insidiouschangesinstromalmatrixfuelcancerprogression...review...

17
Review Insidious Changes in Stromal Matrix Fuel Cancer Progression Fayth L. Miles 1,2 and Robert A. Sikes 2,3 Abstract Reciprocal interactions between tumor and stromal cells propel cancer progression and metastasis. A complete un- derstanding of the complex contributions of the tumor stroma to cancer progression necessitates a careful examination of the extracellular matrix (ECM), which is largely synthesized and modulated by cancer-associated broblasts. This structurally supportive meshwork serves as a signaling scaffold for a myriad of biologic processes and responses favoring tumor progression. The ECM is a repository for growth factors and cytokines that promote tumor growth, proliferation, and metastasis through diverse interactions with soluble and insoluble ECM components. Growth factors activated by proteases are involved in the initiation of cell signaling pathways essential to invasion and survival. Various transmembrane proteins produced by the cancer stroma bind the collagen and bronectin-rich matrix to induce proliferation, adhesion, and migration of cancer cells, as well as protease activation. Integrins are critical liaisons between tumor cells and the surrounding stroma, and with their mechano-sensing ability, induce cell signaling pathways associated with contractility and migration. Proteoglycans also bind and interact with various matrix proteins in the tumor microenvironment to promote cancer progression. Together, these components function to mediate cross-talk between tumor cells and broblasts ultimately to promote tumor survival and metastasis. These stromal factors, which may be expressed differentially according to cancer stage, have prognostic utility and potential. This review examines changes in the ECM of cancer-associated broblasts induced through carcinogenesis, and the impact of these changes on cancer progression. The implication is that cancer progression, even in epithelial cancers, may be based in large part on changes in signaling from cancer-associated stromal cells. These changes may provide early prognostic indicators to further stratify patients during treatment or alter the timing of their follow-up visits and observations. Visual Overview: http://mcr.aacrjournals.org/content/12/3/297/F1.large.jpg. Mol Cancer Res; 12(3); 297312. Ó2014 AACR. Introduction The ECM has a major role in tumor development and progression. Over the last decade, it has become much clearer that reciprocal interactions between tumor and stro- mal cells determine the course of cancer progression. Although much remains to be understood about the chro- nology of events dictating tumor initiation and the context for reciprocal regulatory signals, it is clear that continuous bidirectional paracrine signaling establishes a microenviron- ment conducive for the survival of the tumor. Hence, in the study of the tumor stromal microenvironment, there are two sides to be examined: (i) the effects of tumor cell signaling on the surrounding stroma and (ii) the inuence of paracrine stromal signals on tumor cells. In general, the reciprocal relationship supporting tumor progression might be viewed as follows: tumor cells secrete cytokines, chemokines, and enzymes that activate stromal cells and broblasts. Among a diverse group of molecules, these stromal cells secrete pro- teases that break down the tumor cell basement membrane. Consequently, growth factors are released from the under- lying matrix, which not only initiate the signaling pathways in tumor cells, but also activate broblasts further, inducing broblast secretion of a host of ECM factors responsible for regulating numerous interrelated events. Tumor cells thrive in this convoluted but nourishing milieu, which ultimately promotes their survival, proliferation, and metastasis (Fig. 1). Cancer associated broblasts (CAF) have a profound role on ECM composition and dynamics. The ECM is composed of brillar and structural proteins, proteoglycans, integrins and proteases, all of which may be manufactured by CAFs. Hence, CAFs potentially can modulate levels and activities of all these factors. Indeed, various CAF-secreted soluble factors and proteolytic enzymes modulate the tumor microenviron- ment, altering composition of the connective tissue through remodeling. Because the ECM controls the availability and activation of growth factors and serves as a platform for integrin and growth factor receptors to regulate cell signaling Authors' Afliations: 1 Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, California; 2 Depart- ment of Biological Sciences, Laboratory for Cancer Ontogeny and Ther- apeutics; and 3 Center for Translational Cancer Research, University of Delaware, Newark, Delaware Corresponding Author: Robert A. Sikes, Center for Translational Cancer Research, University of Delaware, 326 Wolf Hall, Biology, Newark, DE 19716. Phone: 302-831-6050; Fax: 302-831-2281; E-mail: [email protected] doi: 10.1158/1541-7786.MCR-13-0535 Ó2014 American Association for Cancer Research. Molecular Cancer Research www.aacrjournals.org 297 on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Upload: others

Post on 04-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

Review

InsidiousChanges inStromalMatrix FuelCancerProgression

Fayth L. Miles1,2 and Robert A. Sikes2,3

AbstractReciprocal interactions between tumor and stromal cells propel cancer progression and metastasis. A complete un-

derstanding of the complex contributions of the tumor stroma to cancer progression necessitates a careful examination ofthe extracellular matrix (ECM), which is largely synthesized and modulated by cancer-associated fibroblasts. Thisstructurally supportive meshwork serves as a signaling scaffold for a myriad of biologic processes and responses favoringtumor progression. The ECM is a repository for growth factors and cytokines that promote tumor growth, proliferation,and metastasis through diverse interactions with soluble and insoluble ECM components. Growth factors activatedby proteases are involved in the initiation of cell signaling pathways essential to invasion and survival. Varioustransmembrane proteins produced by the cancer stroma bind the collagen and fibronectin-rich matrix to induceproliferation, adhesion, andmigration of cancer cells, as well as protease activation. Integrins are critical liaisons betweentumor cells and the surrounding stroma, and with their mechano-sensing ability, induce cell signaling pathwaysassociated with contractility and migration. Proteoglycans also bind and interact with various matrix proteins in thetumor microenvironment to promote cancer progression. Together, these components function to mediate cross-talkbetween tumor cells and fibroblasts ultimately to promote tumor survival and metastasis. These stromal factors, whichmay be expressed differentially according to cancer stage, have prognostic utility and potential. This review examineschanges in the ECM of cancer-associated fibroblasts induced through carcinogenesis, and the impact of these changeson cancer progression. The implication is that cancer progression, even in epithelial cancers, may be based in largepart on changes in signaling from cancer-associated stromal cells. These changesmay provide early prognostic indicatorsto further stratify patients during treatment or alter the timing of their follow-up visits and observations.

Visual Overview: http://mcr.aacrjournals.org/content/12/3/297/F1.large.jpg.Mol Cancer Res; 12(3); 297–312. �2014 AACR.

IntroductionThe ECM has a major role in tumor development and

progression. Over the last decade, it has become muchclearer that reciprocal interactions between tumor and stro-mal cells determine the course of cancer progression.Although much remains to be understood about the chro-nology of events dictating tumor initiation and the contextfor reciprocal regulatory signals, it is clear that continuousbidirectional paracrine signaling establishes a microenviron-ment conducive for the survival of the tumor. Hence, in thestudy of the tumor stromal microenvironment, there are twosides to be examined: (i) the effects of tumor cell signaling onthe surrounding stroma and (ii) the influence of paracrine

stromal signals on tumor cells. In general, the reciprocalrelationship supporting tumor progression might be viewedas follows: tumor cells secrete cytokines, chemokines, andenzymes that activate stromal cells and fibroblasts. Among adiverse group of molecules, these stromal cells secrete pro-teases that break down the tumor cell basement membrane.Consequently, growth factors are released from the under-lying matrix, which not only initiate the signaling pathwaysin tumor cells, but also activate fibroblasts further, inducingfibroblast secretion of a host of ECM factors responsible forregulating numerous interrelated events. Tumor cells thrivein this convoluted but nourishing milieu, which ultimatelypromotes their survival, proliferation, and metastasis(Fig. 1).Cancer associated fibroblasts (CAF) have a profound role

on ECMcomposition and dynamics. The ECM is composedof fibrillar and structural proteins, proteoglycans, integrinsand proteases, all of which may be manufactured by CAFs.Hence, CAFs potentially canmodulate levels and activities ofall these factors. Indeed, various CAF-secreted soluble factorsand proteolytic enzymes modulate the tumor microenviron-ment, altering composition of the connective tissue throughremodeling. Because the ECM controls the availability andactivation of growth factors and serves as a platform forintegrin and growth factor receptors to regulate cell signaling

Authors' Affiliations: 1Department of Epidemiology, Fielding School ofPublic Health, University of California, Los Angeles, California; 2Depart-ment of Biological Sciences, Laboratory for Cancer Ontogeny and Ther-apeutics; and 3Center for Translational Cancer Research, University ofDelaware, Newark, Delaware

Corresponding Author: Robert A. Sikes, Center for Translational CancerResearch, University of Delaware, 326 Wolf Hall, Biology, Newark, DE19716. Phone: 302-831-6050; Fax: 302-831-2281; E-mail:[email protected]

doi: 10.1158/1541-7786.MCR-13-0535

�2014 American Association for Cancer Research.

MolecularCancer

Research

www.aacrjournals.org 297

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 2: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

pathways, fibroblasts modulate a multiplicity of growthfactor-mediated pathways through changes in synthesis ofmatrix components.

Growth FactorsParacrine growth factors secreted by CAFs are central to

the establishment of a microenvironment conducive totumor growth and progression. Growth factors functionat various stages through tumor progression, includingproliferation, metastasis, and dissemination. Such factors,particularly heparin-binding molecules, have the ability to

induce a number of physiologic events leading to tumorprogression because of their diverse interactions with theECM. In the ECM, they may be sequestered or copresentedby other molecules, such as proteoglycans and various cellsurface receptors, accounting for their biologic diversity.The prolific contributions of stromal cell-derived factor 1

