Open access peer-reviewed chapter

Extracellular Matrix in Tumor Angiogenesis

Written By

Gvantsa Kharaishvili

Submitted: 14 March 2022 Reviewed: 24 March 2022 Published: 25 May 2022

DOI: 10.5772/intechopen.104661

From the Edited Volume

Tumor Angiogenesis and Modulators

Edited by Ke Xu

Chapter metrics overview

251 Chapter Downloads

View Full Metrics

Abstract

Extracellular matrix (ECM) is a complex three-dimensional network that provides structure, strength, and contextual information for cellular growth, communication, differentiation, survival, adhesion, and migration. ECM basic proteins resist compressive forces and/or allow rapid diffusion, others strengthen the matrix, and give resilience or modulate cell-matrix interactions. ECM undergoes turnover and remodeling physiologically and during inflammation, wound repair and tumor invasion. Remodeling of the ECM is an integral component of the angiogenic process and depends on the composition of matrix molecules, soluble pro-angiogenic and anti-angiogenic factors, and their spatial regulation. This review will focus on the myriad roles of those molecules and will emphasize their involvement in critical points of angiogenesis.

Keywords

  • extracellular matrix
  • tumor microenvironment
  • angiogenesis
  • pro-angiogenic
  • anti-angiogenic

1. Introduction

“Tumor progression is defined by irreversible change in the tumor characteristics reflecting the sequential appearance of a genetically altered subpopulation of cells with the new characteristics” [1]. The term, “tumor progression” is used to describe phenotypic changes in the preexisting neoplastic lesion. It is a coincidence of complex events characterized by morphological, molecular, and functional changes in tumor cells and their environment and encompasses a wide scale of mechanisms [2]. It is in part recognized as a product of evolving crosstalk between different cell types within the tumor and its surrounding supportive tissue or tumor stroma [3]. Invasive tumor cells interact with their microenvironment in a bidirectional manner and remodel it into a supportive context for tumor growth and progression. The composition of the tumor microenvironment varies between tumor types, but hallmark features include cellular components such as immune cells (T-cells, B-cells, NK-cells, macrophages, neutrophils, dendritic cells), stromal cells, blood vessels, cancer-associated fibroblasts, adipocytes, stellate cells, and noncellular components such as extracellular matrix (ECM) and exosomes [4].

Advertisement

2. Extracellular matrix: its composition and molecular profile

Extracellular matrix (ECM) is a noncellular, proteinaceous component of the stroma. It is a complex three-dimensional network of macromolecules. The ECM provides architectural structure, strength, and contextual information for cellular growth, adhesion, communication, differentiation, migration, and survival. Molecules that provide ECM structure are: glycosaminoglycans and proteoglycans (form a hydrated gel-like substance, resist compressive forces, and allow rapid diffusion) and fibrous proteins and collagens (strengthens the matrix and give resilience). They represent insoluble factors of the matrix [5]. Structural molecules are synthesized mainly by fibroblasts but also by other cells of connective tissue. ECM molecules named, “matricellular proteins” (e.g. thrombospondin-1 and -2, SPARC, tenascin-C, and osteopontin) do not function as structural elements but modulate cell-matrix interactions and cell functions [6]. ECM is in a dynamic state and undergoes turnover and remodeling in conjunction with signals and is enhanced during inflammation, wound repair, and tumor invasion. However, ECM can limit initiation of tumor at an early stage of its development, later, ECM stimulates tumor growth and progression and enhances its aggressiveness. Key enzymes which remodel ECM are matrix metalloproteinases (MMPs) and urokinase-type plasminogen activators (uPAs). They degrade components of the basement membrane as well as proteins and proteoglycans of connective tissue and liberate latent growth factors from their storage sites in the extracellular matrix. Factors that are activated in this fashion are, for example, fibroblast growth factors (FGFs), hepatocyte growth factor (HGF), and transforming growth factors (TGFs) [7]. Tumor growth-induced solid stress, matrix stiffness, increased interstitial fluid pressure, hypoxia and altered tumor pH have been established as a result of tumor growth and on the other hand, neoangiogenesis-supporting conditions. As structural and metabolic alterations of ECM can lead to the development or progression of disease, its molecules can serve as important targets for pharmacotherapy.

2.1 Collagens

Collagen represents 30% of dry weight in the human body and is the most abundant protein synthesized by fibroblasts and by several other cell types distinct by their molecular profile, morphology, distribution function, and involvement in pathologies [8]. Collagens play structural roles and contribute to mechanical properties, organization, and configuration of tissues. Some collagens have a restricted tissue distribution and hence specific biological functions [9]. Collagens are trimeric molecules composed of three polypeptide α chains, which contain the sequence repeat that allows the formation of a triple helix. Besides triple-helical domains, collagens contain non-triple-helical domains, used as building blocks by other extracellular matrix proteins. At present, 28 types of collagens are classified as fibrillar collagens, unconventional collagens including collagen VII, network-forming collagens (VI, VIII, and X), fibril-associated collagens with interrupted triple helix (IX, XII, XIV, XVI, and XIX), basement membrane collagens, transmembrane collagens, and multiplexins [10, 11]. Type I, II, III, V, XI, XXIV, and XXVII collagens belong to the classical fibrillar collagens [12]. Fibrillar collagens can assemble into supramolecular aggregates. Type I collagen is major collagen of tendons, ligaments, skin, cornea, and other connective tissues representing 90% of the total collagen in humans. It is mostly a part of the compound containing either type III collagen seen in skin and reticular fibers [13] or type V collagen found in bone [14]. The biomechanical properties of these compounds (e.g., torsional stability and stiffness or tensile strength) establish the stability and integrity of these tissues [15]. Bourgot and colleagues describe the evolution of fibrillar collagen organization during tumor progression where tumor-derived paracrine signals promote a desmoplasic reaction characterized by the activation of the resident fibroblasts into cancer-associated fibroblasts (CAFs) with enhanced secretory activity, reorganization of the collagen fibers (their cross-linking), augmenting the stiffness of the stroma. Tumor adjacent collagen fibers that promote invasive cancer cell migration can be organized parallel (Tumor Associated Collagen Signature—TACS-2) or perpendicular to the tumor border (TACS-3) [16]. Collagen fibers employ guidance signals for endothelial cell migration during regenerative angiogenesis. Inhibition of collagen cross-linking results in a 70% shorter regeneration area with 50% reduced vessel growth and disintegrated collagen fibers. The disrupted collagen scaffold impedes endothelial cell migration and induces the formation of abnormal angioma-like blood vessels [16]. Type I collagen, potently stimulates angiogenesis in vitro and in vivo [17]. Crucial to its angiogenic activity appears to be ligation and possibly clustering of endothelial cell surface α1β1/α2β1 integrin receptors by the GFPGER (502–507) sequence of the collagen fibril. Authors describe here genetically engineered “angiogenic superpolymers”, containing type I collagen, fibrillar collagens and collagen mimetics, possibilities of their modifications to display ideal angiogenic properties, and prove their usefulness for tissue engineering and human medicine [17].

The vascular basement membrane represents an insoluble structural component of the wall of newly formed capillaries and undergoes several changes during tumor-induced angiogenesis. Initially, the membrane is degraded and disassembled by proteolytic activity of matrix metalloproteinases, mainly MMP2 and 9, but is finally after complex molecular crosstalk by regulation mainly via VEGF signaling, is reorganized to a native state around a newly formed capillary. Such vascular matrix changes during angiogenesis are associated with the expression of matrix proteins that can interact with vascular endothelium and provide endogenous angiogenic and anti-angiogenic signals. Basement membrane molecules play a role also in the process of the relapse of pathological angiogenesis [18]. Rapid relapse of tumor angiogenesis is hypothesized to be facilitated by the empty basement membrane sleeves (ebms) of previously regressed vessels, which are postulated to serve as scaffolding for endothelial cells during new angiogenic sprouting, following cessation of antiangiogenic treatment [19]. Type IV collagen is found in solid and soluble states in ECM, it is composed of three α(IV) chains [20]. The a1 and a2 isoforms are ubiquitously present in human basement membranes. Type IV collagen promotes cell adhesion, migration, differentiation, growth [21], and regulates endothelial cell proliferation and behavior during the critical steps of the angiogenic process. Studies have shown that the function of type IV collagen in the elongation and stabilization of microvessels was dose-dependent with low concentrations of type IV collagen promoting elongation, and high concentrations stabilizing them. Anti-angiogenic properties were associated with inhibitors of collagen metabolism and basement membrane collagen synthesis and deposition were crucial for blood vessel formation and survival [18]. There are six known bioactive peptides generated from collagen type IV [22]. These peptides are fragments of non-collagenous domains from the α1 (arresten), α2 (canstatin), α3 (tumstatin), α4 (tetrastatin), α5 (pentastatin), and α6 chains (hexastatin). Arresten, is an inhibitor of angiogenesis in squamous cell carcinoma, binding with α1β1 integrin in endothelial cells [22, 23, 24]. Carcinoma cells showing overexpression of arresten changed to an endothelial phenotype, suggesting inhibition of migrating carcinoma cells by inducing mesenchymal to endothelial (MET) transition [24]. Role of arresten is demonstrated in modulating the function of capillary endothelial cells and blood vessel formation using in vitro and in vivo models of angiogenesis and tumor growth [25]. Recently, the NC1 domain of the α2 chain of type IV collagen (canstatin) was also identified as an angiogenesis inhibitor. In the study by [25], Canstatin was first identified as vasculogenic mimicry (VM) inhibitor. Vasculogenic mimicry is a neovascularization phenomenon that was first reported in melanoma models. Distinct from classical tumor angiogenesis, VM provides a blood supply for tumor cells independent of endothelial cells and formed by deregulated tumor cells. VM is established in lung cancer [26], hepatocellular carcinoma [27], and glioma [28] and is associated with poor prognosis in cancer patients [29]. Vautrin-Glabik demonstrated that 13 amino acid sequence of tetrastatin decreases VEGF-induced-angiogenesis in vivo using the Matrigel plug model and decreases Human Umbilical Vein Endothelial Cells (HUVEC) migration and pseudotube formation in vitro [30]. Oskimaki et al. recently developed a bioinformatics-based approach to predict over 100 novel endogenous anti-angiogenic peptides [31]. An important peptides determined were tetrastatins, pentastatins, and hexastatins that were validated in vitro in cell proliferation and migration assays on HUVECs [32]. Using pentastatin-1 to an angioreactor-based directed in vivo angiogenesis assay (DIVAA), and in vivo NCI-H82 SCLC xenograft model strong potential for pentastatin-1 as a therapeutic agent for lung cancer was demonstrated [30].

