancer progression is dictated by neoplastic cell features and pro-tumoral crosstalks with their microenvironment. Stroma modifications, such as fibroblast activation into cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM) remodeling, are now recognized as critical events for cancer progression and as potential therapeutic or diagnostic targets.

cancer-associated fibroblasts extracellular matrix cancer therapeutic and diagnostic targets

1. Introduction

More than a century ago, the “seed and soil” theory was proposed by Paget [1], seed being cancer cells and soil the stroma. Molecular characteristics of “seed (cancer cells)” were analyzed in depth and many oncogenes/suppressor genes have been identified and characterized. However, the “soil”, microenvironment encompassing host stromal cells (vascular cells, fibroblasts, immune/inflammatory cells, etc.), as well as non-cellular components (soluble factors, extracellular matrix, etc.) generated by cancer cells themselves and stromal cells, is still under characterization because of its structural and functional complexity. Recent development of novel molecular technologies has revealed the biomedical significance of the “soil” that influences cancer cell biological behaviors and functions, such as proliferation, invasion and metastatic processes. It is now clear that not only the soil promotes the growth of the seed, but also that the seed “educates” the soil to support its needs. Indeed, stromal cells acquire a specific biological phenotype via direct or indirect interactions with cancer cells. As an example, fibroblasts, which are the major components of the tumor microenvironment in most of solid tumors, become activated into cancer-associated fibroblasts (CAFs) under cancer cell stimulation, and, in turn, favor cancer development [2] notably via their secretion of acellular component such as extracellular matrix (ECM) and soluble factors. This abnormal ECM is a key regulator of tumor survival, progression and chemoresistance and represents the “breeding ground” of cancer cells. Globally, as the soil promotes the acquisition and maintenance of each of the cancer hallmarks, over the years several approaches have been used to target it.

2. Cancer-Associated Fibroblasts: Main Actors of Matrix Remodeling

2.1. Cancer-Associated Fibroblasts (CAFs)

Cancer-associated fibroblasts, the major stromal cells of most solid cancers such as breast and pancreatic cancers, have been widely described as key actors in tumor progression through numerous mechanisms including their ability to secrete various exacerbated soluble and insoluble factors (such as ECM). CAFs are defined as all fibroblastic, non-neoplastic, non-vascular, non-epithelial and non-inflammatory cells, activated and found in tumors and metastatic niches [3][4]. As opposed to the physiological activation (observed during wound healing for example), the “activated” phenotype of CAFs is persistent [3].
CAFs have been proposed to be defined as a cellular state rather than a cell type [5] because CAF origin is diverse: whereas they mainly come from the activation of quiescent fibroblasts residing in the tumor host tissue, they also originate from bone marrow derived cells (BMDC), trans-differentiation of pericytes, endothelial and epithelial cells [6][7]. Although there are currently no specific markers defining completely and exclusively CAFs, vimentin, α-SMA (smooth muscle actin), FAP (Fibroblast activation protein), PDGFR-α (Platelet-derived growth factor receptor-α), PDGFR-β, FSP-1 (also known as S100A4) and PDPN (podoplanin) are markers [3][4][8].
During tumorigenesis, quiescent fibroblasts are activated in response to various stimuli such as hypoxia [9], oxidative stress [10], chemokines and cytokines and growth factors such as transforming growth factor superfamily (TGFβ) [11], platelet-derived growth factors (PDGF) [12], epidermal growth factors (EGF), fibroblast growth factors (FGF) [13] and sonic hedgehog (SHH) [14]. Such activation leads to an increase of CAF contractile capacities (with increased expression of α-SMA and vimentin), to a morphological modification (stellate shaped) [5], and to an exacerbated secretion of many factors (soluble and insoluble factors including ECM proteins). CAFs have been shown to first deposit fibronectin, generating intracellular tension involving actin cables. This creates, in the case of wound healing, a positive feedback loop that keeps the fibroblasts in an activated state in which YAP (Yes-associated protein) is translocated to the nucleus and α–SMA overexpressed [15]. Signals from the neo-synthesized ECM activate the Rho-ROCK-Myosin II signaling pathway and the incorporation of α-SMA into actin-myosin fibers leads to an increased contractility of activated fibroblasts [16][17]. These cells then generate tensile forces which, once transmitted to the matrix, trigger its remodeling at different levels. At the biochemical level, activated fibroblasts modify the matrix molecular composition by increasing the deposition of new matrix components, and by modulating the expression of matrix metalloproteinases (MMPs). Mechanically, these cells affect the physical properties of the matrix by modifying its organization and stiffness [18]. Such modifications induce the recruitment of new fibroblasts and their activation, and other components of ECM are produced thereby increasing the deposition of type I collagen, resulting in a decrease in fibronectin/collagen I ratio. While in a physiological context, when the collagen I network is crosslinked, fibronectin fibers are relaxed and fibroblasts resume their quiescence [15], in a tumor context, the ECM remodeling is continuous [19][20][21], altering the distribution of fibronectin zones by preventing the relaxation of fibronectin fibers [22].
Through these secretions, CAFs maintain their activated status, enhance their number and install a dialogue not only with tumor cells but also with the other stromal cells (endothelial cells, immune cells, for example) leading to complex and finely regulated tumor modifications. Indeed, while in the past researchers believed that CAFs had exclusively pro-tumoral functions (promoting tumor cell proliferation, survival, chemoresistance, angiogenesis [23] and immunosuppression [24]), in the past seven years, several publications have shown that CAF deletion or ECM modifications could result, depending on the context, in enhanced tumor progression [25][26].