(SDF-1) to cancer progression in the tumor microenviron-ment are linked largely to its role in recruitment of mesen-chymal stem cells andmyofibroblasts to the tumor, as SDF-1is integral to CAF activation and the desmoplastic reaction.In addition to cancer cells, SDF-1 is largely secreted by CAFsand mesenchymal cells (1, 2) where it stimulates growth,proliferation, invasion, angiogenesis, metastasis, and colo-nization in a number of cancers. It is noteworthy that bonemarrow–derived CAFs secrete higher levels of SDF-1 thanwild-type myofibroblasts (3), highlighting the particularimportance of bone marrow–niche cells in cancer progres-sion. SDF-1 is an important mediator of cross-talk betweentumor cells and fibroblasts. Paracrine stromal SDF-1 signal-ing is stimulated in the presence of TGF-b1, which increasesexpression of epithelial CXCR4, resulting in SDF-1–medi-ated Akt activation and subsequent tumor progression andsurvival (4). In addition, SDF-1 promotes cell–cell and cell–matrix adhesions by regulating integrin expression (5, 6).TGF-b1 plays a major role in activation of fibroblasts by

promoting fibroblast to myofibroblast transdifferentiation.Furthermore, TGF-b1 is actively secreted by CAFs andstromal cells. TGF-b1 is notoriously a complex, pleiotropicfactor because of multiple binding partners and associatednoncanonical signaling pathways. Consequently, it plays arole in both tumor suppression and tumor promotion.Although loss of TGF-b signaling through abrogation ofTbRII in stroma has been shown to correlate with increasedproliferation and progression of cancer cells (7–9), paracrineTGF-b1 signaling frequently induces metastasis and epithe-lial to mesenchymal transformation of cancer cells. Suchdiverse actions of TGF-b seem to be dependent on contex-tual differences that include the tissue localization, cellsource, extracellular mediators, and the cell signaling con-text. We have found that although direct stimulation ofprostate cancer cells with TGF-b1 suppresses growth andinvasion (10), stimulation of bonemarrow stromal cells withTGF-b1 suppresses paracrine apoptotic signals to promoteprostate cancer cell survival (Fig. 2). Similarly, overexpres-sion of TGF-b1 in normal prostate fibroblasts inducesinvasive tumor growth (11). Stromal TGF-b is associatedwith tumor progression and metastasis in breast cancer, andhigh stromal expression is correlated with increased mortal-ity (12). In addition, orthotopic mammary carcinoma mod-els have demonstrated a role for TGF-b in regulating tumorperfusion, as abrogation of TGF-b signaling in the stromaincreases recruitment of perivascular cells, thus improvingchemotherapeutic efficacy (13).TGF-b, among other factors, is an important regulator of

insulin-like growth factor (IGF) and the IGF axis, anothervery important signaling system in prostate cancer progres-sion. In prostate stromal cells, TGF-b1 increases the expres-sion of IGF-1 and IGF-binding protein 3 (IGFBP-3; refs. 14,

Figure 1. Reciprocal tumor: stromal cell signaling in the tumormicroenvironment promotes tumor progression and stromal cellactivation. A. Paracrine signals (soluble factors) from the tumor inducealterations that prime the stroma. B. Subsequent release of stromalproteases triggers breakdown of the tumor basement membrane (BM)and release of additional growth factors from tumor cells, furtheractivating the stroma. C. Consequently, stromal cells secrete amyriad offactors such as growth factors, proteoglycans, proteases, and insolubleproteins to promote tumor growth, migration, andmetastasis. During theprocess, cancer associate fibroblasts (CAFs) may be transformed.

Miles and Sikes

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research298

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 3: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

15), as well as the ratio of IGF-I bound to IGFBP-3, therebydecreasing growth of prostate cancer cells in coculture (14).Such regulation of IGF and binding protein levels providesone explanation for the divergent responses of TGF-b inprostate cancer. Prospective and case–control studies havefound that elevated plasma IGF-1 predicts an increasedprostate cancer risk of several fold (16, 17), and elevatedsera levels correlate with an increased risk for breast andcolorectal cancer, although not statistically significant (18–20). However, increased IGFBP-1 seems to be significantlyassociated with decreased risk for colorectal cancer (OR ¼0.48; 95% confidence interval, 0.23–1.00, P ¼ 0.02;ref. 21). IGF-2 may also play a role in prostate and breastcancer, where it is expressed at higher levels by mammarystromal cells (22, 23), and in particularly large quantities bystromal cells in benign prostatic hyperplasia (24). Althoughthe prognostic significance of total stromal IGF-2 levels inbreast cancer is inconclusive, patients with lower serum levelsof free IGF-2 have larger malignant tumors (25). Interest-ingly, the presence of integrin-a11 on fibroblasts is associ-atedwith increased stromal IGF-2 levels in vivo in non–smallcell lung carcinoma (NSCLC), concomitant with anenhanced rate of tumor growth (26).Similar to other heparin-binding growth factors, platelet-

derived growth factor (PDGF), expressed by leukocytes andtumor cells, promotes angiogenesis, likely through upregu-lating stromal production of VEGF (27), andmay also have amore direct effect through stimulation of endothelial cells

(28). AlthoughCAFs do not commonly express PDGF, theyreadily express PDGF receptor to promote angiogenesis andproliferation, which involves upregulation of fibroblastgrowth factor (FGF)-2 and FGF-7 in cervical cancer (29,30). Inhibition of PDGF decreased stromal heparin-bindingEGF (HB-EGF)-mediated tumor cell growth in coculturesof cervical cancer cells and CAFs from patient tumor tissues,highlighting the significance of PDGF in HB-EGF signal-ing, and cancer progression (31). In addition to cervicalCAFs,HB-EGF has been found to be expressed at high levelson prostate stromal cells (32, 33), where paracrine signalsmediate prostate cancer survival and androgen-independenttumor growth as well as neuroendocrine differentiation (34).The effects of PDGF on cancer cell proliferation andprogression occur largely through EGFR/ErbB signalingleading to activation of mitogen—activated protein kinase(MAPK) and PI3K/Akt. Similarly, hepatocyte growth factor(HGF) activates the RAS/MAPK and phosphoinositide 3-kinase (PI3K) pathways in addition to many others, bybinding c-MET, another receptor tyrosine kinase with ahighly complex signaling profile (35). CAF-secreted HGFpromotes invasiveness of several cancers, including breast,colon, and oral squamous cell carcinoma (36–38). Curious-ly, all HGF signaling in cancer cells may not be mediatedthrough c-MET. Tate and colleagues clearly demonstrate aprostate cancer cell response to HGF in a c-MET-nullbackground, thereby opening up the possibility of signalingthrough cell surface nucleolin (39). Thus, heparin-binding

Figure 2. TGF-b pretreatment ofbone marrow stromal cellsreverses stromal cell conditionedmedia (CM)-induced death ofprostate cancer cells. HS-5 bonemarrow stromal cells werepretreated with or without TGF-b1for 4 days in DMEM supplementedwith 10% FBS. Following PBSwashes, fresh serum-free mediumwas replaced and conditionedmedium was collected after 48hours incubation. C4-2 prostatecancer cells were treated withDMEM, HS-5 CM with vehicle, orHS-5 CM with exogenously addedTGF-b1. A live/dead assay wasperformed using calcein AM andethidium homodimer at aconcentration of 125 nmol/L and2 mmol/L, respectively, todifferentially label live cells fromdead C4-2 cells. Representativeimages are shown from five fieldsper experiment run three timesminimum. Color images, green(calcein AM) and red (ethidiumhomodimer) were converted tograyscale and inverted inphotoshop to produce black andwhite images. Images werecaptured using a Nikon 2000TE.Final magnification �25.

Stromal Paracrine Factors Promote Cancer Progression

www.aacrjournals.org Mol Cancer Res; 12(3) March 2014 299

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 4: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

growth factor-activated tyrosine kinases engage the ECMthrough focal adhesion complexes and associated down-stream signaling cascades, ultimately inducing a variety ofresponses to promote tumorigenesis and cancer progression.

Collagen, Fibronectin, and TenascinCAFs secrete higher levels of collagens than those of

normal tissue. It is well known that deposition of a densecollagen matrix is a major feature of the desmoplasticreaction. This generates a stiff collagen matrix, which isassociated with increased activity of the matrix cross-linkingenzyme lysyl-oxidase (40). Increased collagen density hasbeen shown to promote proliferation of epithelial cells inthree-dimensional (3D) matrices, and tumor formation ofbreast cancer cells, demonstrating the role of a collagen-dense environment in tumor initiation and progression. Theconsequence of enhanced collagen deposition is increasedcell signaling pathways that promote cancer cell invasion,migration, and collagen reorganization. Isoform-specificexpression of collagen may be dependent on tumor typeand stage. Collagen I and III are upregulated commonly byCAFs or stromal cells in several cancers (41–45). In geneexpression studies, collagens X and XI-a1 were increased inthe reactive cancer stroma of invasive breast tumors, forminga prognostic signature (46, 47), although protein levels ofcollagen XI-a1 were shown to be downregulated in malig-nant breast cancer in a separate analysis (48).Upregulation ofcollagens IV, VI, XI, and XV, and downregulation ofcollagen XIV also have been observed in CAFs during theCAF–epithelial interaction in various cancers, in some casesforming a prognostic gene signature (49–52).Carcinomas may be characterized by several variants of

fibronectin or oncofetal fibronectin, which are present in thetumor stroma and expressed by myofibroblasts. Oncofetalisoforms, including ED-A andED-B fibronectin, are derivedby alternative splicing. Because of the varied specializedprotein-binding domains, fibronectin isoforms may beunique in their affinity and specificity for ECM-bindingpartners, which include fibrin, collagen, heparin, tenascin,syndecan, and integrins. Oncofetal fibronectin is associatedwith an invasive phenotype, poor prognosis, or poor differ-entiation in many cancers, as demonstrated in CAF:tumorspheroids as well as human tissues (53–56). ED-B fibro-nectin is an angiogenic marker that is involved in neovascu-larization in advanced and remodeling tumors, and thus hasa role in dissemination of cancer cells (57). Migrationstimulating factor (MSF), a truncated isoform of oncofetalfibronectin containing the gelatin-binding domain, also issecreted by both CAFs and oncofetal fibroblasts and inducesmigration (58). The migratory response of fibroblasts toMSF is based on their ability to adhere to native collagentype I. Because of this MSF-induced migratory response, amicroenvironment is established for the manifestation of aninvasive phenotype in cancer cells (58). Overexpressionof MSF is correlated with poor survival of patients withcancer (56).The dynamic role of tenascin-C in the tumor microen-

vironment stems from its ability to interact with a diverse

group of ECM molecules and surface receptors that initiatecell signaling pathways. Tenascin-C is resident in the stromaof many poorly differentiated tumors, expressed highly byfibroblasts (37, 59–61), and induced by a variety of signalingpathways. In earlier studies, it was proposed to be a stromalmarker for mammary tumors because it was not detected innormal tissue (60), and also was found to be elevated in thesera of patients with cancer (62). Now, it is quite evident thattenascin-C has a prominent role in cancer—promotingangiogenesis, tumor cell proliferation, and metastasis, withlevels increasing with tumor aggressiveness (61). Recently, itwas identified as one among 11 analytes capable of differ-entiating between benign andmalignant ovarian cancer (63).In a mouse model of breast cancer, it was shown thatsecretion of tenascin-C by stromal cells expressing s100AF,a marker associated with metastasis and poor prognosis,promotes metastatic colonization of breast cancer (64).Tenascin-C interacts with several integrins expressed on thesurface of tumor cells and fibroblasts (65, 66). It also bindsfibronectin and interferes with fibronectin-mediated adhe-sion, competing with the syndecan-4–binding site (67, 68).Its antiadhesive properties affect proliferation (68) andstimulate cancer cell migration by promoting disassemblyof focal adhesions, thus interfering with integrin-mediatedadhesions. Tenascin-C also was shown to inhibit activationof RhoA and associated stress fiber formation, and to inducefilopodial extensions (69). In particular, alternatively splicedfibronectin type III repeats are associated with increasedmotility and invasion (70). Tenascin-W, although not ascommon as tenascin-C, has been shown to be a marker foractivated tumor stroma, and is associated with increasedmigration of breast cancer cells (71). Recently, it was foundlocalized around the vasculature of tumor cells, and wasshown to stimulate angiogenic activity in vitro (72). Hence,Tenascin-W may have further utility as a marker for tumorangiogenesis.