2.2 Elastin

Elastin provides elasticity to the ECM. Elastin is roughly 1000 times more flexible than collagens. It is produced as tropoelastin, a 72 kDa precursor protein by fibriblasts, smooth muscle cells, chondrocytes, or endothelial cell and is secreted from the cell to the extracellular space, where it crosslinks with other elastin molecules. Elastin is the primary ECM protein present in arteries where it composes ~50% of their dry weight [33]. During aging, continuous mechanical stress and an increase in elastase activity contribute to the fragmentation of elastic fibers resulting in the release of elastin-derived peptides (EDPs) [34]. EDPs are matrikines—matrix fragments having the ability to regulate cell physiology and display a wide range of biological activities in a number of normal and transformed cells [35]. For example, they potentiate the migration and matrix invasion of tumor cells, stimulate the migration and proliferation of monocytes and skin fibroblasts and up-regulate MMP expression by fibroblasts inducing a remodeling program for melanoma invasion. Additionally, they are pro-angiogenic, chemotactic for inflammatory cells and promote elastase release [36]. Robinet and colleagues showed that elastin-derived peptides enhanced angiogenesis in the chick chorio-allantoic membrane in vivo, augmented pseudotube formation from human vascular and microvascular endothelial cells in the matrigel and promoted cell migration in wound healing assay [37].

2.3 Glycosaminoglycans

Glycosaminoglycans (GAGs) were primarily known as “space fillers” in the ECM, but later appeared as active signaling molecules in cell fate regulation via cytokine production, leukocyte recruitment, or inflammatory response [38]. GAGs are linear polysaccharides with two basic saccharide molecules that vary according to epimerization, sulfation, and deacetylation. Their specificity and functionality depend on the order of the carbohydrate chain and the other chemical modifications [38]. Hyaluronan is the simplest GAG since it is non-sulfated, does not undergo epimerization, and does not use typical covalent bonds for linking to proteins. Other GAGs—chondroitin sulfate, dermatan sulfate, keratan sulfate, and heparan sulfate usually use covalent bonds for attachment to proteins in proteoglycan molecules. Chondroitin and heparan sulfate are further remodeled by sulfation [33, 39]. Hyaluronan is synthesized at the plasma membrane by transmembrane enzymes of the HA synthase family (HAS1–3) [40, 41]. Chain length is dependent on polymerizing enzyme type, for example, HAS1 and HAS2 produce high molecular weight (~2000 kDa) HA and HAS3 produce lower molecular weight (100–1000 kDa) HA. After synthesis via HASes, extracellular HA can be rapidly altered due to its impressive turnover rate via a variety of hyaluronidases (mainly HYAL1 and 2) [40]. Despite the relative simplicity of its molecule, HA regulates a variety of cellular functions including wound repair, inflammation, cell migration, and angiogenesis [41, 42], and recently emerged as a key player in regulating the tumorigenic and inflammatory milieu [43]. Interestingly, its physiological sequel is largely related to the size of the molecule, for example, full-length HA mainly demonstrates anti-inflammatory property whereas its smaller fragments exert pro-inflammatory and pro-angiogenic features [40]. In cancer and other pathologic states, HA fragments are abundantly deposited in the extracellular environment that, in one hand, is a result of increased synthesis of HA via HASes and on the other hand—accelerated degradation via hyaluronidases, reactive oxygen species, and mechanical forces [44] creating a microenvironment supporting angiogenesis and inflammation [41, 45, 46]. Several evidence suggests that aberrant levels of HAS2 promote breast cancer growth, differentiation, lymph node involvement, and worse patient survival [47, 48]. HAS2 knockdown inhibited breast cancer growth and attenuated HA expression. Similarly, HAS2 has a regulatory effect on tumorigenicity and metastasis of prostate, colon, and ovarian tumors through excessive HA synthesis [49, 50]. Recently, Chen and colleagues [40] suggested a novel mechanism of angiogenesis regulation via autophagic degradation of HAS2 in endothelial cells. In [51], colleagues showed that the C-terminal module of perlecan, endorepellin, blocks VEGFR2 kinase activity, thereby evoking a strong pro-autophagic and anti-angiogenic response in vascular endothelial cells both ex vivo and in vivo. Bix and colleagues [52] have also shown that systemic delivery of recombinant endorepellin inhibits tumor growth and angiogenesis and increases tumor hypoxia in squamous and Lewis lung carcinoma xenograft models. Recently, HAS2 was degraded in vascular endothelial cells via autophagy evoked by nutrient deprivation, mTOR inhibition, or pro-autophagic proteoglycan fragments endorepellin and endostatin [40]. Autophagic degradation of HAS2 suppressed extracellular hyaluronan and inhibited ex vivo angiogenesis showed in aortic ring assay where they quantified the extent of active sprouting issued from the aortic rings and measured the radial distance of the newly-formed vessels where they found a significant reduction in angiogenesis [40]. The antiangiogenic activity of the role of endostatin and tumstatin was also emphasized, where tumor suppressor protein p53 prevented an incipient tumor from switching to the angiogenic phenotype mediated in part by endostatin and tumstatin [53].

The role of tumor-associated macrophages in angiogenesis is documented in [54]. TAMs induce tumor vascularization by releasing several factors, including VEGF which is the main angiogenic factor [55]. Monocytes (Mo) and monocyte-derived macrophages (MØ) can bind HA which induces intracellular signals [56, 57], however, the anti-tumor or pro-tumor role, is dependent on the size of HA in colorectal and breast carcinomas. As it is shown in [55] tumor necrosis factor (TNF)-stimulated gene 6 (TSG-6) was downregulated in Mo/MØ by high molecular weight hyaluronan, modulating their angiogenic behavior in breast carcinoma milieu, but not in colorectal carcinoma [55].

2.4 Proteoglycans

Next to collagens, proteoglycans (PGs) constitute a major class of extracellular matrix/cell surface components known to be involved in primary physiological and pathological phenomena; and due to the altered transcription/translation patterns that these PGs exhibit, they have been identified as potential diagnostic/prognostic and therapeutic targets in diverse disease states [58]. Based upon its direct involvement in cell-cell and cell-ECM interactions, this gene family has been strongly implicated in the regulation of cell movement. Assignment of diverse roles of PGs in promoting, or inhibiting, cell movement seems to be dictated by the biological system [58]. The proteoglycan superfamily now contains more than 30 molecules. They sustain the transparency of the cornea, the elasticity of blood vessels, the tensile strength of the skin, tendon, or cartilage, as well as compressive forces of the mineralized matrix of bones. PGs can alter the biology of growth factors and cytokines [59]. The basic proteoglycan unit consists of a “core protein” with one or more covalently attached glycosaminoglycan chain(s). Proteoglycans can be categorized depending upon the nature of their glycosaminoglycan chains and/or by size (kDa). Four major classes of PGs exist: (i) chondroitin sulfate/dermatan sulfate PGs (decorin, biglycan, versican); heparan sulfate/ chondroitin sulfate PGs (testican, perlecan); (ii) chondroitin sulfate (neurocan, aggrecan); (iii) keratan sulfate (fibromodulin, lumican). Among them, decorin, biglycan, testican, fibromodulin, lumican are small proteoglycans, and versican, perlecan, neurocan, and aggrecan are large proteoglycans. The small leucine-rich repeat proteoglycans (SLRPs) form a group of molecules on the basis of their relatively small protein core (36–42 kDa) [60, 61]. Some of these gene products are not classical proteoglycans. Despite being structural proteins, SLRPs constitute a network of signal regulation: being mostly extracellular, they are upstream of multiple intracellular signaling cascades. They affect intracellular phosphorylation and modulate pathways, including those driven by bone morphogenetic protein/transforming growth factor β superfamily members, receptor tyrosine kinases such as ErbB, and the insulin-like growth factor I receptor, and Toll-like receptors.

Decorin was originally discovered as a collagen-binding protein necessary for fibrillogenesis [62, 63], hence related eponym of decorin [64]. Soluble decorin is a high-affinity antagonistic ligand for several key receptor tyrosine kinases resulting in protracted oncostasis and angiostasis [65]. Recently, decorin has emerged as a soluble pro-autophagic cue by initiating endothelial cell autophagy through activation of AMPK, an energy sensor kinase, and evoking tumor cell mitophagy as the mechanistic basis for the oncostatic effects [66]. Decorin, due to its role as a tumor repressor and anti-angiogenic factor was designated as “a guardian from the matrix” [67]. According to the review, decorin suppresses tumor growth and angiogenesis via EGFR and Met where decorin monomer binds a narrow region of an epitope that in part overlaps with the agonist binding site [68]. This binding further augments receptor dimerization, the consequence of which is rapid phosphorylation of the intracellular tails [69]. This event further recruits and activates downstream effectors, e.g. provides caveosome-mediated internalization of the decorin/receptor complex, and eventual degradation in lysosomes [70, 71]. The latter causes a protracted cessation of intracellular receptor signaling. As a major consequence of inhibiting Met, two potent oncogenes, β-catenin, and Myc, are targeted for sustained degradation via the 26S proteasome [72]. Decorin suppresses β-catenin signaling in a non-canonical fashion and the latter is targeted for degradation in a manner consistent with direct phosphorylation of β-catenin by an RTK, such as Met [73, 74, 75]. Wnt/β-catenin signaling activation and its member molecule mutations are well established in colorectal cancer and different epithelial tumor sprouting and nonsprouting angiogenesis. Wnt agonists (e.g., B cell Lymphoma 9 protein (BCL9) is the angiogenesis promoting, where antagonists such as the DKK-4 (also called the Dickkopf Wnt signaling pathway inhibitor 4), in particular, conditioned media from DKK-4 expressing cells promoted the migrative abilities of CRC and formation of capillary-like tubules of human primary microvascular endothelial cells [76].

Versican is a large chondroitin sulfate proteoglycan that forms aggregates with hyaluronan which connects it to the cell surface via hyaluronan receptors such as CD44 [77, 78]. Versican is implicated in many biological processes involving vasculature, such as atherosclerosis and vasculitis [79, 80]. There are five known versican splice isoforms; V0–V4 [81]. Each isoform except V3 has a glycosaminoglycan (GAG) domain with covalently attached chondroitin sulfate (CS) chains. Versican is highly expressed in the early stages of development but becomes downregulated after tissue maturation [82], interestingly, it is reexpressed during wound repair, arteriopathies, pulmonary fibrosis, or tumor formation [83]. Versican is anti-adhesive since it is a poor cell attachment and migration substrate and is excluded from focal adhesions [77, 84, 85]. Several clinical studies have suggested that high versican expression is a poor prognostic factor in gastric, pancreatic, head and neck squamous, or mammary cancers [77]. Increased versican immunostaining has been detected during tumor blood vessel formation [86]. Versican V2 isoform is the major type expressed in brain tissues, and brain tumors are greatly enriched in vascularization, therefore, authors hypothesized that the V2 isoform may play a role in angiogenesis in brain tumors. They injected U87 glioblastoma cells stably transfected with a versican V2 expression construct or a control vector into nude mice and showed that the tumors formed by the V2-transfected cells were visibly enriched in vascularisation, whereas the tumors formed by the vector-transfected cells did not exhibit this phenotype [86]. Furthermore, V2 expression facilitated endothelial-tumor cell interaction observed in tube-like structure formation in matrigel [82]. Koyama and colleagues demonstrated that basic fibroblast growth factor-induced neovascularization was elevated in the presence of either hyaluronan oligosaccharides or a hyaluronan aggregate containing versican, using the Matrigel plug assay. Administration of hyaluronan-versican aggregates, but not native hyaluronan alone, promoted stromal cell recruitment with the infiltration of endothelial cells, suggesting that hyaluronan overproduction accelerates tumor angiogenesis through stromal reaction, notably in the presence of versican [87]. Versican localized preferentially to the vicinity of tumor vasculature and macrophages in the tumor. However, the extracellular protease ADAMTS-generated versican fragment is uniquely localized to vascular endothelium. Members of the family of A disintegrin-like and metalloproteinase with thrombospondin type 1 motifs (ADAMTS) are involved in versican proteolysis and tumor progression [88, 89]. ADAMTS1 was first shown to display anti-angiogenic properties [90]. Later, it’s angiostatic (antiangiogenic) and tumor-suppressive properties have also been shown in model systems [91, 92], but controversial results about its relevance to metastasis and tumor growth have also gained attention [93]. ADAMTS family of secreted zinc-dependent metalloproteinases comprises at least 19 genetically distinct members in humans [94]. The expression of the majority of ADAMTS subtypes is associated with pre- and postnatal growth and onset and progression of cancer [95]. ADAMTS subtypes have been sub-classified as aggrecanases because of their ability to cleave large chondroitin sulfate. Despite their structural similarity to other matrix metalloproteinases, ADAMTS have a narrow substrate specificity. This feature could serve as an advantage for ADAMTS inhibitors in the treatment of cancer [95].