One CAF particularity to take into account when trying to understand their role in cancer, is the recent observation, based on single cell RNA seq analysis, that CAFs are heterogeneous, in terms of morphology, functions and markers. This emergent concept of CAF subpopulations is based on several recent publications reporting the presence of CAF subgroups in PDAC (pancreatic ductal adenocarcinoma), breast carcinoma, colon carcinoma, lung adenocarcinoma and high-grade serous ovarian cancers [27][28] (Table 1). In breast cancer, Costa et al. distinguished four different CAF subpopulations (referred to as CAF-S1 to -S4), that accumulate differentially depending on breast cancer subtypes (luminal, HER2, and triple-negative). Importantly, as authors identified that the CAF-S1 subset highly contributes to immunosuppression, they suggest that patients with CAF-S1 rich tumors may benefit from specific immunotherapeutic strategy [29]. In non-small cell lung cancer, CAF subgrouping, either based on the collagen aspect or on fibroblast density, has prognosis significance [30][31][32]. In PDAC, David Tuveson’s team was the first to report the presence of diverse CAF subtypes: a CAF subpopulation with elevated expression of αSMA located immediately adjacent to neoplastic cells called “myofibroblastic CAFs” (myCAF), and another CAF subpopulation located more distantly from neoplastic cells (lacking elevated αSMA expression) which secreted IL6 and additional inflammatory mediators called “inflammatory CAFs” (iCAF) [33]. Later on, by performing single cell analysis, this same team also identified a third subpopulation named “antigen-presenting CAFs” (ApCAFs) capable of activating CD4+ T cells in an antigen-specific manner, thus with putative immune-modulatory capacity [34].
Over the years, several studies have looked for correlation between CAFs/ECM and PDAC patient prognosis. Erkan et al. showed in a cohort of 233 PDAC patients (who underwent surgical resection and received adjuvant therapy) that collagen deposition was an independent positive prognosis marker whereas the amount of α-SMA expression (CAF activity) was negatively correlated with patient survival, although statistically insignificant [35]. In this study, they defined four major patterns of collagen deposition with regard to PSC (pancreatic stellate cells), the main cell origin of CAFs in PDAC [3][36][37] activity, and they showed that the combination of high stromal activity and low collagen deposition was associated with worse prognosis, whereas the combination of high collagen deposition and low stromal activity was correlated with a better prognosis. Although these results are contradictory to the dogma that collagen-induced signals favor tumor aggressiveness [38][39], they have been confirmed in another publication. Indeed, Bever et al., using computer-aided quantitative method to correlate patient survival with stroma density and activity in pancreatic cancer, observed that high stromal density (ratio of the stroma area to total tumor area), but not stroma activity (measured by α-SMA expression), was significantly associated with longer disease-free survival (DFS) and overall survival (OS) in a cohort of 66 PDAC patients (who underwent pancreaticoduodenectomy and received adjuvant therapy) [40]. This year, R. Kalluri’s group reported, by using a dual-recombinase (integrating the capacity to manipulate genes using both the Cre-loxP and Flp-FRT) genetic mouse model of spontaneous PDAC to delete type 1 collagen specifically in myofibroblasts, that reducing Col1 total stromal content accelerates PanINs (pancreatic intraepithelial neoplasia) and PDAC emergence and decreases mouse overall survival by the establishment of an immunosuppressive microenvironment (recruitment of myeloid-derived suppressor cells) [41]. In contrary, Weaver’s group observed no significant association between the levels of fibrillary collagens and patient survival. The authors demonstrated, using second harmonic generation (SHG) microscopy, that the diameter of the collagen fibers adjacent to the pancreatic lesions was significantly thicker in PDAC patients with the shortest survival suggesting that collagen thickness is indicative of poor prognosis [42]. Following these findings, another group showed, using a SHG-based quantitative approach, that PDAC patients with high collagen alignment (induced by collagen crosslinking) had significantly reduced overall survival compared to patients with low alignment [43]. Then, according to these publications, collagen quality (thickness, alignment) rather than quantity is predictive of poor prognosis; therefore, not only CAF activation is important but also ECM modifications. This concept is supported by Moffitt’s studies that defined, based on PDAC virtual microdissection, “normal” and “activated” stromal subtypes with prognostic and biological relevance; “activated” subtype, corresponding to a stroma encompassing activated CAFs and highly remodeled ECM, has the worse prognosis [44].
Table 1. CAF subgrouping in breast, ovarian, pancreatic, lung and colorectal cancers associated with identified secretions, main tumoral characteristics and markers. Italic refers to genes. Abbreviations: Acta2: Actin Alpha 2, Smooth Muscle; C7: Complement C7; CAV: Caveolin; CCL: C-C Motif Chemokine Ligand; CD74: Cluster of Differentiation 74; Cdh11: Cadherin 11; Clec3b: C-Type Lectin Domain Family 3 Member B; CMH: major histocompatibility complex; Col: collagen; CXCL: chemokine (C-X-C motif) ligand; ENG: Endoglin; FAP: Fibroblast Activation Protein Alpha; FSP1: Fibroblast-Specific Protein-1 = S100A4: S100 Calcium Binding Protein A4; Gas6: Growth Arrest Specific 6; Gpm6a: Glycoprotein M6A; Gsn: Gelsolin; H2-Ab1: histocompatibility 2, class II antigen A, beta 1; HAS1: Hyaluronan Synthase 1; HLA-DRA: Major Histocompatibility Complex, Class II, DR Alpha; Igf: Insulin Like Growth Factor; Irf5: Interferon regulatory factor 5; ITGA11: Integrin Subunit Alpha 11; Lgals7: Galectin 7; LRRC15: Leucine Rich Repeat Containing 15; Lrrn4: Leucine Rich Repeat Neuronal 4; Ly6c1: Lymphocyte antigen 6C1 precursor; Msln: Mesothelin; MYH11: myosin heavy chain 11; MYL9: myosin light chain 9; Nkain4: Sodium/Potassium Transporting ATPase Interacting 4; PDGFR: Platelet Derived Growth Factor Receptor Alpha; PDPN: Podoplanin; POSTN: Periostin; Ptn: Pleiotrophin; Saa3: Serum Amyloid A3Slpi; Tagln: Transgelin; TGFb: Transforming Growth Factor Beta 1; Thy1: Thy-1 Cell Surface Antigen; TIMP1: Tissue Inhibitor of Metalloproteinase 1; Tnc: Tenascin C.