ProteasesProteolysis regulates and induces release of latent growth

factors stored in the matrix, unveils protein domains, andgenerates bioactive fragments. Cleavage of the ECM bymatrix metalloproteinases (MMP) and other proteases leadsto activation of growth factors, as well as cytoskeletal reor-ganization and modification of downstream signaling path-ways. Accordingly, VEGF and basic fibroblast growth factormay be released and activated to promote angiogenesis (73,74). EGF receptor (EGFR) ligands, such as HB-EGF, alsocan be processed by MMPs (75), along with IGF-bindingproteins (76, 77), which regulate the activity and bioavail-ability of IGF (78), along with ligand independent effects ofIGF-BP fragments (79).Numerous MMPs are expressed and secreted by CAFs,

and enable cancer cells to evade tissue boundaries, escape theprimary site, and metastasize. The significance of fibroblastsin synthesis of MMPs has become appreciated only recently,as MMP expression was assessed previously within theconfines of the tumor. However, it is clearer now thatstromal cells are the major source of MMPs, which become

Miles and Sikes

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research300

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 5: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

active at the invasive front (80). Several coculture models oftumor cells and fibroblasts have demonstrated enhancedactivation of MMP-2 or expression of MT1-MMP, anactivator of proMMP-2 (gelatinase; refs. 81–83). In vivomodels also have shown the role of stromal MT1-MMP orMMP-2 in tumor cell growth (84, 85). Notably, introduc-tion of MMP-2-null fibroblasts reduced head and necksquamous cell carcinoma (HNSCC) tumor volume in severecombined immunodeficient (SCID) mice by 90% andabrogated invasion of collagen gels in coculture. MMP-2expression in CAFs has been highlighted as an indicator ofpoor prognosis forNSCLC (86); and, in the prostate,MMP-2 is associated with an invasive phenotype (87, 88). In theprostate, stromal MMP-2 levels are upregulated by TGF-b1(89). The significance of this may reside in the clinicalobservation that high-serum TGF-b levels are associatedwith poor patient survival and increased bonemetastasis (90,91). It has been suggested that proMMP2 is activated inCAFs as a consequence of collagen I binding of integrin b1.Interestingly, b1 integrin is activated by MT1-MMP–induced cleavage of the a-V integrin precursor, promotingcollagen binding and motility of cancer cells, thus providinga potential mechanism for MT1-MMP activation of MMP-2 (81, 92).The unique set of MMPs expressed in the tumor stroma

varies with tumor type and stage, and consequently, iscontext dependent. Accordingly, in a squamous cell carci-noma model of benign and malignant HaCaT clones trans-planted onto nude mice, expression of stromal-derivedMMPs and their correlation with malignancy were exam-ined, and stage-specific differences identified. mRNA ofMMP-2, -9, -13, and -14 was upregulated in the stromaof malignant and invasive tumors, and MMP-3 wasexpressed exclusively in stroma representing very late-stagedisease (93). During progression of breast cancer, MMP-9,-13, and -11 (stromelysin-3) are upregulated in reactivestroma, and have implications in invasion and osteolysis,with MMP-11 also being implicated in mammary tumordevelopment (47, 94–97). MMP-11 and -13 are associatedwith tumor progression, invasion, and poor prognosis inseveral other cancers (98–102), where they may be expressedby both stromal and tumor cells.Other stromal-relevant MMPs are ADAM proteins, of

which ADAM-17, -9, and -12 are expressed by myofibro-blasts or stromal cells in colon cancer, liver metastases, andprostate cancer, respectively (103–105). These ADAM pro-teins have a role in cancer development, progression, orinvasion although the mechanisms have not been elucidatedthoroughly. Production of ADAMTS-1 by fibroblasts ischaracteristic of many carcinomas and correlates withenhanced secretion of TGF-b1 and interleukin (IL)-1b(106). It was shown recently that fibroblasts in coculturewith breast cancer cells upregulate ADAMTS to promoteinvasion in an epigenetic mechanism, likely throughdecreased promoter-associated histone methylation (107).Matrix metalloendopeptidase (CD10), is also expressed byCAFs, elevated in tumor stroma, and has prognostic value inNSCLC and breast cancer, although its precise role in cancer

is still being examined (51, 108–111). Among the MMPinhibitors, TIMP2 was shown to interfere with fibroblast-induced growth of cancer cells in a mouse model (112).Interestingly, however, TIMP along with plasmin, may helpto activate proMMP, and is released with MMP-2(81, 113, 114). TIMP-1 and TIMP-2 activity may besuppressed by cathepsin B, which is localized to fibroblastsand correlated with invasion and shorter patient survival(115, 116).Serine-proteases and their associated inhibitors are very

significant in the continuous loop of proteolytic activitiesthat ultimately promote tumor cell invasiveness. uPA systemcomponents pro-uPA, PAI-1, and PAI-2 are produced byfibroblasts and other stromal cells, and have an importantrole in tumor growth and metastasis. Although high levels ofPAI-1 are associated with tumor progression and poorprognosis, particularly for breast cancer (117, 118),increased PAI-2 has been correlated with better patientprognosis (119). Although both PAI-1 and -2 inhibit uPA,an activator of plasmin, and hence prevent ECM degrada-tion, PAI-1 and PAI-2 interact differently with members ofthe LDL receptor family. PAI-2 binds LDL receptors withlower affinity and consequently does not induce downstreamsignaling leading to cell proliferation, potentially explainingthe observation of decreased metastasis associated with highPAI-2 levels in cancer (120).Microarray studies have shown downregulation of the

protease inhibitor and growth suppressor, WAP-type four-disulfide core (WFDC1), in CAFs (121–123). Overexpres-sion of WFDC1 inhibits proliferation of cancer cells, andWFDC1 expression was shown to decrease with progressionof prostate cancer to metastatic disease (121). The observa-tion of downregulatedWFDC1 in a diverse group of cancershighlights the potential prognostic significance of thismolecule.

ProteoglycansLike other ECM components in the convoluted tumor

milieu, proteoglycans are implicated in many cell signalingpathways. This is largely due to their associated glycosami-noglycans, which bind and interact with a myriad of ECMproteins, macromolecules, cell adhesion molecules, andgrowth factors. An important aspect of proteoglycan bioac-tivity is proteolytic cleavage or shedding. Ectodomain shed-ding affects the biologic function of proteoglycans, alteringproliferation, adhesion, and migration responses. In addi-tion, proteolytic cleavage can yield antiangiogenic peptidefragments to facilitate dissemination (124–126).Syndecan-1 is expressed in the desmoplastic stroma or

cancer fibroblasts of breast, ovarian, head and neck, andother carcinomas (127–131). Its growth-promoting abilityhas been demonstrated in two-dimensional (2D) and 3Dcocultures, as well as knockout studies, particularly in breastcancer models. This growth-promoting function may beattributed to the cleaved, soluble form of syndecan-1 in somecancers (132), whereas the soluble ectodomain may beassociated with decreased proliferation and increased inva-siveness in breast and other cancers (133). Of note,

Stromal Paracrine Factors Promote Cancer Progression

www.aacrjournals.org Mol Cancer Res; 12(3) March 2014 301

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 6: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

immunohistochemical examination showed that syndecan-1, which was redistributed from the epithelium to thestroma, was more than 10-fold higher in aggressive breasttissue compared with normal tissue (134). Similarly, fibro-blast-secreted versican promotes tumor growth, migration,and metastasis in breast, ovarian, and prostate cancer (135–139), and is a product of paracrine TGF-b1 signaling in thelatter. Stromal versican is associated with relapse and pooreroutcome in patients with cancer (140–142). Glypican-1 and-3 are additional proteoglycans that may have roles in tumorpromotion, as they are overexpressed in a variety of carci-nomas or associated stromal fibroblasts (128, 143–145).On the other hand, decorin, expressed primarily by

myofibroblasts, has a tumor-suppressive role, reducingtumor growth and metastasis in murine xenograft models,downregulating EGFR (146). The decorin protein core,specifically, is tumor suppressive triple-negative breast can-cer, inducing expression of stromal genes involved in celladhesion and tumor suppression and suppressing genesinvolved in inflammation (147). Decreased expression ofdecorin is a biomarker for aggressive soft tissue tumors (148).Likewise, lumican, which is increased in the reactive stromaof primary tumors, was shown to inhibit invasion of met-astatic prostate cancer cells in vitro (149).Among the proteoglycans, perlecan is the most diverse in

its bioactivities as a consequence of the multifunctionalnature of its large protein core and heparan sulfate chains.Hence, it is not unexpected for the roles of perlecan to extendbeyond development and normal biologic processes into thetumor microenvironment (150). Perlecan is present at highlevels in the ECM and vascularized stroma of breast, colon,prostate, and other cancers (151). In metastatic melanomas,perlecan is increased up to 15-fold (152). It contributessignificantly to cancer cell proliferation, in part, throughcooperation with other growth factor pathways, such as FGF(153–155) and has additional domains that modulate angio-genic activity (156). Its ability to activate FAK highlightspotential involvement in other tumorigenic and prometa-static signaling pathways (157).The highly prevalent pericellular glycosaminoglycan hya-

luronan provides a hydration shelf to facilitate cell signalingand cancer progression. In addition to its receptor CD44, itinteracts with proteoglycans and growth factor complexes tofacilitate cell migration and metastasis. It has an importantrole in regulating inflammation, and can recruit tumor-associated macrophages, which are necessary for extravasa-tion (158). Increased stromal hyaluronan expression isassociated with poor prognosis and metastasis in breast,colon, prostate, ovarian, and lung cancer. It is clearly a vitalplayer in the stromal tumor microenvironment (159–162).