Asporin, also known as periodontal ligament-associated protein 1 (PLAP1) was identified in 2001 [96, 97]. Asporin mRNA was expressed primarily in the skeleton (perichondrium/periosteum of cartilage/bone) and other specialized connective tissues. Asporin blocks chondrogenesis and inhibits TGF-β1-induced expression of matrix genes and the resulting chondrocyte phenotypes [98]. Knockdown of asporin increases the expression of cartilage marker genes and TGF-β1; in turn, TGF-β1 stimulates asporin expression in articular cartilage cells, suggesting that asporin and TGF-β1 form a regulatory feedback loop. Asporin, like decorin, can bind collagen at the same site, but in contrast to decorin and biglycan, it drives collagen biomineralization [99]. Our laboratory has identified asporin as a novel cancer-related protein in invasive breast cancer [100]. Later, asporin was reported as an important player in tumor microenvironment [101] and experimentally proved that MDA-MB-231 and BT-549 cells invaded faster through collagen matrix which was prepared with the recombinant asporin. This finding was explained to be related to a less dense matrix due to the inhibition of collagen fibrillogenesis by asporin [102]. Recently, asporin was specifically reported in pancreas and prostate cancer by two additional groups [103, 104]. The direct role of asporin in angiogenesis/angiostasis is not been studied yet, however, a search of the Gene Expression Omnibus, revealed high levels of ASPN expression in white adipose-derived (WAT) CD34+ cells that are a very rich reservoir of CD45− CD34+ populations with endothelial differentiation potential/significantly increased levels of angiogenesis-related genes [101]. The multifaceted role of asporin was recently reviewed also in [105] where its emerging role in proliferation, migration, invasion, and angiogenesis through TGF-β, EGFR, and CD44 pathways was described [105].

2.5 Laminin

Laminins are major noncollagenous constituents of the basement membrane. The fragmentation or absence of BM structures seen in malignant tumors is due to active proteolytic degradation, decreased synthesis of BM components, and/or remodeling by the tumor cells [106]. There are 5α, 4β, and 6γ chains of laminin molecule [33]. It has three short and one long arm arranged in a cross-like structure. The α chains have a larger G domain at the C-termini, which is composed of 3 LG domains (LG1-LG3) connected by a binding region to other LG4 and 5 domains. Integrins bind to LG1–3. Heparan sulfate has been shown to bind to LG4 of the α1 chain. Certain laminin isoforms are predominant in vascular basement membranes and may be critical in maintaining the proper development as well as stability of the mature vessel [107]. LN-1 provoked angiogenesis in the chicken chorioallantoic membrane in the same manner as FGF-2, and vessel development in embryoid bodies was further enhanced in a synergistic mode by FGF-2 and LN-1. The latter significantly enhanced the differentiation of endothelial cells in a 3D collagen environment, either in the absence or presence of FGF-2 [108]. In tumors, as in normal tissues, the blood vessels express laminin α4, α5, β1, and γ1 chains, suggesting the presence of laminin-8 and -10, synthesized by VECs. Laminin-10 is more adhesive and migration promoting [109]. Microvessels are expected to express additional laminins α2, α3, and β2 [107]. The cellular origin of the laminin chains in the vessel should be carefully examined, since pericytes are also able to synthesize several laminins [110]. Lugassy and colleagues [111] in their work studied qualitative aspects of tumor cells and vasculature in melanoma and focused on the pericellular matrix. They demonstrated the angio-tumoral complex in which the tumor cell and endothelium are in direct contact via an amorphous matrix. This amorphous matrix lacks an organized lamina and contains predominantly laminin with noticeably less collagen type IV. Interestingly, this was absent in naevi. Authors regarded the laminin found in this amorphous matrix as “free” laminin, is distinct from laminin integrated into an organized lamina, and showed free laminin role in promoting the migration of melanoma cells in contact with vessels and suggested that this angio-tumoral complex represents a marker for metastasis [112]. During intravasation, tumor cells penetrate BM rich in laminin-8 and 10. When in circulation, large tumor cells and cell aggregates are often covered with platelets, that contain and, following stimulation, secrete laminin-8 and other laminin isoforms [113]. Tumor cell extravasation again requires penetration of the vascular BM to generate secondary tumors [107]. Interaction of tumor cells with endothelial cells and the basement membrane seems organ-specific, time and tumor type-dependent in the ultrastructural study on lung, liver, brain, kidney, and adrenal tissues. Study shows that endothelial cells of the lungs and liver can play a much more active role in the process of extravasation [114]. Laminin α3B chain normally expressed in vascular and epithelial basement membranes, was downregulated in skin cancers [115]. Notably, endothelial cell behavior during tumor progression is largely dependent on complex interactions between laminin molecules with integrins (please see also below).

2.6 Fibronectin

Fibronectin is a dimer with a molecular weight of ~270 kDa. There are two fibronectin forms, soluble plasma fibronectin (p-fibronectin), produced by hepatocytes and cellular fibronectin (c-fibronectin) produced in tissues where it is further deposited as a component of the fibrillar matrix. Many of the functions of fibronectin depend on the 3-dimensional structure of the protein and its assembly into a functional fibrillar matrix [116]. In ECM, fibronectin binds collagen, heparin, other fibronectin proteins, and cell surface integrins. Fibronectin binds integrins through the tripeptide motif of arginine, glycine, and aspartic acid (RGD)2,3, α5β1 integrin plays here a major role. Studies to elucidate the mechanisms of fibronectin fibrillogenesis in endothelial cells have revealed a determinant role for integrin beta subunit adaptor (ILK) in this process [117]. Example of how transient c-fibronectin expression participates in a “pro-angiogenic switch” comes from studies on vascular patterning in the developing retinal vasculature [118, 119]. During this process, blood vessels use the existing astrocyte network as a template, and fibronectin is the principal component of the astrocyte-derived extracellular scaffold. Bazigou et al. [120] showed that interaction between integrin α9 and fibronectin containing the EDA domain is required for fibronectin matrix assembly during lymphatic valve morphogenesis [120]. Targeted deletion of α4 in lymphatic vessels or pharmacological inhibition of α4β1 compromise growth factor- and tumor-induced lymphangiogenesis and suppressed metastatic spread in vivo. α4β1 and c-fibronectin were suggested as markers of proliferative lymphatic endothelium in malignant tumors [121]. Fibronectin is a Wnt target gene and lung vascularization and branching morphogenesis are dependent on Wnt and fibronectin signaling [122]. However, fibronectin level is weak in morphogenesis and quiescent vasculature and highly upregulated together with tenascin-C following vessel injury. Tenascin-C expression is also highly associated with angiogenesis in a wide range of disease states, including diabetes, aortic aneurysm, artherosclerosis, ulcerative colitis, inflammatory bowel disease, Crohn‘s disease, vasculitis, and cancer [122]. Both proteins were localized in the vessel wall, where fibronectin was more abundant on the luminal side and tenascin-C on the extraluminal side of the vascular BM. To note, tumor vessels were diversely positive for tenascin-C and oncofetal fibronectin, suggesting a temporally and spatially regulated expression of these ECM proteins in the tumor vasculature and may reflect different maturation states of the vessels. Re-expression of fibronectin occurs during pathological angiogenesis in various diseases such as cancer, late-stage atherosclerosis, and blinding ocular conditions [123, 124].

Advertisement

3. Cell-extracellular matrix interactions and angiogenesis

3.1 Integrins

Integrins are the main receptors involved in cell-matrix contacts. They contain transmembrane subunits α and β, large extracellular domain, and intracellular domain that interacts with cytoskeleton proteins. Subunits form 24 integrins. Integrins provide transmission of chemical and mechanical signals, which results in rearrangement of the cell cytoskeleton and activation of pathways that control cell survival and motility, angiogenesis, differentiation, and apoptosis. The ability of the cell to survive without contact with a substrate is a feature of tumor cells. Integrin expression changes significantly during carcinogenesis and different tumors express different integrins. Integrin α6β4 in cooperation with epidermal growth factor receptor (EGFR) is expressed mostly in breast carcinoma [125], while integrin αVβ3 in cooperation with platelet-derived growth factor (PDGF) and EGFR are expressed in glioblastomas and melanomas [126]. The role of integrins in tumor angiogenesis has been partially discussed above in relation to laminins and will also be discussed below.

Matrix metalloproteinases (MMPs), also known as matrixins, are members of the metzincin protease superfamily of zinc-endopeptidases. There are 187 members of MMPs which are encoded in the human genome and 28 members are secreted MMPs. They can degrade every protein in ECM and basement membranes. Several MMPs are membrane-type which contribute to the precise localization of protease activity, as this is required at the edge of migrating cells. Several MMPs—collagenase, gelatinase, matrilysin degrades collagen, gelatin, and fibronectin, respectively. Stromelysin degrades structural proteins and proteoglycans. MMP activity is regulated by tissue inhibitors of MMPs (TIMPs 1–4) which are produced by more cells than MMPs themselves [127]. MMPs are directly implicated in embryonic growth and tissue morphogenesis that require disruption of ECM barriers for microenvironment remodeling and cell migration and contribute to the formation of a complex microenvironment for tumor development and progression through activation of growth factors, suppression of tumor cell apoptosis, destruction of chemokine gradients developed by host immune response, or release ECM-sequestered angiogenic factors [128]. For example, MMP-11 (human stromelysin-3, hST-3) favored the release of insulin-like growth factor 1 that is bound to specific binding proteins (IGFBPs) [129]. MMP-9 can proteolytically activate TGF-β and promote tumor invasion and angiogenesis [130]. Several other pro-angiogenic factors such as VEGF and basic fibroblast growth factor (bFGF) are induced/activated by MMPs. MMP-14 overexpression by cancer cells increases VEGF synthesis and promotes angiogenesis in glioblastomas [131] and breast carcinomas [132, 133]. VEGF expression was also inspired by MMP-2 in A549 lung adenocarcinoma cells through the binding to αvβ3 and activated integrin signaling [134]. Cancer cell-derived MMP-13 (collagenase-3) also induced VEGF synthesis by endothelial cells and fibroblasts and initiated tumor angiogenesis in vivo [135]. MMP-1, -8, and -13 are collagenases associated with angiogenesis and their loss leads to irreversible rupture of the matrix [136]. The fragmentation of basement membrane type IV collagen is carried out by MMP-2 and MMP-9. Type IV collagenase activity is important in the early steps of endothelial cell morphogenesis/capillary formation. Interstitial collagenase (MMP-1) is a membrane-type 1 matrix metalloproteinase (MT1-MMP) that can also break down collagen types I–III, gelatin, laminin, and other ECM components. MT1-MMP is expressed by endothelial cells and it may regulate angiogenesis by activating pro-MMP2 and by cleaving collagens on the cell surface at a highly localized site [136]. Tissue inhibitors of metalloproteinases regulate them, playing a key role in angiogenesis regulation by inhibiting neovascularization.