Cancer CAF Subpopulations Secretion Main Characteristics Markers/Key Genes
Breast cancer [29][45] and high-grade serous ovarian cancers [28] CAF-S1 CXCL12, CCL2, CCL11, CXCL14 [28][29][45] - Attract CD4+CD25+ T lymphocytes, promote their differentiation into Tregs and subsequent pro-tumoral functions [29]
- Enhance cancer cell migration [45]
- Initiate an epithelial-to-mesenchymal transition (EMT) [45]
CD29MedFAPHi FSP1Low-HiαSMAHi PDGFRβMed-Hi CAV1Low [28][29]
CAF-S2 ND Inactivated CAF [45] CD29LowFAPNeg FSP1Neg-LowαSMANeg PDGFRβNeg CAV1Neg [28][29]
CAF-S3 ND Inactivated CAF [45] CD29MedFAPNeg FSP1Med-HiαSMANeg-Low PDGFRβMed CAV1Neg-Low [28][29]
CAF-S4 CCL2, CCL11, CXCL12, CXCL13, CXCL14 [28][29][45] Induce cancer cell invasion via NOTCH signaling [45] CD29HiFAPNeg FSP1Low-MedαSMAHi PDGFRβLow-Med CAV1Neg-Low [28][29]
PDAC [8][33][34][46][47] Myofibroblastic CAFs (myCAFs) [33][34] ECM proteins Anti-tumor, contractile, stroma-remodeling FAP+ αSMAhigh IL-6low
Tnc, Tgfb1, Thy1, Tagln, Col12a1, Pdgfrb
Inflammatory CAFs (iCAFs) [33][34] IL-6, IL-11, LIF IL-8, CXCL1-2-12, CXCL2, CCL2 Pro-tumor, secrete cytokines and chemokines involved in cancer progression αSMAlow IL-6high
Clec3b, Col14a1, Gsn, Ly6c1, Cxcl12
Antigen-presenting CAFs (ApCAFs) [34] ND Present antigen to T cells CD74, Saa3, Slpi, H2-Ab1, Nkain4, Irf5, CMH class II
FB1 = iCAF like [46] Il-6, CXCL12, CCL2, CCL7 Secretory phenotype Cxcl14, Ptn, and genes mediating insulin-like growth factor signaling (Igf1, Igfbp7, Igfbp4), Pdgfrα
FB3 = myCAF like [46]   Contractile phenotype mesothelial markers (Lrrn4, Gpm6a, Nkain4, Lgals7, and Msln); fibroblast markers (Cav1, Cdh11, and Gas6), Acta2 and Tagln, MHC-II–associated genes
CAF-c1 [47] Collagen I, SPARC, ECM proteins Early tumors CD74+/HLA-DRAlo/Col1a1+/Col3a1+/TIMP1+/FAP+, C7+/ENG+
CAF-c2 = IL1-CAF [47] Il1 Established tumors HAS1+/CXCL1+/CCL2+/FAP+/CD74hi/HLA-DRA+
CAF-c0 = TGFβ-CAF [47] TGFβ Established tumors LRRC15+/TAGLN+/Col11a+/ACTA2+/FAP+/CD74hi/HLA-DRA+
Subtype A [8] ECM proteins Associated with poor/intermediate prognosis POSTNhigh/MYH11low/PDPNlow/αSMA low/PDGFRα/Vimentinlow
Subtype B [8] ECM proteins Associated with intermediate prognosis and with cancer cell protection against gemcitabine MYH11high/POSTNlow/high/PDGFRα/αSMAhigh/Vimentinhigh
Subtype C [8] Inflammatory mediators and ECM proteins Associated with “good” prognostic but with cancer cell protection against gemcitabine PDPNhigh/POSTNlow-high/PDGFRα
Subtype D [8] ECM proteins Associated with bad prognosis and with cancer cell protection against gemcitabine αSMAhigh/Vimentinhigh
Non-small cell lung carcinoma [32] High desmoplastic CAFs ND Enhance collagen matrix remodeling, invasion and tumor growth αSMA+ITGA11+
Low desmoplastic CAFs ND Pro-tumoral functions limited compared to HD-CAFs αSMA+ITGA11+
Colorectal cancer [48] PDPN+ CAFs ND Associated with prolonged disease-free survival PDPN+
PDPN/α-SMAhigh CAFs ND Associated with aggressive tumors PDPN/α-SMAhigh
PDPN/S100A4high CAFs ND Associated with tumor budding and lymphovascular invasion PDPN/S100A4high
Melanoma [49] S1 CAFs CXCL12, CSF1, CCL8 Regulate immune cell recruitment PDPNhigh/PDGFRαhigh/CD34high
S2 CAFs ECM proteins Drive desmoplastic reaction PDPNhigh/PDGFRαhigh/CD34low
S3 CAFs ND Regulate actin cytoskeleton and contractility Acta2high/CD34low

2.2. Extracellular Matrix (ECM)

One of the major features of CAFs is their ability to produce large amounts of ECM proteins, such as collagens, glycoproteins and proteoglycans [50]. The development of new technologies, mainly the “matrisome” approach which is based on mass spectrometry analysis of in vivo samples and enables to characterize the ECM biochemical composition, have revolutionized our understanding of tumoral ECM components and roles [51][52]. ECM is a complex scaffold composed of hundreds of proteins that provide anchoring and support to environmental cells under physiological and pathological conditions [51]. Major ECM structural components are collagens, proteoglycans, and hyaluronic acid that provide supportive framework within which other ECM components (such as laminin or fibronectin for example) and cells interact [50][53]. In addition to this architectural role, ECM proteins provide signals that cells interpret and transduce via cell surface receptors such as integrins. These signals, named mechanotransduction, activate cellular pathways that impact cellular functions such as proliferation, survival, morphology, adhesion and motility [54][55][56]. ECM represents also a growth factor reservoir as matrix proteins can sequester them and modify their signaling properties [57]. ECM modifications (biochemical composition, mechanical properties, integrity) are often observed in diseases such as fibrosis, cardiovascular or musculoskeletal diseases [58][59][60] and cancer [61][62]. It has recently become evident that ECM has biomechanical and physical properties that impact all cancer hallmarks, including the cellular processes that contribute to cancer initiation, progression, spreading [20][63] and metastatic niche formation [64][65]. Moreover, the extraordinarily dense fibrotic stroma, found in PDAC (PDAC fibrotic area accounts for up to 90% of the tumor area [66]) and breast cancer, impedes tumor perfusion and delivery of anticancer drugs [67]. ECM modifications within tumor (quantity, stiffness, etc.) have been shown to correlate with more aggressive tumors and worse prognosis for the patient [68][69][70][71].