Transmembrane proteinsIntegrins, probably the most vital group of transmem-

brane proteins implicated in cancer progression, are integralcomponents of the ECM, serving as liaisons for transmittingextracellular signals. Integrins bind RGD and other speci-ficity sequences on fibronectin and tenascin, as well asproteoglycans, and growth factors. Integrin adhesions are

regulated by proteases, and integrins reciprocally activate andregulate MMPs. Because integrins form attachments withthe matrix and vasculature, they are imperative to tumor celldissemination and extravasation from the primary site (163).Integrin-a11 is localized to mesenchymal cells, and wasfound overexpressed in NSCLC, where it was shown to havea role in tumorigenicity and increased expression of IGF-2(26, 164). This was demonstrated after implantation ofintegrin-a11–deficient fibroblasts and NSCLC cells inSCID mice caused reduction of tumor volume, which wasrestored upon re-expression of a11 in knockout mouseembryonic fibroblasts. Similarly, a population of CAFs fromhepatocellular carcinoma liver tissue demonstrated signifi-cantly higher surface expression of integrin-a4 and lowerexpression of a2 when compared with nonneoplastic tissue(165). CAFs express several a-subunits, including aV anda5, which interact withmultiple b-subunits, and are capableof binding collagen, fibronectin, and soluble factors such asTGF-b1 (166–168). Integrin ligands, ICAM and VCAM,have been shown to be upregulated and form a prognosticgene signature in CAFs (51, 52). These molecules aresignificant largely because of their fundamental roles inconnecting the ECM and cytoskeleton, initiating intracel-lular signaling pathways necessary for adhesion and motility(see discussion below on mechanical modeling).Central to transmembrane proteins implicated in the

fibroblastic tumor microenvironment is fibroblast activationprotein (FAP), a serine protease with gelatinase activityexpressed by stromal cells in various cancers (169). Itincreases proliferation, adhesion, and migration of ovariancancer cells (170). When expressed in breast cancer cells, itpromoted tumor growth, increasing both the size andnumber of tumors (171, 172). However, it is downregulatedduring the transformation of melanocytes, and may have atumor-suppressive role in melanoma (173, 174). It is alsoelevated in kidney, gastric, basal cell, colon, and lungcarcinoma (175–179). Depletion of FAP-expressing cellsin the latter induced necrosis, and highlighted FAP as animmune suppressor (178). FAP interaction with integrin-a3b1 may facilitate tumor cell adhesion and invasion (180).Endosialin/TEM1 is a highly glycosylated integral mem-

brane receptor expressed predominantly by CAFs, althoughinitially characterized as a tumor endothelial marker (181,182). It has been demonstrated to bind ECM proteins suchas fibronectin and collagen, and enhances cell adhesion andmigration on these matrices. In addition, it was shown topromote activation of MMP-9 (183). In vivo experimentsshowed that deficiency of TEM1 decreased growth of lungcarcinoma and fibrosarcoma (184). Endosialin may beexpressed by a subset of pericytes, and play a role in tumorangiogenesis, but this has not been elucidated fully (182).Podoplanin is another integral transmembrane glycopro-

tein, involved in cell adhesion and the lymphatic vascularsystem. A receptor for selectins, it is recognized by theD2-40monoclonal antibody, which has allowed for detection ofthis protein in lymph nodes. Podoplanin has been shown tobe a very specific indicator for the detection of lymphaticinvasion in primary tumors (185). Podoplanin is expressed at

Miles and Sikes

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research302

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 7: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

higher levels in human vascular adventitial fibroblasts thanlung-derived fibroblasts. Podoplanin-positive vascular fibro-blasts demonstrated increased tumor formation, lymph nodeinfiltration, and metastasis relative to podoplanin-negativevascular fibroblasts, underscoring the significance of podo-planin inmetastasis (186). Podoplanin has prognostic utilityfor several cancers. In breast cancer, podoplanin expressionin CAFs correlated negatively with estrogen receptor status,and was associated with poor prognosis (187, 188). Podo-planin expression in CAFs also is associated with poorprognosis in lung (189) and esophageal cancer (190), butinterestingly, is a favorable prognostic indicator in colorectalcancer (191).Protease-activated receptors (PAR) have an important role

in tumor progression andmetastasis of prostate cancer. PAR-1 is expressed mainly by myofibroblasts in the reactivestroma of primary prostate cancer tissues, whereas PAR-2is expressed predominantly by prostate cancer cells. PAR-1was found to be increased, particularly, in the stroma of high-grade prostate cancers, and also was found in the endothe-lium and fibroblasts of the bone-reactive stroma (192). Inthe prostate, stromal PAR-1 may be upregulated byhuman glandular kallikrein-4 produced by prostate cancercells, triggering secretion of IL-6 from stroma, in turnpromoting cell proliferation and upregulation of kallikreinsin prostate cancer cells (193). The activation and subsequenttumor-promoting effects of PAR-1 in the prostate tumormicroenvironment provide a nice illustration of the signif-icance of reciprocal tumor:stromal signals during cancerprogression.

Secreted proteinsThe secreted ECM glycoprotein osteonectin or add

abbreviation after cysteine: (SPARC) is overexpressed instromal or tumor cells of many cancers (194–197). It wasshown to increase microvessel density in hepatocellularcarcinoma, and therefore may promote angiogenesis. It ishighly prevalent in bone, and is a chemoattractant forbone-metastasizing cancers such as breast and prostatecancer (195). Although it was antimetastatic when over-expressed in breast cancer cells (198), stromal expressionof osteonectin along with CD10 was shown to be aprognostic indicator of recurrence for ductal carcinomain situ (199). Interestingly, stromal and tumor-derivedosteonectin were shown to suppress metastasis of bladdercancer in a murine syngeneic model. Cocultures of bladdercancer and stromal cells demonstrated that osteonectinsuppressed the inflammatory phenotype of tumor-associ-ated macrophages and CAFs by inhibiting reactive oxygenspecies formation via p38-JNK-AP1 and NF-kB. Further-more, osteonectin correlated with decreased expression ofIL-6, SDF-1, VEGF, and TGF-b, and TNF-a in fibro-blasts (200). Consistent with a beneficial role for osteo-nectin, overexpression of osteonectin has been shown toincrease susceptibility of ovarian and colon cancer tochemo- and radiation therapy (201, 202).The related protein, osteopontin, is overexpressed inmany

cancers (203). It is particularly abundant in the stroma of

breast cancer, where it is associated with poor outcome, andskin fibroblasts, where it promotes preneoplastic and malig-nant tumor progression (204–206). Similar to osteonectin,osteopontin promotes prostate cancer growth and progres-sion. The finding of higher osteopontin and osteonectinexpression in bone-metastasizing human prostate cancertissue highlights the potential prognostic value of osteopon-tin and other bone-resident proteins in cancer (207, 208).Osteopontin levels are also higher in triple-negative breastcancers (209). Periostin is another secreted stromal proteinoverexpressed in many cancers, including prostate, breast,colon, ovarian, pancreatic, and melanoma stroma. It isparticularly important in the stromal reaction of cancer andfunctions at the tumor stromal interface (47, 210–213). Inthe prostate, it correlates with poor prognosis, and is report-edly elevated 9-fold compared with BPH (47, 214).Collectively, these proteins can bind collagen, fibronectin,tenascin, andmany integrins, and therefore play a potentiallysignificant role in matrix remodeling in the tumormicroenvironment.

Biophysical and mechanical modeling of the ECMThe study of tumor–stromal interactions requires appro-

priate model systems. Much progress has been made with invitromodels, as the shift from2D to 3Dmodels have allowedfor more physiologically relevant interactions—phenotypi-cally and morphologically approximating the in vivo micro-environment. Spheroids have been used to examine theseinteractions (215), as well as other 3D cocultures usinghydrogels and biologically relevant matrices (216–218),which are also useful in examining the mechanical aspectsof the ECM and related tumor responses. Cells displaydistinct responses on matrix proteins and fibers, as a reflec-tion of their physiologic environment, emphasizing theimportance ofmatrix selection in the study of the fibroblastictumor microenvironment.ECM tension and rigidity, which can be recapitulated in

vitro with mechanical loading and occur in vivo withenhanced matrix deposition and remodeling, influence stro-magenesis (219), and tumor and fibroblast responses (220,221). In vitro studies have demonstrated that fibroblasts in3D or loaded collagen gels are stressed, and this is reflected infibroblast phenotype and signaling events (222–224).Mechanical force triggers transcription of a-smooth muscleactin and collagen to promote differentiation of myofibro-blasts, with the latter requiring cooperation of TGF-b (225,226). These events are necessary for the invasion andproliferation of tumor cells (227, 228). In a mechanicalinvasion assay utilizing paramagnetic micro beads within acollagen/fibronectin matrix and a rotating rare earth mag-net, Menon and colleagues demonstrated that mechanicalforce applied to the matrix induced tumor cell invasion ina mechanism involving cofilin, and that fibronectin wasrequired for this mechanosensory response (229). Stretch-ing of fibronectin induces mechanical stress resulting ininitiation of various signaling pathways (220, 230). Theactivation state of integrins is determined by mechanicalinfluences, among other factors, and different integrin

Stromal Paracrine Factors Promote Cancer Progression

www.aacrjournals.org Mol Cancer Res; 12(3) March 2014 303

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 8: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

conformations bind to different surfaces of fibronectin. Inthe case of a5b1, this generates complexes of differentialbinding strengths, which ultimately will affect integrin-mediated signaling cascades (230–232), thus affectingtumor cell responses.Mechanosensing integrins also respond to matrix rigidity

or stiffness with enhanced signaling, activating the FAK,extracellular signal—regulated kinase, and RhoGTPasepathways (40, 233–235). This is associated with increasedcontractility and migration in tumor cells, along with reor-ganization and stretching of the matrix. Examination of thisresponse using breast cancer cells in a 3D collagen geldemonstrated radial realignment of collagen fibers (236),a characteristic effect of migrating cells.Additional studies examining the mechano-signaling

pathways mediated by integrins will help elucidatemechanisms of tumor cell progression. Mechanical forcealso is associated with activity of tenascin-C, which canlead to enhanced invasiveness of tumor cells (237), as wellas TGF-b, which has mechanoregulatory ability, stimu-lates matrix contraction, and is itself activated in responseto stress (238–240). Thus, it is the interconnected anddynamic network of external (mechanical) and intracel-lular signals that promotes cancer progression in thestromal tumor microenvironment.

Targeted ApproachesExperimental studies have generated quantitative infor-

mation about ECM molecules relevant to the fibroblastictumor microenvironment that ultimately must be translatedinto therapeutic targets or clinically useful prognostic indi-cators (241). Mass screening has limitations, as it elucidatesonly transcribed genes, and may not be as sensitive as isnecessary for accurate identification of therapeutic targets.Assessment of mRNA expression and protein levels fre-quently generates different results, and therefore, proteinanalysis should parallel gene expression studies when seekingtherapeutic targets. Targeted inhibition of CAF-derivedECM proteins has proven difficult. For example, FAP wasinitially a very exciting target for reactive tumor stroma, butlater studies with inhibitory antibodies reported no signif-icant results (242). However, other transmembrane proteinsmay prove to be useful biomarkers. Podoplanin, in partic-ular, shows promise as a marker of lymph node metastasis.This has obvious implications in cancer treatment andmanagement, as there is an overwhelming need to distin-guish metastatic cancers from organ-confined or locallyinvasive tumors. PAR-1, as well as a11b1 and endosialinhave shown potential as therapeutic targets for prostate andlung cancer, respectively. Importantly, CAF-downregulatedproteins, such as WFDC1 may be equally significant in thedevelopment of therapeutic modalities, and their utilityshould not be overlooked (Table 1). Inhibition of proteaseshas been challenging most likely because of their uniqueexpression at different stages of progression, and varyingregulatory functions in tumor and stromal cells. This elu-cidates the necessity of highly specific and strategic targeting

of MMPs. Nonetheless, PAI-1 and -2 show encouragingprognostic utility particularly for breast cancer, which issupported by many studies.Matrix proteins expressed at the target site may have

prognostic significance when detected in the primarytumor tissue, as in the case of osteopontin and osteonec-tin. Because of their metastasis-promoting and chemoat-tractant properties, these proteins could be targeted spe-cifically using competitive peptides to prevent coloniza-tion of tumor cells in bone. Bone stromal cells also displayincreased expression of the ECM proteins, versican andtenascin, which promote tumor growth in 3D coculturewith advanced prostate cancer cells (243). This providesfurther rationale for targeting tumor-promoting ECMproteins at the metastatic or secondary site, as it ispresumable that cancer cells induce expression of ECMproteins in target tissue that contribute to their growth.Perlecan, with its diverse biologic activities and contribu-tions to tumor growth, is abundant in bone marrow, andmay be another desired target for bone-metastasizingcancers (150, 244, 245). Finally, given the role of pro-teases in tumor metastasis, fragments of many of theseECM molecules will likely prove to have significantbiologic effects quite varied from the native molecule. Assuch, these fragments may be either therapeutic targets oreasily detectable prognostic markers.