3.2 Matrix topology, stiffness, and solid stress

Physical and chemical features of the tumor environment determine matrix topology (architecture) and stiffness that depends on the size of biopolymer fibers and the density of the fiber network [137]. Connective tissue is characterized by different fiber arrangements. Different combinations and densities of the cells, fibers, and other ECM components as well as different fiber arrangements ranging from loose or random to highly aligned structures, produce graded variations of connective tissue. ECM topology can represent an important regulator of cell motility through physical signals that geometrically impel adhesion foci to conduct directional migration [138]. Cancer cells perform contact guidance mediated by mechanosensory integrins through which they, using contractile force, actively remodel the ECM fibers surrounding a tumor (align them perpendicularly to the tumor) [137, 138, 139]. Dense fibrillar collagen that is characteristic of breast cancer stroma forms radial patterns extending away from tumors. On the other hand, the reticular arrangement of the collagen matrix surrounding mammary glands may anchor and/or hinder cells. Thus, ECM topography, in particular, its non-linear pattern reduces invasion while linear structure promotes it. Matrix concentration and post-translational modifications such as glycosylation and cross-linking affect the mechanical properties, including viscoelasticity or stiffness. Tumors exhibit a higher degree of stiffness than their normal adjacent counterpart. For example, the healthy mammary gland is highly compliant (elastic modulus E = ~200 Pa), while the average tumor is stiffer (E = ~4000 Pa). Both the tumor-surrounding stroma and vasculature exhibit increased stiffness (E = ~800–1000 Pa and ~450 Pa [140].

Changes in ECM topology and stiffness can shape mechanosensing events and activate intracellular signaling processes involved in cell migration. Among signaling pathways/genes involved in directionally persistent migration are, for example, vinculin, talin, FAK, p130CAS, and filamin A. Integrin receptors and the physical arrangement of adhesions assure orientation of the cytoskeleton while leading-edge protrusions can be stabilized by matrix orientation [137, 138]. When cancer cells experience an increase in ECM stiffness, they respond to the change by generating increased traction forces on their surroundings by regulating growth factor signaling and focal adhesion formation. For this purpose, the cell has several alternatives: for example, it can either force the network fibers apart and remodel the shape, form trails of variable caliber until it can pass through the pore, or the tumor cell degrades the fiber matrix via multistep pericellular proteolysis that was observed in individual and collective cancer cell migration [140]. Increased tumor tissue stiffness has been linked to tumor progression, direct stem cell differentiation, cell-cell and cell–matrix adhesion, hyaluronan synthesis, and expression of genes that play important roles in invasion and metastasis [128, 141, 142, 143]. A computational model was used to investigate the effect of ECM topography on vascular morphogenesis and explanation of mechanisms that control cell shape and orientation, sprout extension speeds, and sprout morphology. Sprout extension speed and morphology depend on matrix density, fiber network connectedness, and fiber orientation and varying matrix fiber density affect the likelihood of capillary sprout branching. The authors calculated optimal density for capillary network formation and suggested matrix heterogeneity as a mechanism for sprout branching. The density of the matrix fibers has a strong effect on the extension speed and the morphology of a new blood vessel pointing to new targets for pro- and anti-angiogenesis therapies [144].

Another important tumor characteristic is tumor growth-induced solid stress. As tumor cells proliferate they sequentially create new solid material (i.e. cells and matrix components) which pushes against the surrounding tumor microenvironment. Uncontrolled proliferation of cancer cells leads to ignorance of contact inhibition, their expansion imposes elastic tension on the surrounding tumor microenvironment, storing stress through the deformation of adaptable structures, and collapsing delicate structures, such as blood and lymphatic vessels. Interestingly, solid stress is accumulated within the tumor and is still sustained after the tumor excision [145]. Collagen and hyaluronan molecules are the main contributors of the ECM to solid stress. Collagen, as it becomes stiffer when stretched, is responsible for tensile stress. This observation is valid for both capsular and interstitial collagen. When hyaluronan resists compression, its negatively charged chains are pushed away, owing to electrostatic repulsion and trap water, therefore matrix becomes poorly compressible [145]. The compression of vessels by solid stress may create potential obstacles to drug delivery: the collapse of blood vessels hampers access to systemically administered drugs. This collapse might explain the fact that neoplasias with more ECM might be more resistant to treatment. For instance, chondrosarcomas, chordomas, or pancreatic ductal adenocarcinoma (the latter has the highest solid stress magnitude 7 kPa = 52.5 mmHg) are tumors rich in ECM and refractory to chemotherapy [146, 147, 148]. Further, the lack of lymphatic vessel function induces drainage compromise, leading to uniformly elevated interstitial fluid pressure. As a result, the transport of therapeutics, like antibodies and nanoparticles, is reduced because the dominant mode of transport becomes diffusion which is an inadequately slow process for large particles [149]. In this sense, decreasing solid stress by the angiotensin inhibitor, losartan, decompress tumor blood vessels, enhances drug delivery, and potentiates chemotherapy effects [150].

As stated above, endothelial activation is believed to be predominantly related to biochemical signals. However, mechanical forces have more recently also been demonstrated to regulate endothelial cell phenotype and function. Recent work has shown that mechanical forces control endothelial cell proliferation, survival, and migration [151, 152] and fluid shear stress from blood flow plays a critical role in regulating vessel morphogenesis, sprouting, and barrier function [153, 154]. To convert mechanical forces and biophysical signals into intracellular biochemical reaction cascades, endothelial cells employ a complex system of mechanosensors (actin cytoskeleton, integrins, cell-cell adhesion receptors, receptor tyrosine kinases, ion channels, and G-protein-coupled receptors) to sense and respond to mechanical forces [155]. Matrix stiffening enhanced integrin-mediated Rho/Rho-associated protein kinase (ROCK) activity and contraction in tumor epithelial and endothelial cells [156, 157, 158]. Tumor endothelial cells have abnormal mechanosensitivity to uniaxial cyclic strain transmitted through the ECM, which is mediated by vigorous regulation of Rho activity and cytoskeletal tension. Normal and tumor endothelial cells express similar levels of active β1 and β3 integrins [159]. Tumor endothelial cells demonstrate constitutively high baseline activity of Rho and ROCK, thicker stress fibers, higher adhesion strength, and augmented cytoskeletal tension. Logically, described features are mainly due to higher intrinsic Rho and ROCK-related cytoskeletal tension in the background of unchanged levels of integrins. These dynamics between normal and tumor endothelial cells in response to mechanical impulses suggest that the aberrant mechanical forces from the tumor microenvironment may cause tumor endothelial cells to gradually obtain an altered phenotype. Such alteration may further enable tumor endothelial cells to spread over a wider range of matrix stiffness [155, 158]. Specific integrins have been demonstrated to contribute to non-tumor and tumor angiogenesis. The expression of α1β1 and α2β1 integrins is upregulated by VEGF in endothelial cells [160], and the combined antagonism of α1β1 and α2β1 reduced human squamous cell carcinoma growth and angiogenesis [161]. The α5β1 integrin is selectively expressed in angiogenic vasculature. Upregulated αvβ3 and αvβ5 integrins in endothelial cells are necessary for the growth and survival facilitation of neovessels [162]. As already mentioned, αv integrins are also involved in cytokine-dependent pathways of angiogenesis. Integrin αvβ3 is incumbent in pathways activated by FGF or TNFα while integrin αvβ5 is necessary for angiogenic pathways activated by VEGF or TGFα [163]. Specifically, the αvβ5 integrin pathway downstream of VEGF causes activation of FAK and Src kinase [164]. The αvβ3 integrin has also been associated with VEGFR2 and the binding of αvβ3 to its corresponding ECM ligands has been shown to increase VEGF signaling [165]. Integrin αvβ3 is overexpressed in newly developed vasculature of mammary carcinoma [166], the expression level of αvβ3 and αvβ5 integrins in tumor neovessels were found to be associated with the neuroblastoma grade [167]. The experimental inhibition of αvβ3 integrin suppressed angiogenesis and related breast tumor growth in immunodeficient (SCID) mouse/human chimera [166] and resulted in tumor reduction in human clinical trials [168]. Combined inhibition of αvβ3 and αvβ5 integrins also significantly reduced growth of human melanoma xenografts in SCID mice [169]. Integrin α6β4 signaling has similarly been involved in incipient invasive phase of pathological angiogenesis. The β4 substrate domain promotes bFGF-mediated angiogenesis in matrigel plug assay and hypoxia-inducible factor VEGF-mediated angiogenesis in the retinal neovascularization model regulates sprouting angiogenesis by forced nuclear translocation of activated ERK and NF-κB in migrating endotheliocytes [170]. Furthermore, targeted deletion of the signaling domain of the integrin β4 significantly reduced the size and microvascular density in various tumors including melanoma, lung cancer, lymphoma, or fibrosarcoma [170]. These data demonstrate the role of cytoskeletal- and integrin-mediated mechanosensory pathways in facilitating tumor angiogenesis.

3.3 Hypoxia and interstitial fluid pressure

Hypoxia is another feature of the abnormal tumor microenvironment that is intrinsically linked to the formation of neovasculature and clinically manifests with metastatic progression and worse patient survival [171, 172]. Diffusion-limited hypoxia is a sequel of tumor cells located distantly from the blood-supplied areas. Such cells “suffer” from prolonged hypoxia and tumor cells are kept viable for hours to a few days in such environment [173]. Within the cell, hypoxia induces oncogenes, enhances DNA mutation chance, and selects for cells with increased apoptotic rate [171, 174]. Extracellularly, hypoxia supports tumor progression by increased matrix deposition, turnover, cross-linking, and remodeling [175]. HIF-1α increases vascularization in hypoxic areas and allows for the survival and proliferation of cancer cells, its inhibition prevents the expansion of neoplasia [176]. Along with known angiogenic factors, novel ones and their receptors include VEGF, VEGFR-1, -2, bFGF, platelet-derived growth factor B (PDGF), insulin-like growth factor II (IGF2), adrenomedullin, and epidermal growth factor (EGF) are targets of the HIF transcription factors. Several of these angiogenesis-related gene products, including iNOS, endothelin, adrenomedullin, and heme oxygenase 1, are also implicated in the modulation of local blood flow by regulating the vascular tone [177]. The well-known EMT activators such as Snail, Slug, and Twist are also induced by hypoxia [178]. Hypoxia also affects stem cells [179] that become pluripotent and aggressive with high metastatic potential. Resistance to anti-angiogenic therapy thus may be mediated by HIF-1α activated genes. Therapeutical targeting of hypoxia includes bioreductive prodrugs, HIF-1 targeting, and genetic engineering of anaerobic bacteria [180].