Collagens are by far the most abundant and best characterized ECM components. Collagen I is responsible for the majority of the desmoplastic reaction [72][73][74], and high levels of its deposition have been associated with reduced survival in PDAC patients [38]. In breast cancer, as in PDAC, accumulation of fibrillary collagens I, III, and V occurs [39][75][76] and increased level of Col1a1 or Col3 is associated with a metastasis status [76] and correlates with shorter survival [77]. Mechanistically, fibrillary collagens regulate tumor cell functions via the integrin activation promoting tumor cell proliferation, migration and preventing apoptosis [78]. In contrast, type IV collagen, which is mainly present in the basement membrane (BM) that underlies epithelium and endothelial cells, is decreased in both PDAC and breast cancer [39]. In PDAC mouse model (KTC: Tgfbr2flox/wt; KrasLSL-G12D/+Tgfbr2flox/wtPtf1a-Cre) [79], as in human PDAC tumors [80], not only Col4 is decreased but also BM proteins in general. This BM destruction facilitates invasion and metastasis in many cancers [63][81]. Interestingly, the tumoral ECM remodeling, involving protease-mediated ECM cleavage, generates ECM fragments, named matrikines or matricryptins, capable of influencing tumor progression and dissemination [60]. Indeed, increased level of MMP-mediated degradation of type I, II, III, and IV collagens release C-terminal collagen domains named C1M, C3M C4M and C4M12a1, respectively, that are often found in PDAC patient serum and are associated with significantly shorter survival [82][83].
Glycoproteins are the second main ECM subgroup deregulated during cancer that encompasses fibronectin, laminins and many other proteins. Fibronectin (FN) is found to be overexpressed in several cancers and reported to participate in several steps of tumorigenesis including growth, invasion, and metastasis. When analyzed as a potential prognosis factor for cancer patients, FN’s role in cancer progression appears to be complex, as FN deposited in tumor microenvironment (TME) or FN tumor cell endogenous expression have opposite correlations with patient prognosis [84][85]. A better understanding of such paradoxical role of FN in tumorigenesis is of high interest and has been recently extensively well reviewed by Tsung-Cheng Lin et al. [85]. In fibrotic solid tumors, FN expression is associated with poor clinical outcome [39], tumor aggressiveness [86], and participates in the resistance to radiotherapy via the FN-specific α5β1-integrin pathway [87]. Other glycoproteins such as periostin and galectin-1 are upregulated in PDAC [88][89] and their expressions are negatively correlated to patient survival [88][89][90]. Numbers of glycoproteins are found to be enhanced in cancer patient serum and used as diagnostic and prognostic biomarkers: for example, CA125 and CA19-9 are two glycoproteins used as ovarian cancer and pancreatic cancer biomarkers, respectively [91].
Finally, proteoglycan expression is also modified during tumorigenesis and one major example is the hyaluronan (HA). HA is a negatively charged glycosaminoglycan found to be highly accumulated in solid cancers [38][92][93]. HA expression within tumor, through a mechanism involving its high hydrophobic properties, enhances interstitial tumor pressure, and its accumulation correlates with poor prognosis [38] and metastasis [94].

References

  1. Paget, S. The distribution of secondary growths in cancer of the breast. Lancet 1889, 133, 571–573.
  2. Socovich, A.M.; Naba, A. The cancer matrisome: From comprehensive characterization to biomarker discovery. Semin. Cell Dev. Biol. 2019, 89, 157–166.
  3. Kalluri, R.; Zeisberg, M. Fibroblasts in cancer. Nat. Rev. Cancer 2006, 6, 392–401.
  4. Öhlund, D.; Elyada, E.; Tuveson, D. Fibroblast heterogeneity in the cancer wound. J. Exp. Med. 2014, 211, 1503–1523.
  5. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598.
  6. Potenta, S.; Zeisberg, E.M.; Kalluri, R. The role of endothelial-to-mesenchymal transition in cancer progression. Br. J. Cancer 2008, 99, 1375–1379.
  7. Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Investig. 2009, 119, 1420–1428.