SummaryThe reactive stroma is characterized by a diverse milieu

of soluble and membrane-bound proteins mediating mul-tiple deregulated biologic processes. Reciprocal commu-nication between tumor and stromal cells relies on sig-naling platforms regulated by the diverse components ofthe ECM, ultimately promoting tumor growth, survival,and eventual colonization of the metastatic site. Thus, theECM deserves special attention in the study of tumordevelopment and progression and quest for therapeutictargets.Expression of ECM components and associated signal-

ing events dictating cancer cell responses are determinedby tumor stage and grade, as well as cancer type (47).Therefore, these factors must be considered whenattempting to understand the tumor stromal microenvi-ronment. In the context of the continuous and intricatestring of reciprocal signals in the tumor microenviron-ment, elucidation of relevant stromal components shouldparallel the discovery of tumor-secreted molecules inducedby paracrine signaling.Our knowledge of the functional significance of the ECM

is based predominantly on standard 2D bioassays andmurine models, but it will take more innovative modelscarefully designed to recapitulate tumor–stromal interac-tions in 3D to ascertain the clinicopathologic relevance ofspecific matrix molecules. Hence, prospective studies ofsufficient sample size, necessary first steps to uncover thetrue prognostic value of ECM proteins, must be proceededby comprehensive studies in appropriate model systems to

Miles and Sikes

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research304

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 9: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

analyze and confirm the roles of these prospective targets. Asthe dynamics of tumor–stromal interactions at differenttumor stages become clearer, targeted approaches to preventand predict cancer progression can be introduced.

ImplicationsTumor expansion and metastasis depend not only on the

biology and signaling from or within the epithelial com-partment of the cancer, but also to a large degree on theparacrine factors and matrix composition of the cancer-associated stroma. Future cancer therapy should targethighly relevant stromal cell-derived matrix factors, conse-

quently attenuating stromal-mediated paracrine signals asso-ciated with cancer progression.

Disclosure of Potential Conflicts of InterestR.A. Sikes is a consultant/advisory board member of Delaware Prostate Cancer

Coalition, 1st State Prostate Cancer Survivors, and Sisters on a Mission. No potentialconflicts of interest were disclosed by the other authors.

AcknowledgmentsThis work was supported by the Center for Translational Cancer Research, the

Delaware INBRE program, NIH NIGMS (8 P20 GM103446-13), and NIH P01-CA98912 on Prostate Cancer Bone Metastasis: Biology and Targeting.

Received October 7, 2013; revised December 30, 2013; accepted December 30,2013; published OnlineFirst January 22, 2014.

References1. Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T,

Naeem R, et al. Stromal fibroblasts present in invasive human breastcarcinomas promote tumor growth and angiogenesis through ele-vated SDF-1/CXCL12 secretion. Cell 2005;121:335–48.

Table 1. Stromal ECM factors and their clinical relevance/prognostic value in cancer

Factor Expression Clinical significance Cancer

Oncofetal fibronectin Stroma Malignancy, poor prognosis Oral (53), ovarian (54), colorectal (55),breast (56)

TGF-b stroma Mortality, tumor growth Prostate (11), breast (12)SDF-1 CAFs Tumor progression, angiogenesis Breast (1, 2)HB-EGF Stroma, CAFs Tumor growth Cervical (31), prostate (32)Tenascin-C, -W Stroma Malignancy, metastasis Breast (60, 64, 71)MMP-2 Stroma, sera Poor prognosis, metastasis,

invasionNSCLC (86), prostate (88), skin SCC (93)

MMP-13 Myofibroblasts, stroma Poor prognosis, invasion Skin (93), HNSCC (99), colorectal (100),breast (102)

MMP-11 Fibroblasts, stroma Local invasiveness Breast (47), HNSCC (101)Adam-9 Myofibroblasts Metastasis Colorectal (104)Adam-12 Stroma Tumor progression Prostate (105)PAI-1, uPA Tumor and stroma Poor prognosis Breast (117, 118)PAI-2 Tumor and stroma Favorable prognosis Breast (117, 119)WFDC-1 CAFs (decreased) Metastasis Prostate (121)Syndecan-1 Stroma Progression, poor survival Ovarian (128), endometrial (130), breast (134)Versican Stroma Malignancy, progression Ovarian (141), prostate (142)Decorin protein core Myofibroblasts Tumor suppression Triple-negative breast (147)Hyaluronan Stroma Poor survival, metastasis Ovarian (159), breast (160), prostate (161),

NSCLC (162)ICAM CAFs Progression NSCLC (51)VCAM CAFs Metastasis Colorectal (52)a11b1 Fibroblasts Tumorigenicity NSCLC (26)CD10 (MME) CAFs, stroma Progression, poor prognosis,

recurrenceNSCLC (51), melanoma (108), breast (109)

Podoplanin CAFs 1. Poor prognosis Breast (187, 188), lung (189), esoph. (190)Colorectal (191)2. Favorable prognosis

PAR-1 Stroma Progression Prostate (192)Osteonectin (SPARC) Stroma 1. Recurrence, poor prognosis Breast (199), pancreatic (194)

2. Reduced metastasis Bladder (200)Osteopontin Stroma Tumor progression Breast (204)Periostin Stroma 1. Favorable prognosis Breast (47)

2. Poor prognosis, progression Prostate (47, 214)

Stromal Paracrine Factors Promote Cancer Progression

www.aacrjournals.org Mol Cancer Res; 12(3) March 2014 305

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 10: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

2. Tait LR, Pauley RJ, Santner SJ, Heppner GH, HengHH, Rak JW, et al.Dynamic stromal-epithelial interactions during progression ofMCF10DCIS.com xenografts. Int J Cancer 2007;120:2127–34.

3. Quante M, Tu SP, Tomita H, Gonda T, Wang SS, Takashi S, et al.Bone marrow-derived myofibroblasts contribute to the mesenchy-mal stem cell niche and promote tumor growth. Cancer Cell2011;19:257–72.

4. AoM, FrancoOE, Park D, RamanD,Williams K, Hayward SW. Cross-talk between paracrine-acting cytokine and chemokine pathwayspromotes malignancy in benign human prostatic epithelium. CancerRes 2007;67:4244–53.

5. BurgerM,GlodekA,HartmannT,Schmitt-Gr€aff A, Silberstein LE, FujiiN, et al. Functional expression of CXCR4 (CD184) on small-cell lungcancer cells mediates migration, integrin activation, and adhesion tostromal cells. Oncogene 2003;22:8093–101.

6. Hartmann TN, Burger JA, Glodek A, Fujii N, Burger M. CXCR4chemokine receptor and integrin signaling co-operate in mediatingadhesion and chemoresistance in small cell lung cancer (SCLC) cells.Oncogene 2005;24:4462–71.

7. Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S,et al. TGF-beta signaling in fibroblasts modulates the oncogenicpotential of adjacent epithelia. Science 2004;303:848–51.

8. Cheng N, Bhowmick NA, Chytil A, Gorksa AE, Brown KA,Muraoka R,et al. Loss of TGF-beta type II receptor in fibroblasts promotesmammary carcinoma growth and invasion through upregulation ofTGF-alpha-, MSP- and HGF-mediated signaling networks. Onco-gene 2005;24:5053–68.

9. Achyut BR, Bader DA, Robles AI, Wangsa D, Harris CC, Ried T, et al.Inflammation-mediated genetic and epigenetic alterations drive can-cer development in the neighboring epithelium upon stromal abro-gation of TGF-beta signaling. PLoS Genet 2013;9:e1003251.

10. Miles FL, Tung NS, Aguiar AA, Kurtoglu S, Sikes RA. Increased TGF-beta1-mediated suppression of growth andmotility in castrate-resis-tant prostate cancer cells is consistent with Smad2/3 signaling.Prostate 2012;72:1339–50.

11. Franco OE, Jiang M, Strand DW, Peacock J, Fernandez S, JacksonRS II, et al. Altered TGF-beta signaling in a subpopulation of humanstromal cells promotes prostatic carcinogenesis. Cancer Res2011;71:1272–81.

12. Richardsen E, Uglehus RD, Johnsen SH, Busund LT. Immunohisto-chemical expression of epithelial and stromal immunomodulatorysignalling molecules is a prognostic indicator in breast cancer. BMCRes Notes 2012;5:110.

13. Liu J, Liao S, Diop-Frimpong B, Chen W, Goel S, Naxerova K, et al.TGF-beta blockade improves the distribution and efficacy of thera-peutics in breast carcinoma by normalizing the tumor stroma. ProcNatl Acad Sci U S A 2012;109:16618–23.

14. Kawada M, Inoue H, Arakawa M, Ikeda D. Transforming growthfactor-beta1 modulates tumor-stromal cell interactions of prostatecancer through insulin-like growth factor-I. Anticancer Res 2008;28:721–30.

15. Massoner P, Haag P, Seifarth C, Jurgeit A, Rogatsch H, Doppler W,et al. Insulin-like growth factor binding protein-3 (IGFBP-3) in theprostate and in prostate cancer: local production, distribution andsecretion pattern indicate a role in stromal-epithelial interaction.Prostate 2008;68:1165–78.

16. Chan JM, Stampfer MJ, Giovannucci E, Gann PH, Ma J, Wilkinson P,et al. Plasma insulin-like growth factor-I and prostate cancer risk: aprospective study. Science 1998;279:563–6.

17. Wolk A, Mantzoros CS, Andersson SO, Bergstr€om R, Signorello LB,Lagiou P, et al. Insulin-like growth factor 1 and prostate cancer risk: apopulation-based, case-control study. J Natl Cancer Inst 1998;90:911–5.