Abnormal metabolism in the tumor is further characterized by a decrease in extracellular pH. The known sources of H + ions in tumors are by- or end-products of anaerobic glycolysis, such as lactic acid and carbonic acid [181, 182]. The dysbalance between production and removal of H + ions lowers the extracellular pH in tumors. The level of pH also decreases in tumors with increasing distance from nearest blood vessels. Low extracellular pH causes stress-induced alteration of VEGF and IL-8 gene upregulation and relevant protein expression in three different tumor cells in vitro [183]. When the possible relationship between pH, pO2, and their effect on VEGF expression in vivo was examined using GFP imaging of tissues, pO2 and pH appear to regulate VEGF transcription in tumors independently. For example, in the hypoxic state or neutral pH, VEGF-promoter activity increased, with a decrease in pO2 and independent of pH. In decreased pH or oxygenated conditions, VEGF-promoter activity increased, with a decrease in pH and independent of pO2 [184]. To conclude, these key microenvironmental factors regulate angiogenic profiles in a complementary mode.

Another pathophysiologic feature of the tumor microenvironment is elevated interstitial fluid pressure (IFP) in the range of 10–100 mmHg [185, 186]. IFP of normal tissue is around zero [187]. The driving force in increasing tumor IFP is the tumor vasculature [188, 189]. In contrast to normal vessels which are characterized by dichotomous branching, tumor vasculature is chaotic, with trifurcations and branches with unsteady calibers, larger inter-endothelial junctions, multiple fenestrations, vesicles, vesico-vacuolar channels and a disruption of normal basement membrane [190]. Due to described ultrastructural alterations, vascular permeability in solid tumors is generally higher compared to normal counterparts. Tumors, also either lack lymphatics or the intratumoral vessels are non-functional [191], as a result, excess fluid accumulates in the interstitium resulted in elevated IFP. In IFP regulation model, fibroblasts actively regulate the tension applied to the ECM through integrins which enable fibroblasts to modify collagen fiber tension and modulate the elasticity of the ECM in response to hyaluronan and proteoglycan expansion. According to [192], interestingly, a significantly dense and stiffer collagen framework and related higher IFP is also a result of the synthesis of another important proteoglycan fibromodulin by stromal fibroblasts, which is mainly promoted by emerged inflammatory processes in malignant tumors. Interstitial fluid pressure may serve as another target for cancer therapy. Roh and colleagues [193] reported an inverse relationship between tumor IFP and degree of tissue oxygenation and suggested IFP’s role in predicting radiotherapy effect. Increased tumor IFP can also act as an obstacle to drug delivery, which makes questionable their efficacy. Several studies have also demonstrated advanced amelioration of chemotherapeutics following a reduction in tumor IFP [150].

Advertisement

4. Conclusions

The extracellular matrix in non-tumor states regulates tissue development and homeostasis, and its deregulation imparts to neoplasia and its progression. It serves not only as the mechanical milieu upon which cells/tissues inhabit but creates and exerts critical biochemical and biomechanical messages that drive cell growth, survival, differentiation, migration, and manage neoangiogenesis and immune scaffold. The cellular mechanisms inducing both angiogenesis and immunosuppressive responses are often reached by the same cell types and soluble factors. Studies point out that combinatorial strategies toward many potential targets with emphasis on angiogenesis should be adapted as a useful therapeutic approach to hinder/reverse tumor progression.