  8. Neuzillet, C.; Tijeras-Raballand, A.; Ragulan, C.; Cros, J.; Patil, Y.; Martinet, M.; Erkan, M.; Kleeff, J.; Wilson, J.; Apte, M.; et al. Inter- and intra-tumoural heterogeneity in cancer-associated fibroblasts of human pancreatic ductal adenocarcinoma. J. Pathol. 2019, 248, 51–65.
  9. Kugeratski, F.G.; Atkinson, S.J.; Neilson, L.J.; Lilla, S.; Knight, J.R.P.; Serneels, J.; Juin, A.; Ismail, S.; Bryant, D.M.; Markert, E.K.; et al. Hypoxic cancer–associated fibroblasts increase NCBP2-AS2/HIAR to promote endothelial sprouting through enhanced VEGF signaling. Sci. Signal. 2019, 12, eaan8247.
  10. Chan, J.S.K.; Tan, M.J.; Sng, M.K.; Teo, Z.; Phua, T.; Choo, C.C.; Li, L.; Zhu, P.; Tan, N.S. Cancer-associated fibroblasts enact field cancerization by promoting extratumoral oxidative stress. Cell Death Dis. 2018, 8, e2562.
  11. Casey, T.M.; Eneman, J.; Crocker, A.; White, J.; Tessitore, J.; Stanley, M.; Harlow, S.; Bunn, J.Y.; Weaver, D.; Muss, H.; et al. Cancer associated fibroblasts stimulated by transforming growth factor beta1 (TGF-beta 1) increase invasion rate of tumor cells: A population study. Breast Cancer Res. Treat. 2008, 110, 39–49.
  12. Rizvi, S.; Mertens, J.C.; Bronk, S.F.; Hirsova, P.; Dai, H.; Roberts, L.R.; Kaufmann, S.H.; Gores, G.J. Platelet-derived growth factor primes cancer-associated fibroblasts for apoptosis. J. Biol. Chem. 2014, 289, 22835–22849.
  13. Bordignon, P.; Bottoni, G.; Xu, X.; Popescu, A.S.; Truan, Z.; Guenova, E.; Kofler, L.; Jafari, P.; Ostano, P.; Röcken, M.; et al. Dualism of FGF and TGF-β Signaling in Heterogeneous Cancer-Associated Fibroblast Activation with ETV1 as a Critical Determinant. Cell Rep. 2019, 28, 2358–2372.
  14. Yauch, R.L.; Gould, S.E.; Scales, S.J.; Tang, T.; Tian, H.; Ahn, C.P.; Marshall, D.; Fu, L.; Januario, T.; Kallop, D.; et al. A paracrine requirement for hedgehog signalling in cancer. Nature 2008, 455, 406–410.
  15. Kollmannsberger, P.; Bidan, C.M.; Dunlop, J.W.C.; Fratzl, P.; Vogel, V. Tensile forces drive a reversible fibroblast-to-myofibroblast transition during tissue growth in engineered clefts. Sci. Adv. 2018, 4, eaao4881.
  16. Provenzano, P.P.; Keely, P.J. Mechanical signaling through the cytoskeleton regulates cell proliferation by coordinated focal adhesion and Rho GTPase signaling. J. Cell Sci. 2011, 124, 1195–1205.
  17. 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.
  18. Jang, I.; Beningo, K.A. Integrins, CAFs and Mechanical Forces in the Progression of Cancer. Cancers 2019, 11, 721.
  19. Leight, J.L.; Drain, A.P.; Weaver, V.M. Extracellular Matrix Remodeling and Stiffening Modulate Tumor Phenotype and Treatment Response. Annu. Rev. Cancer Biol. 2017, 1, 313–334.
  20. Pickup, M.W.; Mouw, J.K.; Weaver, V.M. The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 2014, 15, 1243–1253.
  21. Cox, T.R.; Erler, J.T. Remodeling and homeostasis of the extracellular matrix: Implications for fibrotic diseases and cancer. Dis. Model. Mech. 2011, 4, 165–178.
  22. Barbazán, J.; Matic Vignjevic, D. Cancer associated fibroblasts: Is the force the path to the dark side? Curr. Opin. Cell Biol. 2019, 56, 71–79.
  23. 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.
  24. Kraman, M.; Bambrough, P.J.; Arnold, J.N.; Roberts, E.W.; Magiera, L.; Jones, J.O.; Gopinathan, A.; Tuveson, D.A.; Fearon, D.T. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science 2010, 330, 827–830.
  25. Amakye, D.; Jagani, Z.; Dorsch, M. Unraveling the therapeutic potential of the Hedgehog pathway in cancer. Nat. Med. 2013, 19, 1410–1422.
  26. Van Cutsem, E.; Tempero, M.A.; Sigal, D.; Oh, D.-Y.; Fazio, N.; Macarulla, T.; Hitre, E.; Hammel, P.; Hendifar, A.E.; Bates, S.E.; et al. Randomized Phase III Trial of Pegvorhyaluronidase Alfa With Nab-Paclitaxel Plus Gemcitabine for Patients With Hyaluronan-High Metastatic Pancreatic Adenocarcinoma. J. Clin. Oncol. 2020, 38, 3185–3194.
  27. 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, 1–13.
  28. 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, 1056.
  29. 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.
  30. Takahashi, Y.; Ishii, G.; Taira, T.; Fujii, S.; Yanagi, S.; Hishida, T.; Yoshida, J.; Nishimura, M.; Nomori, H.; Nagai, K.; et al. Fibrous stroma is associated with poorer prognosis in lung squamous cell carcinoma patients. J. Thorac. Oncol. 2011, 6, 1460–1467.
  31. Minami, Y.; Matsuno, Y.; Iijima, T.; Morishita, Y.; Onizuka, M.; Sakakibara, Y.; Noguchi, M. Prognostication of small-sized primary pulmonary adenocarcinomas by histopathological and karyometric analysis. Lung Cancer 2005, 48, 339–348.