18. Giovannucci E, Pollak M, Platz EA, Willett WC, Stampfer MJ, MajeedN, et al. Insulin-like growth factor I (IGF-I), IGF-binding protein-3 andthe risk of colorectal adenoma and cancer in the Nurses' HealthStudy. Growth Horm IGF Res 2000;10 Suppl A:S30–1.

19. McCarty MF. Androgenic progestins amplify the breast cancer riskassociated with hormone replacement therapy by boosting IGF-Iactivity. Med Hypotheses 2001;56:213–6.

20. Rinaldi S, Peeters PH, Berrino F, Dossus L, Biessy C, Olsen A, et al.IGF-I, IGFBP-3 and breast cancer risk in women: the EuropeanProspective Investigation into Cancer and Nutrition (EPIC). EndocrRelat Cancer 2006;13:593–605.

21. Kaaks R, Toniolo P, Akhmedkhanov A, Lukanova A, Biessy C,Dechaud H, et al. Serum C-peptide, insulin-like growth factor(IGF)-I, IGF-binding proteins, and colorectal cancer risk in women.J Natl Cancer Inst 2000;92:1592–600.

22. GianiC, PincheraA,RasmussenA, FierabracciP,BonacciR,CampiniD, et al. Stromal IGF-II messenger RNA in breast cancer: relationshipwith progesterone receptor expressed bymalignant epithelial cells. JEndocrinol Invest 1998;21:160–5.

23. Rasmussen AA, Cullen KJ. Paracrine/autocrine regulation of breastcancer by the insulin-like growth factors. Breast Cancer Res Treat1998;47:219–33.

24. Cohen P, Peehl DM, Baker B, Liu F, Hintz RL, Rosenfeld RG. Insulin-like growth factor axis abnormalities in prostatic stromal cells frompatients with benign prostatic hyperplasia. J Clin Endocrinol Metab1994;79:1410–5.

25. Singer CF,MoggM, KoestlerW, PacherM,Marton E, Kubista E, et al.Insulin-like growth factor (IGF)-I and IGF-II serum concentrations inpatients with benign and malignant breast lesions: free IGF-II iscorrelated with breast cancer size. Clin Cancer Res 2004;10(12 Pt1):4003–9.

26. ZhuCQ,PopovaSN,BrownER,Barsyte-LovejoyD,NavabR,ShihW,et al. Integrin alpha 11 regulates IGF2 expression in fibroblasts toenhance tumorigenicity of human non-small-cell lung cancer cells.Proc Natl Acad Sci U S A 2007;104:11754–9.

27. Cao Y. Direct role of PDGF-BB in lymphangiogenesis and lymphaticmetastasis. Cell Cycle 2005;4:228–30.

28. Cao R, Bj€orndahl MA, Religa P, Clasper S, Garvin S, Galter D, et al.PDGF-BB induces intratumoral lymphangiogenesis and promoteslymphatic metastasis. Cancer Cell 2004;6:333–45.

29. Jain RK, Lahdenranta J, Fukumura D. Targeting PDGF signaling incarcinoma-associated fibroblasts controls cervical cancer in mousemodel. PLoS Med 2008;5:e24.

30. Pietras K, Pahler J, Bergers G, Hanahan D. Functions of paracrinePDGF signaling in the proangiogenic tumor stroma revealed bypharmacological targeting. PLoS Med 2008;5:e19.

31. Murata T, Mizushima H, Chinen I, Moribe H, Yagi S, Hoffman RM,et al. HB-EGF and PDGF mediate reciprocal interactions of carcino-macellswith cancer-associated fibroblasts to support progressionofuterine cervical cancers. Cancer Res 2011;71:6633–42.

32. Adam RM, Borer JG, Williams J, Eastham JA, Loughlin KR, FreemanMR. Amphiregulin is coordinately expressed with heparin-bindingepidermal growth factor-like growth factor in the interstitial smoothmuscle of the human prostate. Endocrinology 1999;140:5866–75.

33. FreemanMR, Paul S, Kaefer M, IshikawaM, AdamRM, RenshawAA,et al. Heparin-binding EGF-like growth factor in the human prostate:synthesis predominantly by interstitial and vascular smooth musclecells and action as a carcinoma cell mitogen. J Cell Biochem1998;68:328–38.

34. Adam RM, Kim J, Lin J, Orsola A, Zhuang L, Rice DC, et al. Heparin-binding epidermal growth factor-like growth factor stimulates andro-gen-independent prostate tumor growth and antagonizes androgenreceptor function. Endocrinology 2002;143:4599–608.

35. Comoglio PM, Giordano S, Trusolino L. Drug development of METinhibitors: targeting oncogene addiction and expedience. Nat RevDrug Discov 2008;7:504–16.

36. Daly AJ, McIlreavey L, Irwin CR. Regulation of HGF and SDF-1expression by oral fibroblasts–implications for invasion of oral can-cer. Oral Oncol 2008;44:646–51.

37. De Wever O, Nguyen QD, Van Hoorde L, Bracke M, Bruyneel E,Gespach C, et al. Tenascin-C and SF/HGF produced by myofibro-blasts in vitro provide convergent pro-invasive signals to humancolon cancer cells through RhoA and Rac. FASEB J 2004;18:1016–8.

38. Jedeszko C, Victor BC, Podgorski I, Sloane BF. Fibroblast hepato-cyte growth factor promotes invasion of human mammary ductalcarcinoma in situ. Cancer Res 2009;69:9148–55.

Miles and Sikes

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research306

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 11: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

39. Tate A, Isotani S, Bradley MJ, Sikes RA, Davis R, Chung LW, et al.Met-Independent Hepatocyte Growth Factor-mediated regulation ofcell adhesion in human prostate cancer cells. BMC Cancer2006;6:197.

40. Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, et al.Matrix crosslinking forces tumor progression by enhancing integrinsignaling. Cell 2009;139:891–906.

41. Crispino P, De Toma G, Ciardi A, Bella A, Rivera M, Cavallaro G, et al.Role of desmoplasia in recurrence of stage II colorectal cancer withinfive years after surgery and therapeutic implication. Cancer Invest2008;26:419–25.

42. Faouzi S, Le Bail B, Neaud V, Boussarie L, Saric J, Bioulac-Sage P,et al. Myofibroblasts are responsible for collagen synthesis in thestroma of human hepatocellular carcinoma: an in vivo and in vitrostudy. J Hepatol 1999;30:275–84.

43. Huijbers IJ, Iravani M, Popov S, Robertson D, Al-Sarraj S, Jones C,et al. A role for fibrillar collagen deposition and the collagen internal-ization receptor endo180 in glioma invasion. PLoS ONE 2010;5:e9808.

44. Kauppila S, Stenb€ack F, Risteli J, Jukkola A, Risteli L. Aberrant type Iand type III collagen gene expression in human breast cancer in vivo.J Pathol 1998;186:262–8.

45. Tuxhorn JA, Ayala GE, Smith MJ, Smith VC, Dang TD, Rowley DR,et al. Reactive stroma in human prostate cancer: induction of myofi-broblast phenotype and extracellular matrix remodeling. Clin CancerRes 2002;8:2912–23.

46. Bauer M, Su G, Casper C, He R, Rehrauer W, Friedl A, et al. Hetero-geneity of gene expression in stromal fibroblasts of human breastcarcinomas and normal breast. Oncogene 2010;29:1732–40.

47. Planche A, Bacac M, Provero P, Fusco C, Delorenzi M, Stehle JC,et al. Identification of prognostic molecular features in the reactivestroma of human breast and prostate cancer. PLoS ONE 2011;6:e18640.

48. Halsted KC, Bowen KB, Bond L, Luman SE, Jorcyk CL, Fyffe WE,et al. Collagen alpha1(XI) in normal and malignant breast tissue. ModPathol 2008;21:1246–54.

49. Allinen M, Beroukhim R, Cai L, Brennan C, Lahti-Domenici J, HuangH, et al. Molecular characterization of the tumor microenvironment inbreast cancer. Cancer Cell 2004;6:17–32.

50. Fukushima N, Sato N, Prasad N, Leach SD, Hruban RH, Goggins M.Characterization of gene expression inmucinouscystic neoplasmsofthe pancreas using oligonucleotide microarrays. Oncogene2004;23:9042–51.

51. Navab R, Strumpf D, Bandarchi B, Zhu CQ, Pintilie M, Ramnarine VR,et al. Prognostic gene-expression signature of carcinoma-associat-ed fibroblasts in non-small cell lung cancer. Proc Natl Acad Sci U S A2011;108:7160–5.

52. Nakagawa H, Liyanarachchi S, Davuluri RV, Auer H, Martin EW Jr, dela Chapelle A, et al. Role of cancer-associated stromal fibroblasts inmetastatic colon cancer to the liver and their expression profiles.Oncogene 2004;23:7366–77.

53. Mandel U, Gaggero B, Reibel J, Therkildsen MH, Dabelsteen E,Clausen H. Oncofetal fibronectins in oral carcinomas: correlation oftwo different types. APMIS 1994;102:695–702.

54. Menzin AW, Loret de Mola JR, Bilker WB, Wheeler JE, Rubin SC,Feinberg RF. Identification of oncofetal fibronectin in patients withadvanced epithelial ovarian cancer: detection in ascitic fluid andlocalization to primary sites and metastatic implants. Cancer1998;82:152–8.

55. InufusaH,NakamuraM,Adachi T,Nakatani Y, ShindoK,YasutomiM,et al. Localization of oncofetal and normal fibronectin in colorectalcancer. Correlation with histologic grade, liver metastasis, and prog-nosis. Cancer 1995;75:2802–8.

56. Schor AM, Schor SL. Multiple Fibroblast Phenotypes in CancerPatients: Heterogeneity in Expression of Migration Stimulating Fac-tor, in Tumor-Associated Fibroblasts and Their Matrix: Tumor Stro-ma,Mueller M.M., Fusenig F., editors. Springer-Verlag: New York;2011.

57. SantimariaM,Moscatelli G, VialeGL,Giovannoni L,Neri G,Viti F, et al.Immunoscintigraphic detection of the ED-B domain of fibronectin, a

marker of angiogenesis, in patients with cancer. Clin Cancer Res2003;9:571–9.

58. Schor SL, Ellis IR, Jones SJ, Baillie R, Seneviratne K, Clausen J, et al.Migration-stimulating factor: a genetically truncated onco-fetal fibro-nectin isoform expressed by carcinoma and tumor-associated stro-mal cells. Cancer Res 2003;63:8827–36.

59. Lightner VA, SlempCA, Erickson HP. Localization and quantitation ofhexabrachion (tenascin) in skin, embryonic brain, tumors, and plas-ma. Ann N Y Acad Sci 1990;580:260–75.

60. Mackie EJ, Chiquet-Ehrismann R, Pearson CA, Inaguma Y, Taya K,Kawarada Y, et al. Tenascin is a stromal marker for epithelial malig-nancy in the mammary gland. Proc Natl Acad Sci U S A 1987;84:4621–5.