References

  1. 1. Ayoubi S, Dunn IF, Al-Mefty O. 31—Meningiomas. In: Kaye AH, Laws ER, editors. Brain Tumors. 3rd ed. London: W.B.Saunders; 2012. pp. 600-629. ISBN 9780443069673
  2. 2. Conti CJ. 14.16 Mechanisms of tumor progression. In: McQueen CA, editor. Comprehensive Toxicology. 2nd ed. Oxford, England: Elsevier; 2010. pp. 335-347. ISBN 9780080468846,2
  3. 3. Liotta LA, Kohn EC. The microenvironment of the tumour-host interface. Nature. 2001;411(6835):375-379
  4. 4. Anderson NM, Simon MC. The tumor microenvironment. Current Biology. 2020;30(16):R921-Rr25
  5. 5. Erler JT, Weaver VM. Three-dimensional context regulation of metastasis. Clinical & Experimental Metastasis. 2009;26(1):35-49
  6. 6. Järveläinen H, Sainio A, Koulu M, et al. Extracellular matrix molecules: Potential targets in pharmacotherapy. Pharmacological Reviews. 2009;61(2):198-223
  7. 7. Schulz S. C-type natriuretic peptide and guanylyl cyclase B receptor. Peptides. 2005;26(6):1024-1034
  8. 8. Mescher AL, Mescher AL, Junqueira LCU. Junqueira's Basic Histology: Text and Atlas. 14th ed. New York: McGraw-Hill Education; 2016
  9. 9. Ricard-Blum S. The collagen family. Cold Spring Harbor Perspectives in Biology. 2011;3(1):a004978
  10. 10. Heino J. The collagen family members as cell adhesion proteins. BioEssays. 2007;29(10):1001-1010
  11. 11. Ricard-Blum S, Ruggiero F. The collagen superfamily: From the extracellular matrix to the cell membrane. Pathologie Biologie. 2005;53(7):430-442
  12. 12. Mander, Lewis N, Liu, Hung-wen. Comprehensive Natural Products II: Chemistry and Biology. Lewis Mander, Hung-Wen (Ben) Liu. The Netherlands: Elsevier Amsterdam; 2010
  13. 13. Fleischmajer R, Perlish JS, Burgeson RE, et al. Type I and type III collagen interactions during fibrillogenesis. Annals of the New York Academy of Sciences. 1990;580:161-175
  14. 14. Niyibizi C, Eyre DR. Bone type V collagen: Chain composition and location of a trypsin cleavage site. Connective Tissue Research. 1989;20(1-4):247-250
  15. 15. Mayne R. Cartilage collagens. What is their function, and are they involved in articular disease? Arthritis and Rheumatism. 1989;32(3):241-246
  16. 16. Bourgot I, Primac I, Louis T, et al. Reciprocal interplay between fibrillar collagens and collagen-binding integrins: Implications in cancer progression and metastasis. Frontiers in Oncology. 2020;10:1488
  17. 17. Twardowski T, Fertala A, Orgel JP, et al. Type I collagen and collagen mimetics as angiogenesis promoting superpolymers. Current Pharmaceutical Design. 2007;13(35):3608-3621
  18. 18. Mukwaya A, Jensen L, Lagali N. Relapse of pathological angiogenesis: Functional role of the basement membrane and potential treatment strategies. Experimental & Molecular Medicine. 2021;53(2):189-201
  19. 19. Mancuso MR, Davis R, Norberg SM, et al. Rapid vascular regrowth in tumors after reversal of VEGF inhibition. The Journal of Clinical Investigation. 2006;116(10):2610-2621
  20. 20. Wu Y, Ge G. Complexity of type IV collagens: From network assembly to function. Biological Chemistry. 2019;400(5):565-574
  21. 21. Khoshnoodi J, Pedchenko V, Hudson BG. Mammalian collagen IV. Microscopy Research and Technique. 2008;71(5):357-370
  22. 22. Kisling A, Lust RM, Katwa LC. What is the role of peptide fragments of collagen I and IV in health and disease? Life Sciences. 2019;228:30-34
  23. 23. Ricard-Blum S, Vallet SD. Fragments generated upon extracellular matrix remodeling: Biological regulators and potential drugs. Matrix Biology. 2019;75-76:170-189
  24. 24. Aikio M, Alahuhta I, Nurmenniemi S, et al. Arresten, a collagen-derived angiogenesis inhibitor, suppresses invasion of squamous cell carcinoma. PLoS One. 2012;7(12):e51044
  25. 25. Colorado PC, Torre A, Kamphaus G, et al. Anti-angiogenic cues from vascular basement membrane collagen. Cancer Research. 2000;60(9):2520-2526
  26. 26. Williamson SC, Metcalf RL, Trapani F, et al. Vasculogenic mimicry in small cell lung cancer. Nature Communications. 2016;7:13322
  27. 27. Chiablaem K, Lirdprapamongkol K, Keeratichamroen S, et al. Curcumin suppresses vasculogenic mimicry capacity of hepatocellular carcinoma cells through STAT3 and PI3K/AKT inhibition. Anticancer Research. 2014;34(4):1857-1864
  28. 28. Chen YS, Chen ZP. Vasculogenic mimicry: A novel target for glioma therapy. Chinese Journal of Cancer. 2014;33(2):74-79
  29. 29. Ma Y, Wu T, Zhou H, et al. Canstatin represses glioma growth by inhibiting formation of VM-like structures. Translational Neuroscience. 2021;12(1):309-319
  30. 30. Vautrin-Glabik A, Devy J, Bour C, et al. Angiogenesis inhibition by a short 13 amino acid peptide sequence of tetrastatin, the α4(IV) NC1 domain of collagen IV. Frontiers in Cell and Development Biology. 2020;8:775
  31. 31. Karagiannis ED, Popel AS. A systematic methodology for proteome-wide identification of peptides inhibiting the proliferation and migration of endothelial cells. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(37):13775-13780
  32. 32. Karagiannis ED, Popel AS. Identification of novel short peptides derived from the alpha 4, alpha 5, and alpha 6 fibrils of type IV collagen with anti-angiogenic properties. Biochemical and Biophysical Research Communications. 2007;354(2):434-439
  33. 33. Rhodes JM, Simons M. The extracellular matrix and blood vessel formation: Not just a scaffold. Journal of Cellular and Molecular Medicine. 2007;11(2):176-205
  34. 34. Baud S, Duca L, Bochicchio B, et al. Elastin peptides in aging and pathological conditions. Biomolecular Concepts. 2013;4(1):65-76
  35. 35. Duca L, Floquet N, Alix AJ, et al. Elastin as a matrikine. Critical Reviews in Oncology/Hematology. 2004;49(3):235-244
  36. 36. Salesse S, Odoul L, Chazée L, et al. Elastin molecular aging promotes MDA-MB-231 breast cancer cell invasiveness. FEBS Open Bio. 2018;8(9):1395-1404
  37. 37. Robinet A, Fahem A, Cauchard JH, et al. Elastin-derived peptides enhance angiogenesis by promoting endothelial cell migration and tubulogenesis through upregulation of MT1-MMP. Journal of Cell Science. 2005;118(Pt 2):343-356
  38. 38. Taylor KR, Gallo RL. Glycosaminoglycans and their proteoglycans: Host-associated molecular patterns for initiation and modulation of inflammation. The FASEB Journal. 2006;20(1):9-22
  39. 39. Khoshnoodi J, Cartailler JP, Alvares K, et al. Molecular recognition in the assembly of collagens: Terminal noncollagenous domains are key recognition modules in the formation of triple helical protomers. The Journal of Biological Chemistry. 2006;281(50):38117-38121
  40. 40. Chen CG, Gubbiotti MA, Kapoor A, et al. Autophagic degradation of HAS2 in endothelial cells: A novel mechanism to regulate angiogenesis. Matrix Biology. 2020;90:1-19
  41. 41. Heldin P, Lin CY, Kolliopoulos C, et al. Regulation of hyaluronan biosynthesis and clinical impact of excessive hyaluronan production. Matrix Biology. 2019;78-79:100-117
  42. 42. Tolg C, Yuan H, Flynn SM, et al. Hyaluronan modulates growth factor induced mammary gland branching in a size dependent manner. Matrix Biology. 2017;63:117-132
  43. 43. Soldevila-Barreda JJ, Romero-Canelón I, Habtemariam A, et al. Transfer hydrogenation catalysis in cells as a new approach to anticancer drug design. Nature Communications. 2015;6: 6582
  44. 44. Tammi MI, Oikari S, Pasonen-Seppänen S, et al. Activated hyaluronan metabolism in the tumor matrix—Causes and consequences. Matrix Biology. 2019;78-79:147-164
  45. 45. Bourguignon LYW, Earle C, Shiina M. Hyaluronan-CD44 interaction promotes HPV 16 E6 oncogene-mediated oropharyngeal cell carcinoma survival and chemoresistance. Matrix Biology. 2019;78-79:180-200
  46. 46. Karalis TT, Heldin P, Vynios DH, et al. Tumor-suppressive functions of 4-MU on breast cancer cells of different ER status: Regulation of hyaluronan/HAS2/CD44 and specific matrix effectors. Matrix Biology. 2019;78-79:118-138
  47. 47. Chanmee T, Ontong P, Itano N. Hyaluronan: A modulator of the tumor microenvironment. Cancer Letters. 2016;375(1):20-30
  48. 48. Bernert B, Porsch H, Heldin P. Hyaluronan synthase 2 (HAS2) promotes breast cancer cell invasion by suppression of tissue metalloproteinase inhibitor 1 (TIMP-1). Journal of Biological Chemistry. 2011;286(49):42349-42359
  49. 49. Caon I, Bartolini B, Parnigoni A, et al. Revisiting the hallmarks of cancer: The role of hyaluronan. Seminars in Cancer Biology. 2020;62:9-19
  50. 50. Kim YH, Lee SB, Shim S, et al. Hyaluronic acid synthase 2 promotes malignant phenotypes of colorectal cancer cells through transforming growth factor beta signaling. Cancer Science. 2019;110(7):2226-2236
  51. 51. Goyal A, Gubbiotti MA, Chery DR, et al. Endorepellin-evoked autophagy contributes to angiostasis. Journal of Biological Chemistry. 2016;291(37):19245-19256
  52. 52. Bix G, Castello R, Burrows M, et al. Endorepellin in vivo: Targeting the tumor vasculature and retarding cancer growth and metabolism. Journal of the National Cancer Institute. 2006;98(22):1634-1646
  53. 53. Folkman J. Tumor suppression by p53 is mediated in part by the antiangiogenic activity of endostatin and tumstatin. Science's STKE. 2006;2006(354):pe35
  54. 54. Mantovani A, Marchesi F, Malesci A, et al. Tumour-associated macrophages as treatment targets in oncology. Nature Reviews Clinical Oncology. 2017;14(7):399-416
  55. 55. Spinelli FM, Vitale DL, Icardi A, et al. Hyaluronan preconditioning of monocytes/macrophages affects their angiogenic behavior and regulation of TSG-6 expression in a tumor type-specific manner. The FEBS Journal. 2019;286(17):3433-3449
  56. 56. Sokolowska M, Chen LY, Eberlein M, et al. Low molecular weight hyaluronan activates cytosolic phospholipase A2α and eicosanoid production in monocytes and macrophages. The Journal of Biological Chemistry. 2014;289(7):4470-4488
  57. 57. Rayahin JE, Buhrman JS, Zhang Y, et al. High and low molecular weight hyaluronic acid differentially influence macrophage activation. ACS Biomaterials Science & Engineering. 2015;1(7):481-493
  58. 58. Cattaruzza S, Perris R. Proteoglycan control of cell movement during wound healing and cancer spreading. Matrix Biology. 2005;24(6):400-417
  59. 59. Murdoch AD, Iozzo RV. Prokaryotic expression of proteoglycans. Methods in Molecular Biology. 2001;171:231-238
  60. 60. Iozzo RV, Murdoch AD. Proteoglycans of the extracellular environment: Clues from the gene and protein side offer novel perspectives in molecular diversity and function. The FASEB Journal. 1996;10(5):598-614
  61. 61. Iozzo RV. The family of the small leucine-rich proteoglycans: Key regulators of matrix assembly and cellular growth. Critical Reviews in Biochemistry and Molecular Biology. 1997;32(2):141-174
  62. 62. Chen S, Young MF, Chakravarti S, et al. Interclass small leucine-rich repeat proteoglycan interactions regulate collagen fibrillogenesis and corneal stromal assembly. Matrix Biology. 2014;35:103-111
  63. 63. Reese SP, Underwood CJ, Weiss JA. Effects of decorin proteoglycan on fibrillogenesis, ultrastructure, and mechanics of type I collagen gels. Matrix Biology. 2013;32(7-8):414-423
  64. 64. Schaefer L, Iozzo RV. Small leucine-rich proteoglycans, at the crossroad of cancer growth and inflammation. Current Opinion in Genetics & Development. 2012;22(1):56-57
  65. 65. Järveläinen H, Sainio A, Wight TN. Pivotal role for decorin in angiogenesis. Matrix Biology. 2015;43:15-26
  66. 66. Goyal A, Neill T, Owens RT, et al. Decorin activates AMPK, an energy sensor kinase, to induce autophagy in endothelial cells. Matrix Biology. 2014;34:46-54
  67. 67. Neill T, Schaefer L, Iozzo RV. Decorin: A guardian from the matrix. The American Journal of Pathology. 2012;181(2):380-387
  68. 68. Santra M, Reed CC, Iozzo RV. Decorin binds to a narrow region of the epidermal growth factor (EGF) receptor, partially overlapping but distinct from the EGF-binding epitope. The Journal of Biological Chemistry. 2002;277(38):35671-35681
  69. 69. Schlessinger J. Ligand-induced, receptor-mediated dimerization and activation of EGF receptor. Cell. 2002;110(6):669-672