  32. Hao, J.; Zeltz, C.; Pintilie, M.; Li, Q.; Sakashita, S.; Wang, T.; Cabanero, M.; Martins-Filho, S.N.; Wang, D.Y.; Pasko, E.; et al. Characterization of Distinct Populations of Carcinoma-Associated Fibroblasts from Non–Small Cell Lung Carcinoma Reveals a Role for ST8SIA2 in Cancer Cell Invasion. Neoplasia 2019, 21, 482–493.
  33. Öhlund, D.; Handly-Santana, A.; Biffi, G.; Elyada, E.; Almeida, A.S.; Ponz-Sarvise, M.; Corbo, V.; Oni, T.E.; Hearn, S.A.; Lee, E.J.; et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 2017, 214, 579–596.
  34. Elyada, E.; Bolisetty, M.; Laise, P.; Flynn, W.F.; Courtois, E.T.; Burkhart, R.A.; Teinor, J.A.; Belleau, P.; Biffi, G.; Lucito, M.S.; et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 2019, 9, 1102–1123.
  35. Erkan, M.; Michalski, C.W.; Rieder, S.; Reiser-Erkan, C.; Abiatari, I.; Kolb, A.; Giese, N.A.; Esposito, I.; Friess, H.; Kleeff, J. The activated stroma index is a novel and independent prognostic marker in pancreatic ductal adenocarcinoma. Clin. Gastroenterol. Hepatol. 2008, 6, 1155–1161.
  36. Erkan, M.; Reiser-Erkan, C.; Michalski, C.W.; Kong, B.; Esposito, I.; Friess, H.; Kleeff, J. The impact of the activated stroma on pancreatic ductal adenocarcinoma biology and therapy resistance. Curr. Mol. Med. 2012, 12, 288–303.
  37. Erkan, M.; Hausmann, S.; Michalski, C.W.; Fingerle, A.A.; Dobritz, M.; Kleeff, J.; Friess, H. The role of stroma in pancreatic cancer: Diagnostic and therapeutic implications. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 454–467.
  38. Whatcott, C.J.; Diep, C.H.; Jiang, P.; Watanabe, A.; LoBello, J.; Sima, C.; Hostetter, G.; Shepard, H.M.; Von Hoff, D.D.; Han, H. Desmoplasia in Primary Tumors and Metastatic Lesions of Pancreatic Cancer. Clin. Cancer Res. 2015, 21, 3561–3568.
  39. Insua-Rodríguez, J.; Oskarsson, T. The extracellular matrix in breast cancer. Adv. Drug Deliv. Rev. 2016, 97, 41–55.
  40. Bever, K.M.; Sugar, E.A.; Bigelow, E.; Sharma, R.; Laheru, D.; Wolfgang, C.L.; Jaffee, E.M.; Anders, R.A.; De Jesus-Acosta, A.; Zheng, L. The prognostic value of stroma in pancreatic cancer in patients receiving adjuvant therapy. HPB 2015, 17, 292–298.
  41. Chen, Y.; Kim, J.; Yang, S.; Wang, H.; Wu, C.-J.; Sugimoto, H.; LeBleu, V.S.; Kalluri, R. Type I collagen deletion in αSMA+ myofibroblasts augments immune suppression and accelerates progression of pancreatic cancer. Cancer Cell 2021, 39, 548–565.
  42. Laklai, H.; Miroshnikova, Y.A.; Pickup, M.W.; Collisson, E.A.; Kim, G.E.; Barrett, A.S.; Hill, R.C.; Lakins, J.N.; Schlaepfer, D.D.; Mouw, J.K.; et al. Genotype tunes pancreatic ductal adenocarcinoma tissue tension to induce matricellular fibrosis and tumor progression. Nat. Med. 2016, 22, 497–505.
  43. Drifka, C.R.; Loeffler, A.G.; Mathewson, K.; Keikhosravi, A.; Eickhoff, J.C.; Liu, Y.; Weber, S.M.; Kao, W.J.; Eliceiri, K.W. Highly aligned stromal collagen is a negative prognostic factor following pancreatic ductal adenocarcinoma resection. Oncotarget 2016, 7, 76197–76213.
  44. Moffitt, R.A.; Marayati, R.; Flate, E.L.; Volmar, K.E.; Loeza, S.G.H.; Hoadley, K.A.; Rashid, N.U.; Williams, L.A.; Eaton, S.C.; Chung, A.H.; et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat. Genet. 2015, 47, 1168–1178.
  45. 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.
  46. Hosein, A.N.; Huang, H.; Wang, Z.; Parmar, K.; Du, W.; Huang, J.; Maitra, A.; Olson, E.; Verma, U.; Brekken, R.A. Cellular heterogeneity during mouse pancreatic ductal adenocarcinoma progression at single-cell resolution. JCI Insight 2019, 4, e129212.
  47. Dominguez, C.X.; Müller, S.; Keerthivasan, S.; Koeppen, H.; Hung, J.; Gierke, S.; Breart, B.; Foreman, O.; Bainbridge, T.W.; Castiglioni, A.; et al. Single-Cell RNA Sequencing Reveals Stromal Evolution into LRRC15+ Myofibroblasts as a Determinant of Patient Response to Cancer Immunotherapy. Cancer Discov. 2020, 10, 232–253.
  48. 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.
  49. Davidson, S.; Efremova, M.; Riedel, A.; Mahata, B.; Pramanik, J.; Huuhtanen, J.; Kar, G.; Vento-Tormo, R.; Hagai, T.; Chen, X.; et al. Single-Cell RNA Sequencing Reveals a Dynamic Stromal Niche That Supports Tumor Growth. Cell Rep. 2020, 31, 107628.