61. Brellier F, Chiquet-EhrismannR. Howdo tenascins influence the birthand life of a malignant cell? J Cell Mol Med 2012;16:32–40.

62. Schenk S, Muser J, Vollmer G, Chiquet-Ehrismann R. Tenascin-C inserum: a questionable tumor marker. Int J Cancer 1995;61:443–9.

63. Amonkar SD, Bertenshaw GP, Chen TH, Bergstrom KJ, Zhao J,Seshaiah P, et al. Development and preliminary evaluation of amultivariate indexassay for ovariancancer. PLoSONE2009;4:e4599.

64. O'Connell JT, Sugimoto H, Cooke VG, MacDonald BA, Mehta AI,LeBleu VS, et al. VEGF-A and Tenascin-C produced by S100A4þstromal cells are important for metastatic colonization. Proc NatlAcad Sci U S A 2011;108:16002–7.

65. Yokosaki Y, Monis H, Chen J, Sheppard D. Differential effects of theintegrins alpha9beta1, alphavbeta3, and alphavbeta6 on cell prolif-erative responses to tenascin. Roles of the beta subunit extracellularand cytoplasmic domains. J Biol Chem 1996;271:24144–50.

66. Yokosaki Y, Palmer EL, Prieto AL, Crossin KL, BourdonMA, Pytela R,et al. The integrin alpha 9 beta 1 mediates cell attachment to a non-RGDsite in the third fibronectin type III repeat of tenascin. JBiol Chem1994;269:26691–6.

67. HuangW, Chiquet-Ehrismann R, Moyano JV, Garcia-Pardo A, OrendG. Interference of tenascin-C with syndecan-4 binding to fibronectinblocks cell adhesion and stimulates tumor cell proliferation. CancerRes 2001;61:8586–94.

68. Orend G, Huang W, Olayioye MA, Hynes NE, Chiquet-Ehrismann R.Tenascin-C blocks cell-cycle progression of anchorage-dependentfibroblasts on fibronectin through inhibition of syndecan-4. Onco-gene 2003;22:3917–26.

69. Wenk MB, Midwood KS, Schwarzbauer JE. Tenascin-C suppressesRho activation. J Cell Biol 2000;150:913–20.

70. Hancox RA, Allen MD, Holliday DL, Edwards DR, Pennington CJ,Guttery DS, et al. Tumour-associated tenascin-C isoforms promotebreast cancer cell invasion and growth by matrix metalloproteinase-dependent and independent mechanisms. Breast Cancer Res2009;11:R24.

71. Scherberich A, Tucker RP, Degen M, Brown-Luedi M, Andres AC,Chiquet-Ehrismann R. Tenascin-W is found in malignant mammarytumors, promotes alpha8 integrin-dependent motility and requiresp38MAPK activity for BMP-2 and TNF-alpha induced expression invitro. Oncogene 2005;24:1525–32.

72. Martina E, DegenM, R€ueggC,Merlo A, LinoMM,Chiquet-EhrismannR, et al. Tenascin-W is a specific marker of glioma-associated bloodvessels and stimulates angiogenesis in vitro. FASEB J 2010;24:778–87.

73. LederleW, Hartenstein B,Meides A, KunzelmannH,Werb Z, Angel P,et al. MMP13 as a stromal mediator in controlling persistent angio-genesis in skin carcinoma. Carcinogenesis 2010;31:1175–84.

74. Whitelock JM, Murdoch AD, Iozzo RV, Underwood PA. The degra-dation of human endothelial cell-derived perlecan and release ofbound basic fibroblast growth factor by stromelysin, collagenase,plasmin, and heparanases. J Biol Chem 1996;271:10079–86.

75. Suzuki M, Raab G, Moses MA, Fernandez CA, Klagsbrun M. Matrixmetalloproteinase-3 releases active heparin-bindingEGF-like growthfactor by cleavage at a specific juxtamembrane site. J Biol Chem1997;272:31730–7.

76. Fowlkes JL, Enghild JJ, Suzuki K, Nagase H. Matrix metalloprotei-nases degrade insulin-like growth factor-binding protein-3 in dermalfibroblast cultures. J Biol Chem 1994;269:25742–6.

Stromal Paracrine Factors Promote Cancer Progression

www.aacrjournals.org Mol Cancer Res; 12(3) March 2014 307

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 12: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

77. Thrailkill KM, Quarles LD, NagaseH, Suzuki K, Serra DM, Fowlkes JL.Characterization of insulin-like growth factor-binding protein 5-degrading proteases produced throughout murine osteoblast differ-entiation. Endocrinology 1995;136:3527–33.

78. Degraff DJ, Aguiar AA, Sikes RA. Disease evidence for IGFBP-2 as akey player in prostate cancer progression and development of osteo-sclerotic lesions. Am J Transl Res 2009;1:115–30.

79. Gregory CW, Degeorges A, Sikes RA. The role of the IGF axis in thedevelopment and progression of prostate cancer., in RecentResearch Developments in Cancer,Pandalai S.G., Mukhtar H., LabrieF., editors. Transworld Research Network: Kerala, India; 2011. p.437–62.

80. Nelson AR, Fingleton B, Rothenberg ML, Matrisian LM. Matrixmetalloproteinases: biologic activity and clinical implications. J ClinOncol 2000;18:1135–49.

81. Boyd RS, Balkwill FR. MMP-2 release and activation in ovariancarcinoma: the role of fibroblasts. Br J Cancer 1999;80:315–21.

82. Ko K, Yazumi S, Yoshikawa K, Konda Y, Nakajima M, Chiba T, et al.Activation of fibroblast-derived matrix metalloproteinase-2 by colon-cancer cells in non-contact Co-cultures. Int J Cancer 2000;87:165–71.

83. Tokumaru Y, Fujii M, Otani Y, Kameyama K, Imanishi Y, Igarashi N,et al. Activation of matrix metalloproteinase-2 in head and necksquamous cell carcinoma: studies of clinical samples and in vitrocell lines co-cultured with fibroblasts. Cancer Lett 2000;150:15–21.

84. Taniwaki K, Fukamachi H, Komori K, Ohtake Y, Nonaka T, SakamotoT, et al. Stroma-derived matrix metalloproteinase (MMP)-2 promotesmembrane type 1-MMP-dependent tumor growth in mice. CancerRes 2007;67:4311–9.

85. Zhang W, Matrisian LM, Holmbeck K, Vick CC, Rosenthal EL. Fibro-blast-derived MT1-MMP promotes tumor progression in vitro and invivo. BMC Cancer 2006;6:52.

86. IshikawaS, TakenakaK, Yanagihara K,Miyahara R, KawanoY,OtakeY, et al. Matrix metalloproteinase-2 status in stromal fibroblasts, notin tumor cells, is a significant prognostic factor in non-small-cell lungcancer. Clin Cancer Res 2004;10:6579–85.

87. Giannoni E, Bianchini F, Masieri L, Serni S, Torre E, Calorini L, et al.Reciprocal activation of prostate cancer cells and cancer-associatedfibroblasts stimulates epithelial-mesenchymal transition and cancerstemness. Cancer Res 2010;70:6945–56.

88. Gohji K, Fujimoto N, Hara I, Fujii A, Gotoh A, Okada H, et al. Serummatrix metalloproteinase-2 and its density in men with prostatecancer as a new predictor of disease extension. Int J Cancer1998;79:96–101.

89. YuL,WangCY,Shi J,Miao L,DuX,MayerD, et al. Estrogens promoteinvasion of prostate cancer cells in a paracrine manner through up-regulation of matrix metalloproteinase 2 in prostatic stromal cells.Endocrinology 2011;152:773–81.

90. Shariat SF, ShalevM,Menesses-Diaz A, Kim IY, KattanMW,WheelerTM, et al. Preoperative plasma levels of transforming growth factorbeta(1) (TGF-beta(1)) strongly predict progression in patients under-going radical prostatectomy. J Clin Oncol 2001;19:2856–64.

91. Wikstrom P, Stattin P, Franck-Lissbrant I, Damber JE, Bergh A.Transforming growth factor beta1 is associated with angiogenesis,metastasis, and poor clinical outcome in prostate cancer. Prostate1998;37:19–29.

92. Baciu PC, Suleiman EA, Deryugina EI, Strongin AY. Membranetype-1 matrix metalloproteinase (MT1-MMP) processing of pro-alphav integrin regulates cross-talk between alphavbeta3 andalpha2beta1 integrins in breast carcinoma cells. Exp Cell Res2003;291:167–75.

93. Vosseler S, Lederle W, Airola K, Obermueller E, Fusenig NE, MuellerMM. Distinct progression-associated expression of tumor and stro-malMMPs inHaCaT skin SCCscorrelateswith onset of invasion. Int JCancer 2009;125:2296–306.

94. Nielsen BS, Egeblad M, Rank F, Askautrud HA, Pennington CJ,Pedersen TX, et al. Matrix metalloproteinase 13 is induced in fibro-blasts in polyomavirus middle T antigen-driven mammary carcinomawithout influencing tumor progression. PLoS ONE 2008;3:e2959.

95. Wang TN, Albo D, Tuszynski GP. Fibroblasts promote breast cancercell invasion by upregulating tumor matrix metalloproteinase-9 pro-duction. Surgery 2002;132:220–5.

96. MassonR, LefebvreO,No€el A, FahimeME,ChenardMP,WendlingC,et al. In vivo evidence that the stromelysin-3 metalloproteinasecontributes in a paracrine manner to epithelial cell malignancy. J CellBiol 1998;140:1535–41.

97. Andarawewa KL, Boulay A, Masson R, Mathelin C, Stoll I, TomasettoC, et al. Dual stromelysin-3 function during natural mouse mammarytumor virus-ras tumor progression. Cancer Res 2003;63:5844–9.

98. Boulay A, Masson R, Chenard MP, El Fahime M, Cassard L, BellocqJP, et al. High cancer cell death in syngeneic tumors developed inhost mice deficient for the stromelysin-3 matrix metalloproteinase.Cancer Res 2001;61:2189–93.

99. Culhaci N, Metin K, Copcu E, Dikicioglu E. Elevated expression ofMMP-13 and TIMP-1 in head and neck squamous cell carcinomasmay reflect increased tumor invasiveness. BMC Cancer 2004;4:42.

100. Leeman MF, McKay JA, Murray GI. Matrix metalloproteinase 13activity is associated with poor prognosis in colorectal cancer. J ClinPathol 2002;55:758–62.

101. Muller D, Wolf C, Abecassis J, Millon R, Engelmann A, Bronner G,et al. Increased stromelysin 3 gene expression is associated withincreased local invasiveness in head and neck squamous cell car-cinomas. Cancer Res 1993;53:165–9.