  70. 70. Zhu JX, Goldoni S, Bix G, et al. Decorin evokes protracted internalization and degradation of the epidermal growth factor receptor via caveolar endocytosis. The Journal of Biological Chemistry. 2005;280(37):32468-32479
  71. 71. Moreth K, Iozzo RV, Schaefer L. Small leucine-rich proteoglycans orchestrate receptor crosstalk during inflammation. Cell Cycle. 2012;11(11):2084-2091
  72. 72. Buraschi S, Pal N, Tyler-Rubinstein N, et al. Decorin antagonizes Met receptor activity and down-regulates {beta}-catenin and Myc levels. The Journal of Biological Chemistry. 2010;285(53):42075-42085
  73. 73. Danilkovitch-Miagkova A, Miagkov A, Skeel A, et al. Oncogenic mutants of RON and MET receptor tyrosine kinases cause activation of the beta-catenin pathway. Molecular and Cellular Biology. 2001;21(17):5857-5868
  74. 74. Kharaishvili G, Simkova D, Makharoblidze E, et al. Wnt signaling in prostate development and carcinogenesis. Biomedical Papers of the Medical Faculty of the University Palacky, Olomouc, Czech Republic. 2011;155(1):11-18
  75. 75. Rasola A, Fassetta M, De Bacco F, et al. A positive feedback loop between hepatocyte growth factor receptor and beta-catenin sustains colorectal cancer cell invasive growth. Oncogene. 2007;26(7):1078-1087
  76. 76. Kasprzak A. Angiogenesis-related functions of Wnt signaling in colorectal carcinogenesis. Cancers. 2020;12(12):3601
  77. 77. Asano K, Nelson CM, Nandadasa S, et al. Stromal versican regulates tumor growth by promoting angiogenesis. Scientific Reports. 2017;7(1):17225
  78. 78. Ricciardelli C, Russell DL, Ween MP, et al. Formation of hyaluronan- and versican-rich pericellular matrix by prostate cancer cells promotes cell motility. The Journal of Biological Chemistry. 2007;282(14):10814-10825
  79. 79. Wight TN. Versican: A versatile extracellular matrix proteoglycan in cell biology. Current Opinion in Cell Biology. 2002;14(5):617-623
  80. 80. Wight TN, Kinsella MG, Evanko SP, et al. Versican and the regulation of cell phenotype in disease. Biochimica et Biophysica Acta. 2014;1840(8):2441-2451
  81. 81. Nandadasa S, Foulcer S, Apte SS. The multiple, complex roles of versican and its proteolytic turnover by ADAMTS proteases during embryogenesis. Matrix Biology. 2014;35:34-41
  82. 82. Yang W, Yee AJ. Versican V2 isoform enhances angiogenesis by regulating endothelial cell activities and fibronectin expression. FEBS Letters. 2013;587(2):185-192
  83. 83. Lin H, Wilson JE, Roberts CR, et al. Biglycan, decorin, and versican protein expression patterns in coronary arteriopathy of human cardiac allograft: Distinctness as compared to native atherosclerosis. The Journal of Heart and Lung Transplantation. 1996;15(12):1233-1247
  84. 84. Dutt S, Kléber M, Matasci M, et al. Versican V0 and V1 guide migratory neural crest cells. The Journal of Biological Chemistry. 2006;281(17):12123-12131
  85. 85. Yamagata M, Kimata K. Repression of a malignant cell-substratum adhesion phenotype by inhibiting the production of the anti-adhesive proteoglycan, PG-M/versican. Journal of Cell Science. 1994;107(Pt 9):2581-2590
  86. 86. Paulus W, Baur I, Dours-Zimmermann MT, et al. Differential expression of versican isoforms in brain tumors. Journal of Neuropathology and Experimental Neurology. 1996;55(5):528-533
  87. 87. Koyama H, Hibi T, Isogai Z, et al. Hyperproduction of hyaluronan in neu-induced mammary tumor accelerates angiogenesis through stromal cell recruitment: Possible involvement of versican/PG-M. The American Journal of Pathology. 2007;170(3):1086-1099
  88. 88. Masui T, Hosotani R, Tsuji S, et al. Expression of METH-1 and METH-2 in pancreatic cancer. Clinical Cancer Research. 2001;7(11):3437-3443
  89. 89. Fernández-Rodríguez R, Rodríguez-Baena FJ, Martino-Echarri E, et al. Stroma-derived but not tumor ADAMTS1 is a main driver of tumor growth and metastasis. Oncotarget. 2016;7(23):34507-34519
  90. 90. Vázquez F, Hastings G, Ortega MA, et al. METH-1, a human ortholog of ADAMTS-1, and METH-2 are members of a new family of proteins with angio-inhibitory activity. The Journal of Biological Chemistry. 1999;274(33):23349-23357
  91. 91. Reynolds LE, Watson AR, Baker M, et al. Tumour angiogenesis is reduced in the Tc1 mouse model of Down's syndrome. Nature. 2010;465(7299):813-817
  92. 92. Casal C, Torres-Collado AX, Plaza-Calonge Mdel C, et al. ADAMTS1 contributes to the acquisition of an endothelial-like phenotype in plastic tumor cells. Cancer Research. 2010;70(11):4676-4686
  93. 93. Ricciardelli C, Frewin KM, Tan Ide A, et al. The ADAMTS1 protease gene is required for mammary tumor growth and metastasis. The American Journal of Pathology. 2011;179(6):3075-3085
  94. 94. Lu X, Wang Q , Hu G, et al. ADAMTS1 and MMP1 proteolytically engage EGF-like ligands in an osteolytic signaling cascade for bone metastasis. Genes & Development. 2009;23(16):1882-1894
  95. 95. Lima MA, Dos Santos L, Turri JA, et al. Prognostic value of ADAMTS proteases and their substrates in epithelial ovarian cancer. Pathobiology. 2016;83(6):316-326
  96. 96. Henry SP, Takanosu M, Boyd TC, et al. Expression pattern and gene characterization of asporin. A newly discovered member of the leucine-rich repeat protein family. The Journal of Biological Chemistry. 2001;276(15):12212-12221
  97. 97. Lorenzo P, Aspberg A, Onnerfjord P, et al. Identification and characterization of asporin. A novel member of the leucine-rich repeat protein family closely related to decorin and biglycan. The Journal of Biological Chemistry. 2001;276(15):12201-12211
  98. 98. Nakajima M, Kizawa H, Saitoh M, et al. Mechanisms for asporin function and regulation in articular cartilage. The Journal of Biological Chemistry. 2007;282(44):32185-32192
  99. 99. Kalamajski S, Aspberg A, Lindblom K, et al. Asporin competes with decorin for collagen binding, binds calcium and promotes osteoblast collagen mineralization. The Biochemical Journal. 2009;423(1):53-59
  100. 100. Turashvili G, Bouchal J, Baumforth K, et al. Novel markers for differentiation of lobular and ductal invasive breast carcinomas by laser microdissection and microarray analysis. BMC Cancer. 2007;7:55
  101. 101. Simkova D, Kharaishvili G, Slabakova E, et al. Glycoprotein asporin as a novel player in tumour microenvironment and cancer progression. Biomedical Papers of the Medical Faculty of the University Palacky, Olomouc, Czech Republic. 2016;160(4):467-473
  102. 102. Simkova D, Kharaishvili G, Korinkova G, et al. The dual role of asporin in breast cancer progression. Oncotarget. 2016;7(32):52045-52060
  103. 103. Turtoi A, Musmeci D, Wang Y, et al. Identification of novel accessible proteins bearing diagnostic and therapeutic potential in human pancreatic ductal adenocarcinoma. Journal of Proteome Research. 2011;10(9):4302-4313
  104. 104. Orr B, Riddick AC, Stewart GD, et al. Identification of stromally expressed molecules in the prostate by tag-profiling of cancer-associated fibroblasts, normal fibroblasts and fetal prostate. Oncogene. 2012;31(9):1130-1142
  105. 105. Zhan S, Li J, Ge W. Multifaceted roles of asporin in cancer: Current understanding. Frontiers in Oncology. 2019;9:948
  106. 106. Sasaki T, Fässler R, Hohenester E. Laminin: The crux of basement membrane assembly. The Journal of Cell Biology. 2004;164(7):959-963
  107. 107. Patarroyo M, Tryggvason K, Virtanen I. Laminin isoforms in tumor invasion, angiogenesis and metastasis. Seminars in Cancer Biology. 2002;12(3):197-207
  108. 108. Dixelius J, Jakobsson L, Genersch E, et al. Laminin-1 promotes angiogenesis in synergy with fibroblast growth factor by distinct regulation of the gene and protein expression profile in endothelial cells. The Journal of Biological Chemistry. 2004;279(22):23766-23772
  109. 109. Doi M, Thyboll J, Kortesmaa J, et al. Production, purification, and migration-promoting activity on vascular endothelial cells. The Journal of Biological Chemistry. 2002;277(15):12741-12748
  110. 110. Jeon H, Ono M, Kumagai C, et al. Pericytes from microvessel fragment produce type IV collagen and multiple laminin isoforms. Bioscience, Biotechnology, and Biochemistry. 1996;60(5):856-861
  111. 111. Lugassy C, Shahsafaei A, Bonitz P, et al. Tumor microvessels in melanoma express the beta-2 chain of laminin. Implications for melanoma metastasis. Journal of Cutaneous Pathology. 1999;26(5):222-226
  112. 112. Lugassy C, Eyden BP, Christensen L, et al. Angio-tumoral complex in human malignant melanoma characterised by free laminin: Ultrastructural and immunohistochemical observations. Journal of Submicroscopic Cytology and Pathology. 1997;29(1):19-28
  113. 113. Miner JH, Patton BL, Lentz SI, et al. The laminin alpha chains: Expression, developmental transitions, and chromosomal locations of alpha1-5, identification of heterotrimeric laminins 8-11, and cloning of a novel alpha3 isoform. The Journal of Cell Biology. 1997;137(3):685-701
  114. 114. Paku S, Döme B, Tóth R, et al. Organ-specificity of the extravasation process: An ultrastructural study. Clinical & Experimental Metastasis. 2000;18(6):481-492
  115. 115. Kariya Y, Mori T, Yasuda C, et al. Localization of laminin alpha3B chain in vascular and epithelial basement membranes of normal human tissues and its down-regulation in skin cancers. Journal of Molecular Histology. 2008;39(4):435-446
  116. 116. Mao Y, Schwarzbauer JE. Fibronectin fibrillogenesis, a cell-mediated matrix assembly process. Matrix Biology. 2005;24(6):389-399
  117. 117. Vouret-Craviari V, Boulter E, Grall D, et al. ILK is required for the assembly of matrix-forming adhesions and capillary morphogenesis in endothelial cells. Journal of Cell Science. 2004;117(Pt 19):4559-4569
  118. 118. Gerhardt H, Golding M, Fruttiger M, et al. VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. The Journal of Cell Biology. 2003;161(6):1163-1177
  119. 119. Uemura A, Kusuhara S, Wiegand SJ, et al. Tlx acts as a proangiogenic switch by regulating extracellular assembly of fibronectin matrices in retinal astrocytes. The Journal of Clinical Investigation. 2006;116(2):369-377
  120. 120. Bazigou E, Xie S, Chen C, et al. Integrin-alpha9 is required for fibronectin matrix assembly during lymphatic valve morphogenesis. Developmental Cell. 2009;17(2):175-186
  121. 121. Garmy-Susini B, Avraamides CJ, Schmid MC, et al. Integrin alpha4beta1 signaling is required for lymphangiogenesis and tumor metastasis. Cancer Research. 2010;70(8):3042-3051
  122. 122. Van Obberghen-Schilling E, Tucker RP, Saupe F, et al. Fibronectin and tenascin-C: Accomplices in vascular morphogenesis during development and tumor growth. The International Journal of Developmental Biology. 2011;55(4-5):511-525
  123. 123. Astrof S, Hynes RO. Fibronectins in vascular morphogenesis. Angiogenesis. 2009;12(2):165-175
  124. 124. Neri D, Bicknell R. Tumour vascular targeting. Nature Reviews. Cancer. 2005;5(6):436-446
  125. 125. Soung YH, Chung J. Curcumin inhibition of the functional interaction between integrin α6β4 and the epidermal growth factor receptor. Molecular Cancer Therapeutics. 2011;10(5):883-891
  126. 126. Desgrosellier JS, Cheresh DA. Integrins in cancer: Biological implications and therapeutic opportunities. Nature Reviews. Cancer. 2010;10(1):9-22
  127. 127. Zucker S, Hymowitz M, Conner CE, et al. Rapid trafficking of membrane type 1-matrix metalloproteinase to the cell surface regulates progelatinase a activation. Laboratory Investigation. 2002;82(12):1673-1684
  128. 128. Folgueras AR, Pendás AM, Sánchez LM, et al. Matrix metalloproteinases in cancer: From new functions to improved inhibition strategies. The International Journal of Developmental Biology. 2004;48(5-6):411-424
  129. 129. Mañes S, Mira E, Barbacid MM, et al. Identification of insulin-like growth factor-binding protein-1 as a potential physiological substrate for human stromelysin-3. The Journal of Biological Chemistry. 1997;272(41):25706-25712
  130. 130. Yu Q , Stamenkovic I. Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-beta and promotes tumor invasion and angiogenesis. Genes & Development. 2000;14(2):163-176