  50. Walker, C.; Mojares, E.; Hernández, A.D.R. Role of Extracellular Matrix in Development and Cancer Progression. Int. J. Mol. Sci. 2018, 19, 3028.
  51. Hynes, R.O.; Naba, A. Overview of the matrisome--an inventory of extracellular matrix constituents and functions. Cold Spring Harb. Perspect. Biol. 2012, 4, a004903.
  52. Naba, A.; Clauser, K.R.; Hoersch, S.; Liu, H.; Carr, S.A.; Hynes, R.O. The matrisome: In silico definition and in vivo characterization by proteomics of normal and tumor extracellular matrices. Mol. Cell. Proteom. MCP 2012, 11, M111.014647.
  53. Mouw, J.K.; Ou, G.; Weaver, V.M. Extracellular matrix assembly: A multiscale deconstruction. Nat. Rev. Mol. Cell Biol. 2014, 15, 771–785.
  54. Rozario, T.; DeSimone, D.W. The extracellular matrix in development and morphogenesis: A dynamic view. Dev. Biol. 2010, 341, 126–140.
  55. Charras, G.; Sahai, E. Physical influences of the extracellular environment on cell migration. Nat. Rev. Mol. Cell Biol. 2014, 15, 813–824.
  56. Hastings, J.F.; Skhinas, J.N.; Fey, D.; Croucher, D.R.; Cox, T.R. The extracellular matrix as a key regulator of intracellular signalling networks. Br. J. Pharmacol. 2019, 176, 82–92.
  57. Hynes, R.O. Extracellular matrix: Not just pretty fibrils. Science 2009, 326, 1216–1219.
  58. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801.
  59. Lu, P.; Takai, K.; Weaver, V.M.; Werb, Z. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb. Perspect. Biol. 2011, 3, a005058.
  60. Iozzo, R.V.; Gubbiotti, M.A. Extracellular matrix: The driving force of mammalian diseases. Matrix Biol. 2018, 71–72, 1–9.
  61. Kaukonen, R.; Jacquemet, G.; Hamidi, H.; Ivaska, J. Cell-derived matrices for studying cell proliferation and directional migration in a complex 3D microenvironment. Nat. Protoc. 2017, 12, 2376–2390.
  62. Theocharis, A.D.; Karamanos, N.K. Proteoglycans remodeling in cancer: Underlying molecular mechanisms. Matrix Biol. 2019, 75–76, 220–259.
  63. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674.
  64. Oskarsson, T. Extracellular matrix components in breast cancer progression and metastasis. Breast 2013, 22, S66–S72.
  65. Høye, A.M.; Erler, J.T. Structural ECM components in the premetastatic and metastatic niche. Am. J. Physiol. Physiol. 2016, 310, C955–C967.
  66. Xie, D.; Xie, K. Pancreatic cancer stromal biology and therapy. Genes Dis. 2015, 2, 133–143.
  67. Au, J.L.-S.; Yeung, B.Z.; Wientjes, M.G.; Lu, Z.; Wientjes, M.G. Delivery of cancer therapeutics to extracellular and intracellular targets: Determinants, barriers, challenges and opportunities. Adv. Drug Deliv. Rev. 2016, 97, 280–301.
  68. Beck, A.H.; Sangoi, A.R.; Leung, S.; Marinelli, R.J.; Nielsen, T.O.; van de Vijver, M.J.; West, R.B.; van de Rijn, M.; Koller, D. Systematic analysis of breast cancer morphology uncovers stromal features associated with survival. Sci. Transl. Med. 2011, 3, 108ra113.
  69. Park, J.; Yoon, J. Schizandrin inhibits fibrosis and epithelial—mesenchymal transition in transforming growth factor-β1-stimulated AML12 cells. Int. Immunopharmacol. 2015, 25, 276–284.
  70. Ahn, S.G.; Dong, S.M.; Oshima, A.; Kim, W.H.; Lee, H.M.; Lee, S.A.; Kwon, S.-H.; Lee, J.-H.; Lee, J.M.; Jeong, J.; et al. LOXL2 expression is associated with invasiveness and negatively influences survival in breast cancer patients. Breast Cancer Res. Treat. 2013, 141, 89–99.
  71. Huijbers, A.; Tollenaar, R.A.E.M.; 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.
  72. Imamura, T.; Manabe, T.; Ohshio, G.; Wang, Z.; Yamaki, K.; Yoshimura, T.; Suwa, H.; Imamura, M. Immunohistochemical staining for type IV collagen and laminin in the stroma of human pancreatic cancer. Int. J. Pancreatol. 1995, 18, 95–99.
  73. Mollenhauer, J.; Roether, I.; Kern, H.F. Distribution of extracellular matrix proteins in pancreatic ductal adenocarcinoma and its influence on tumor cell proliferation in vitro. Pancreas 1987, 2, 14–24.
  74. Linder, S.; Castaños-Velez, E.; von Rosen, A.; Biberfeld, P. Immunohistochemical expression of extracellular matrix proteins and adhesion molecules in pancreatic carcinoma. Hepato-gastroenterology 2001, 48, 1321–1327.
  75. Badaoui, M.; Mimsy-Julienne, C.; Saby, C.; Van Gulick, L.; Peretti, M.; Jeannesson, P.; Morjani, H.; Ouadid-Ahidouch, H. Collagen type 1 promotes survival of human breast cancer cells by overexpressing Kv10.1 potassium and Orai1 calcium channels through DDR1-dependent pathway. Oncotarget 2017, 9, 24653–24671.
  76. Kauppila, S.; Stenbäck, F.; Risteli, J.; Jukkola, A.; Risteli, L. Aberrant type I and type III collagen gene expression in human breast cancer In Vivo. J. Pathol. 1998, 186, 262–268.