  131. 131. Deryugina EI, Quigley JP. Tumor angiogenesis: MMP-mediated induction of intravasation-and metastasis-sustaining neovasculature. Matrix Biology. 2015;44-46:94-112
  132. 132. Sounni NE, Devy L, Hajitou A, et al. MT1-MMP expression promotes tumor growth and angiogenesis through an up-regulation of vascular endothelial growth factor expression. The FASEB Journal. 2002;16(6):555-564
  133. 133. Sounni NE, Roghi C, Chabottaux V, et al. Up-regulation of vascular endothelial growth factor-A by active membrane-type 1 matrix metalloproteinase through activation of Src-tyrosine kinases. The Journal of Biological Chemistry. 2004;279(14):13564-13574
  134. 134. Chetty C, Lakka SS, Bhoopathi P, et al. MMP-2 alters VEGF expression via alphaVbeta3 integrin-mediated PI3K/AKT signaling in A549 lung cancer cells. International Journal of Cancer. 2010;127(5):1081-1095
  135. 135. Kudo Y, Iizuka S, Yoshida M, et al. Matrix metalloproteinase-13 (MMP-13) directly and indirectly promotes tumor angiogenesis. The Journal of Biological Chemistry. 2012;287(46):38716-38728
  136. 136. Sang QX. Complex role of matrix metalloproteinases in angiogenesis. Cell Research. 1998;8(3):171-177
  137. 137. Brábek J, Mierke CT, Rösel D, et al. The role of the tissue microenvironment in the regulation of cancer cell motility and invasion. Cell Communication and Signaling: CCS. 2010;8:22
  138. 138. Petrie RJ, Doyle AD, Yamada KM. Random versus directionally persistent cell migration. Nature Reviews. Molecular Cell Biology. 2009;10(8):538-549
  139. 139. Kraning-Rush CM, Reinhart-King CA. Controlling matrix stiffness and topography for the study of tumor cell migration. Cell Adhesion & Migration. 2012;6(3):274-279
  140. 140. Kharaishvili G, Simkova D, Bouchalova, et al. The role of cancer-associated fibroblasts, solid stress and other microenvironmental factors in tumor progression and therapy resistance. Cancer Cell International. 2014;14:41
  141. 141. Levental KR, Yu H, Kass L, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009;139(5):891-906
  142. 142. Palumbo A Jr, Meireles Da Costa N, et al. Esophageal cancer development: Crucial clues arising from the extracellular matrix. Cell. 2020;9(2):455
  143. 143. Reinhart-King CA. How matrix properties control the self-assembly and maintenance of tissues. Annals of Biomedical Engineering. 2011;39(7):1849-1856
  144. 144. Bauer AL, Jackson TL, Jiang Y. Topography of extracellular matrix mediates vascular morphogenesis and migration speeds in angiogenesis. PLoS Computational Biology. 2009;5(7):e1000445
  145. 145. Stylianopoulos T, Martin JD, Chauhan VP, et al. Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(38):15101-15108
  146. 146. Provenzano PP, Cuevas C, Chang AE, et al. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21(3):418-429
  147. 147. Olive KP, Jacobetz MA, Davidson CJ, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324(5933):1457-1461
  148. 148. Chauhan VP, Martin JD, Liu H, et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nature Communications. 2013;4:2516
  149. 149. Honn KV, Tang DG. Adhesion molecules and tumor cell interaction with endothelium and subendothelial matrix. Cancer Metastasis Reviews. 1992;11(3-4):353-375
  150. 150. Horino Y, Takahashi S, Miura T, et al. Prolonged hypoxia accelerates the posttranscriptional process of collagen synthesis in cultured fibroblasts. Life Sciences. 2002;71(26):3031-3045
  151. 151. Li S, Huang NF, Hsu S. Mechanotransduction in endothelial cell migration. Journal of Cellular Biochemistry. 2005;96(6):1110-1126
  152. 152. Li YS, Haga JH, Chien S. Molecular basis of the effects of shear stress on vascular endothelial cells. Journal of Biomechanics. 2005;38(10):1949-1971
  153. 153. Kutys ML, Chen CS. Forces and mechanotransduction in 3D vascular biology. Current Opinion in Cell Biology. 2016;42:73-79
  154. 154. Conway DE, Breckenridge MT, Hinde E, et al. Fluid shear stress on endothelial cells modulates mechanical tension across VE-cadherin and PECAM-1. Current Biology. 2013;23(11):1024-1030
  155. 155. Zanotelli MR, Reinhart-King CA. Mechanical forces in tumor angiogenesis. Advances in Experimental Medicine and Biology. 2018;1092:91-112
  156. 156. Paszek MJ, Zahir N, Johnson KR, et al. Tensional homeostasis and the malignant phenotype. Cancer Cell. 2005;8(3):241-254
  157. 157. Paszek MJ, Weaver VM. The tension mounts: Mechanics meets morphogenesis and malignancy. Journal of Mammary Gland Biology and Neoplasia. 2004;9(4):325-342
  158. 158. Ghosh K, Thodeti CK, Dudley AC, et al. Tumor-derived endothelial cells exhibit aberrant Rho-mediated mechanosensing and abnormal angiogenesis in vitro. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(32):11305-11310
  159. 159. Tzima E, del Pozo MA, Shattil SJ, et al. Activation of integrins in endothelial cells by fluid shear stress mediates Rho-dependent cytoskeletal alignment. The EMBO Journal. 2001;20(17):4639-4647
  160. 160. Senger DR, Claffey KP, Benes JE, et al. Angiogenesis promoted by vascular endothelial growth factor: Regulation through alpha1beta1 and alpha2beta1 integrins. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(25):13612-13617
  161. 161. Senger DR, Perruzzi CA, Streit M, et al. The alpha(1)beta(1) and alpha(2)beta(1) integrins provide critical support for vascular endothelial growth factor signaling, endothelial cell migration, and tumor angiogenesis. The American Journal of Pathology. 2002;160(1):195-204
  162. 162. Weis SM, Cheresh DA. αV integrins in angiogenesis and cancer. Cold Spring Harbor Perspectives in Medicine. 2011;1(1):a006478
  163. 163. Friedlander M, Brooks PC, Shaffer RW, et al. Definition of two angiogenic pathways by distinct alpha v integrins. Science. 1995;270(5241):1500-1502
  164. 164. Hood JD, Frausto R, Kiosses WB, et al. Differential alphav integrin-mediated Ras-ERK signaling during two pathways of angiogenesis. The Journal of Cell Biology. 2003;162(5):933-943
  165. 165. Soldi R, Mitola S, Strasly M, et al. Role of alphavbeta3 integrin in the activation of vascular endothelial growth factor receptor-2. The EMBO Journal. 1999;18(4):882-892
  166. 166. Brooks PC, Strömblad S, Klemke R, et al. Antiintegrin alpha v beta 3 blocks human breast cancer growth and angiogenesis in human skin. The Journal of Clinical Investigation. 1995;96(4):1815-1822
  167. 167. Erdreich-Epstein A, Shimada H, Groshen S, et al. Integrins alpha(v)beta3 and alpha(v)beta5 are expressed by endothelium of high-risk neuroblastoma and their inhibition is associated with increased endogenous ceramide. Cancer Research. 2000;60(3):712-721
  168. 168. Eliceiri BP, Cheresh DA. The role of alphav integrins during angiogenesis: Insights into potential mechanisms of action and clinical development. The Journal of Clinical Investigation. 1999;103(9):1227-1230
  169. 169. Kumar CC, Malkowski M, Yin Z, et al. Inhibition of angiogenesis and tumor growth by SCH221153, a dual alpha(v)beta3 and alpha(v)beta5 integrin receptor antagonist. Cancer Research. 2001;61(5):2232-2238
  170. 170. Nikolopoulos SN, Blaikie P, Yoshioka T, et al. Integrin beta4 signaling promotes tumor angiogenesis. Cancer Cell. 2004;6(5):471-483
  171. 171. Lunt SJ, Chaudary N, Hill RP. The tumor microenvironment and metastatic disease. Clinical & Experimental Metastasis. 2009;26(1):19-34
  172. 172. Grum-Schwensen B, Klingelhofer J, Berg CH, et al. Suppression of tumor development and metastasis formation in mice lacking the S100A4(mts1) gene. Cancer Research. 2005;65(9):3772-3780
  173. 173. Sutherland RM, Franko AJ. On the nature of the radiobiologically hypoxic fraction in tumors. International Journal of Radiation Oncology, Biology, Physics. 1980;6(1):117-120
  174. 174. Duff MD, Mestre J, Maddali S, et al. Analysis of gene expression in the tumor-associated macrophage. The Journal of Surgical Research. 2007;142(1):119-128
  175. 175. Helmlinger G, Sckell A, Dellian M, et al. Acid production in glycolysis-impaired tumors provides new insights into tumor metabolism. Clinical Cancer Research. 2002;8(4):1284-1291
  176. 176. Schauer IG, Sood AK, Mok S, et al. Cancer-associated fibroblasts and their putative role in potentiating the initiation and development of epithelial ovarian cancer. Neoplasia. 2011;13(5):393-405
  177. 177. Bagley RG. Endosialin: From vascular target to biomarker for human sarcomas. Biomarkers in Medicine. 2009;3(5):589-604
  178. 178. Sun S, Ning X, Zhang Y, et al. Hypoxia-inducible factor-1alpha induces twist expression in tubular epithelial cells subjected to hypoxia, leading to epithelial-to-mesenchymal transition. Kidney International. 2009;75(12):1278-1287
  179. 179. Keith B, Simon MC. Hypoxia-inducible factors, stem cells, and cancer. Cell. 2007;129(3):465-472
  180. 180. Sun JD, Liu Q , Wang J, et al. Selective tumor hypoxia targeting by hypoxia-activated prodrug TH-302 inhibits tumor growth in preclinical models of cancer. Clinical Cancer Research. 2012;18(3):758-770
  181. 181. Garin-Chesa P, Old LJ, Rettig WJ. Cell surface glycoprotein of reactive stromal fibroblasts as a potential antibody target in human epithelial cancers. Proceedings of the National Academy of Sciences of the United States of America. 1990;87(18):7235-7239
  182. 182. Pouysségur J, Mechta-Grigoriou F. Redox regulation of the hypoxia-inducible factor. Biological Chemistry. 2006;387(10-11):1337-1346
  183. 183. Xu L, Fukumura D, Jain RK. Acidic extracellular pH induces vascular endothelial growth factor (VEGF) in human glioblastoma cells via ERK1/2 MAPK signaling pathway: Mechanism of low pH-induced VEGF. The Journal of Biological Chemistry. 2002;277(13):11368-11374
  184. 184. Fukumura D, Xu L, Chen Y, et al. Hypoxia and acidosis independently up-regulate vascular endothelial growth factor transcription in brain tumors in vivo. Cancer Research. 2001;61(16):6020-6024
  185. 185. Nathanson SD, Nelson L. Interstitial fluid pressure in breast cancer, benign breast conditions, and breast parenchyma. Annals of Surgical Oncology. 1994;1(4):333-338
  186. 186. Milosevic M, Fyles A, Hedley D, et al. Interstitial fluid pressure predicts survival in patients with cervix cancer independent of clinical prognostic factors and tumor oxygen measurements. Cancer Research. 2001;61(17):6400-6405
  187. 187. Erler JT, Cawthorne CJ, Williams KJ, et al. Hypoxia-mediated down-regulation of Bid and Bax in tumors occurs via hypoxia-inducible factor 1-dependent and -independent mechanisms and contributes to drug resistance. Molecular and Cellular Biology. 2004;24(7):2875-2889
  188. 188. Lunt SJ, Kalliomaki TM, Brown A, et al. Interstitial fluid pressure, vascularity and metastasis in ectopic, orthotopic and spontaneous tumours. BMC Cancer. 2008;8:2
  189. 189. Jain RK, Tong RT, Munn LL. Effect of vascular normalization by antiangiogenic therapy on interstitial hypertension, peritumor edema, and lymphatic metastasis: Insights from a mathematical model. Cancer Research. 2007;67(6):2729-2735
  190. 190. Winkler F, Kozin SV, Tong RT, et al. Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: Role of oxygenation, angiopoietin-1, and matrix metalloproteinases. Cancer Cell. 2004;6(6):553-563
  191. 191. Padera TP, Kadambi A, di Tomaso E, et al. Lymphatic metastasis in the absence of functional intratumor lymphatics. Science. 2002;296(5574):1883-1886
  192. 192. Oldberg A, Kalamajski S, Salnikov AV, et al. Collagen-binding proteoglycan fibromodulin can determine stroma matrix structure and fluid balance in experimental carcinoma. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(35):13966-13971
  193. 193. Roh HD, Boucher Y, Kalnicki S, et al. Interstitial hypertension in carcinoma of uterine cervix in patients: Possible correlation with tumor oxygenation and radiation response. Cancer Research. 1991;51(24):6695-6698

Written By

Gvantsa Kharaishvili

Submitted: 14 March 2022 Reviewed: 24 March 2022 Published: 25 May 2022