  77. Liu, J.; Shen, J.-X.; Wu, H.-T.; Li, X.-L.; Wen, X.-F.; Du, C.-W.; Zhang, G.-J. Collagen 1A1 (COL1A1) promotes metastasis of breast cancer and is a potential therapeutic target. Discov. Med. 2018, 25, 211–223.
  78. Armstrong, T.; Packham, G.; Murphy, L.B.; Bateman, A.C.; Conti, J.A.; Fine, D.R.; Johnson, C.D.; Benyon, R.C.; Iredale, J.P. Type I collagen promotes the malignant phenotype of pancreatic ductal adenocarcinoma. Clin. Cancer Res. 2004, 10, 7427–7437.
  79. Barrett, A.S. Compartment resolved proteomics reveals a dynamic matrisome in a biomechanically driven model of pancreatic ductal adenocarcinoma. J. Immunol. Regen. Med. 2018, 1, 67–75.
  80. Ryschich, E.; Khamidjanov, A.; Kerkadze, V.; Büchler, M.W.; Zöller, M.; Schmidt, J. Promotion of tumor cell migration by extracellular matrix proteins in human pancreatic cancer. Pancreas 2009, 38, 804–810.
  81. Lochter, A.; Bissell, M.J. Involvement of extracellular matrix constituents in breast cancer. Semin. Cancer Biol. 1995, 6, 165–173.
  82. Willumsen, N.; Bager, C.L.; Leeming, D.J.; Smith, V.; Karsdal, M.A.; Dornan, D.; Bay-Jensen, A.-C. Extracellular matrix specific protein fingerprints measured in serum can separate pancreatic cancer patients from healthy controls. BMC Cancer 2013, 13, 1–6.
  83. Hušáková, M.; Bay-Jensen, A.-C.; Forejtová, Š.; Zegzulková, K.; Tomčík, M.; Gregová, M.; Bubová, K.; Hořínková, J.; Gatterová, J.; Pavelka, K.; et al. Metabolites of type I, II, III, and IV collagen may serve as markers of disease activity in axial spondyloarthritis. Sci. Rep. 2019, 9, 11218.
  84. Takei, H.; Iino, Y.; Horiguchi, J.; Yokoe, T. Immunohistochemical fibronectin staining pattern and prognosis in invasive breast carcinoma. Oncology 1995, 52, 106–111.
  85. Lin, T.-C.; Yang, C.-H.; Cheng, L.-H.; Chang, W.-T.; Lin, Y.-R.; Cheng, H.-C. Fibronectin in Cancer: Friend or Foe. Cells 2019, 9, 27.
  86. Bae, Y.K.; Kim, A.; Kim, M.K.; Choi, J.E.; Kang, S.H.; Lee, S.J. Fibronectin expression in carcinoma cells correlates with tumor aggressiveness and poor clinical outcome in patients with invasive breast cancer. Hum. Pathol. 2013, 44, 2028–2037.
  87. Yao, H.; Zeng, Z.-Z.; Fay, K.S.; Veine, D.M.; Staszewski, E.D.; Morgan, M.; Wilder-Romans, K.; Williams, T.M.; Spalding, A.C.; Ben-Josef, E.; et al. Role of α(5)β(1) Integrin Up-regulation in Radiation-Induced Invasion by Human Pancreatic Cancer Cells. Transl. Oncol. 2011, 4, 282–292.
  88. Chen, R.; Pan, S.; Ottenhof, N.A.; de Wilde, R.F.; Wolfgang, C.L.; Lane, Z.; Post, J.; Bronner, M.P.; Willmann, J.K.; Maitra, A.; et al. Stromal galectin-1 expression is associated with long-term survival in resectable pancreatic ductal adenocarcinoma. Cancer Biol. Ther. 2012, 13, 899–907.
  89. Chen, R.; Dawson, D.W.; Pan, S.; Ottenhof, N.A.; de Wilde, R.F.; Wolfgang, C.L.; May, D.H.; Crispin, D.A.; Lai, L.A.; Lay, A.R.; et al. Proteins associated with pancreatic cancer survival in patients with resectable pancreatic ductal adenocarcinoma. Lab. Investig. 2015, 95, 43–55.
  90. Astorgues-Xerri, L.; Riveiro, M.E.; Tijeras-Raballand, A.; Serova, M.; Neuzillet, C.; Albert, S.; Raymond, E.; Faivre, S. Unraveling galectin-1 as a novel therapeutic target for cancer. Cancer Treat. Rev. 2014, 40, 307–319.
  91. Kailemia, M.J.; Park, D.; Lebrilla, C.B. Glycans and Glycoproteins as Specific Biomarkers for Cancer. Anal. Bioanal. Chem. 2017, 409, 395–410.
  92. Delpech, B.; Chevallier, B.; Reinhardt, N.; Julien, J.-P.; Duval, C.; Maingonnat, C.; Bastit, P.; Asselain, B. Serum hyaluronan (hyaluronic acid) in breast cancer patients. Int. J. Cancer 1990, 46, 388–390.
  93. Schwertfeger, K.L.; Cowman, M.K.; Telmer, P.G.; Turley, E.A.; McCarthy, J.B. Hyaluronan, Inflammation, and Breast Cancer Progression. Front. Immunol. 2015, 6, 236.
  94. Schmaus, A.; Sleeman, J.P. Hyaluronidase-1 expression promotes lung metastasis in syngeneic mouse tumor models without affecting accumulation of small hyaluronan oligosaccharides in tumor interstitial fluid. Glycobiology 2015, 25, 258–268.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 470
Revisions: 2 times (View History)
Update Date: 29 Mar 2022
1000/1000