102. Nielsen BS, Rank F, L�opez JM, Balbin M, Vizoso F, Lund LR, et al.Collagenase-3 expression in breast myofibroblasts as a molecularmarker of transition of ductal carcinoma in situ lesions to invasiveductal carcinomas. Cancer Res 2001;61:7091–100.

103. Blanchot-Jossic F, Jarry A, Masson D, Bach-Ngohou K, Paineau J,DenisMG, et al. Up-regulated expression of ADAM17 in human coloncarcinoma: co-expression with EGFR in neoplastic and endothelialcells. J Pathol 2005;207:156–63.

104. MazzoccaA, Coppari R, De Franco R, Cho JY, Libermann TA, PinzaniM, et al. A secreted form of ADAM9 promotes carcinoma invasionthrough tumor-stromal interactions. Cancer Res 2005;65:4728–38.

105. Peduto L, Reuter VE, Sehara-FujisawaA, Shaffer DR,ScherHI, BlobelCP. ADAM12 is highly expressed in carcinoma-associated stromaand is required for mouse prostate tumor progression. Oncogene2006;25:5462–6.

106. Rocks N, Paulissen G, El Hour M, Quesada F, Crahay C, Gueders M,et al. Emerging roles of ADAM and ADAMTS metalloproteinases incancer. Biochimie 2008;90:369–79.

107. Tyan SW, Hsu CH, Peng KL, Chen CC, KuoWH, Lee EY, et al. Breastcancer cells induce stromal fibroblasts to secrete ADAMTS1 forcancer invasion through an epigenetic change. PLoS ONE 2012;7:e35128.

108. Bilalovic N, Sandstad B, Golouh R, Nesland JM, Selak I, TorlakovicEE. CD10 protein expression in tumor and stromal cells of malignantmelanoma is associated with tumor progression. Mod Pathol2004;17:1251–8.

109. Makretsov NA, Hayes M, Carter BA, Dabiri S, Gilks CB, HuntsmanDG. Stromal CD10 expression in invasive breast carcinoma corre-lates with poor prognosis, estrogen receptor negativity, and highgrade. Mod Pathol 2007;20:84–9.

110. Dall'Era MA, True LD, Siegel AF, Porter MP, Sherertz TM, Liu AY.Differential expression of CD10 in prostate cancer and its clinicalimplication. BMC Urol 2007;7:3.

111. Cui L, Ohuchida K, Mizumoto K, Moriyama T, Onimaru M, Nakata K,et al. Prospectively isolated cancer-associated CD10(þ) fibroblastshave stronger interactionswithCD133(þ) colon cancer cells thanwithCD133(�) cancer cells. PLoS ONE 2010;5:e12121.

112. Noel A, Hajitou A, L'Hoir C, Maquoi E, Baramova E, Lewalle JM, et al.Inhibitionof stromalmatrixmetalloproteases: effects onbreast-tumorpromotion by fibroblasts. Int J Cancer 1998;76:267–73.

113. KitamuraH,OosawaY,KawanoN,KamedaY,HayashiH,Nakatani Y,et al. Basement membrane patterns, gelatinase A and tissue inhibitorof metalloproteinase-2 expressions, and stromal fibrosis during thedevelopment of peripheral lung adenocarcinoma. Hum Pathol1999;30:331–8.

Miles and Sikes

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research308

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 13: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

114. Ra HJ, Parks WC. Control of matrix metalloproteinase catalyticactivity. Matrix Biol 2007;26:587–96.

115. GrafM, Baici A, Strauli P. Histochemical localization of cathepsin B atthe invasion front of the rabbit V2 carcinoma. Lab Invest 1981;45:587–96.

116. Campo E, Mu~noz J, Miquel R, Palacín A, Cardesa A, Sloane BF, et al.Cathepsin B expression in colorectal carcinomas correlates withtumor progression and shortened patient survival. Am J Pathol1994;145:301–9.

117. Foekens JA, Look MP, Peters HA, van Putten WL, Portengen H, KlijnJG. Urokinase-type plasminogen activator and its inhibitor PAI-1:predictors of poor response to tamoxifen therapy in recurrent breastcancer. J Natl Cancer Inst 1995;87:751–6.

118. Look MP, van Putten WL, Duffy MJ, Harbeck N, Christensen IJ,Thomssen C, et al. Pooled analysis of prognostic impact of uroki-nase-type plasminogen activator and its inhibitor PAI-1 in 8377breast cancer patients. J Natl Cancer Inst 2002;94:116–28.

119. Duggan C, Kennedy S, Kramer MD, Barnes C, Elvin P, McDermott E,et al. Plasminogen activator inhibitor type 2 in breast cancer. Br JCancer 1997;76:622–7.

120. Croucher DR, Saunders DN, Lobov S, Ranson M. Revisiting thebiological roles of PAI2 (SERPINB2) in cancer. Nat Rev Cancer2008;8:535–45.

121. Madar S, Brosh R, Buganim Y, Ezra O, Goldstein I, Solomon H, et al.Modulated expression of WFDC1 during carcinogenesis and cellularsenescence. Carcinogenesis 2009;30:20–7.

122. Sanchez-Carbayo M, Socci ND, Lozano J, Saint F, Cordon-Cardo C.Defining molecular profiles of poor outcome in patients with invasivebladder cancer using oligonucleotide microarrays. J Clin Oncol2006;24:778–89.

123. Wachi S, Yoneda K, Wu R. Interactome-transcriptome analysisreveals the high centrality of genes differentially expressed in lungcancer tissues. Bioinformatics 2005;21:4205–8.

124. Endo K, Takino T, Miyamori H, Kinsen H, Yoshizaki T, Furukawa M,et al. Cleavage of syndecan-1 by membrane type matrix metallopro-teinase-1 stimulates cell migration. J Biol Chem 2003;278:40764–70.

125. Rodriguez-Manzaneque JC, Carpizo D, Plaza-Calonge Mdel C,Torres-Collado AX, Thai SN, Simons M, et al. Cleavage of synde-can-4 by ADAMTS1 provokes defects in adhesion. Int J BiochemCellBiol 2009;41:800–10.

126. Iozzo RV, Zoeller JJ, Nystrom A. Basement membrane proteogly-cans:modulators Par Excellence of cancer growth and angiogenesis.Mol Cells 2009;27:503–13.

127. Maeda T, Alexander CM, Friedl A. Induction of syndecan-1 expres-sion in stromal fibroblasts promotes proliferation of human breastcancer cells. Cancer Res 2004;64:612–21.

128. Davies EJ, Blackhall FH, Shanks JH, David G, McGown AT,Swindell R, et al. Distribution and clinical significance of heparansulfate proteoglycans in ovarian cancer. Clin Cancer Res 2004;10:5178–86.

129. Maeda T, Desouky J, Friedl A. Syndecan-1 expression by stromalfibroblasts promotes breast carcinoma growth in vivo and stimulatestumor angiogenesis. Oncogene 2006;25:1408–12.

130. Hasengaowa, Kodama J, Kusumoto T, Shinyo Y, Seki N, HiramatsuY. Prognostic significance of syndecan-1 expression in human endo-metrial cancer. Ann Oncol 2005;16:1109–15.

131. AnttonenA, KajantiM,Heikkil€aP, JalkanenM, JoensuuH. Syndecan-1 expression has prognostic significance in head and neck carcino-ma. Br J Cancer 1999;79:558–64.

132. YangY, YaccobyS, LiuW, Langford JK, PumphreyCY, TheusA, et al.Soluble syndecan-1 promotes growth of myeloma tumors in vivo.Blood 2002;100:610–7.

133. Nikolova V, Koo CY, Ibrahim SA,Wang Z, Spillmann D, Dreier R, et al.Differential roles for membrane-bound and soluble syndecan-1(CD138) in breast cancer progression. Carcinogenesis 2009;30:397–407.

134. Lofgren L, Sahlin L, Jiang S, Von Schoultz B, Fernstad R, Skoog L,et al. Expression of syndecan-1 in paired samples of normal andmalignant breast tissue from postmenopausal women. AnticancerRes 2007;27:3045–50.

135. Du WW, Yang BB, Shatseva TA, Yang BL, Deng Z, Shan SW, et al.Versican G3 promotes mouse mammary tumor cell growth, migra-tion, and metastasis by influencing EGF receptor signaling. PLoSONE 2011;5:e13828.

136. Kischel P, Waltregny D, Dumont B, Turtoi A, Greffe Y, Kirsch S, et al.Versican overexpression in human breast cancer lesions: known andnew isoforms for stromal tumor targeting. Int J Cancer 2010;126:640–50.

137. Ricciardelli C, Russell DL, Ween MP, Mayne K, Suwiwat S, Byers S,et al. Formation of hyaluronan- and versican-rich pericellular matrixby prostate cancer cells promotes cell motility. J Biol Chem2007;282:10814–25.

138. Sakko AJ, Ricciardelli C, Mayne K, Tilley WD, Lebaron RG, HorsfallDJ. Versican accumulation in human prostatic fibroblast cultures isenhanced by prostate cancer cell-derived transforming growth factorbeta1. Cancer Res 2001;61:926–30.

139. Skandalis SS, Labropoulou VT, Ravazoula P, Likaki-Karatza E, DobraK, Kalofonos HP, et al. Versican but not decorin accumulation isrelated to malignancy in mammographically detected high densityand malignant-appearing microcalcifications in non-palpable breastcarcinomas. BMC Cancer 2011;11:314.

140. Kusumoto T, Kodama J, Seki N, Nakamura K, Hongo A, Hiramatsu Y.Clinical significanceof syndecan-1and versican expression in humanepithelial ovarian cancer. Oncol Rep 2010;23:917–25.

141. Voutilainen K, Anttila M, Sillanp€a€a S, Tammi R, Tammi M, SaarikoskiS, et al. Versican in epithelial ovarian cancer: relation to hyaluronan,clinicopathologic factors and prognosis. Int J Cancer 2003;107:359–64.

142. Ricciardelli C,Mayne K, Sykes PJ, RaymondWA,McCaul K,MarshallVR, et al. Elevated levels of versican but not decorin predict diseaseprogression in early-stage prostate cancer. Clin Cancer Res1998;4:963–71.

143. Baumhoer D, Tornillo L, Stadlmann S, Roncalli M, Diamantis EK,Terracciano LM. Glypican 3 expression in human nonneoplastic,preneoplastic, and neoplastic tissues: a tissue microarray analysisof 4,387 tissue samples. Am J Clin Pathol 2008;129:899–906.

144. Capurro MI, Xiang YY, Lobe C, Filmus J. Glypican-3 promotes thegrowth of hepatocellular carcinoma by stimulating canonical Wntsignaling. Cancer Res 2005;65:6245–54.

145. Kleeff J, Ishiwata T, Kumbasar A, Friess H, B€uchler MW, Lander AD,et al. The cell-surface heparan sulfate proteoglycan glypican-1 reg-ulates growth factor action in pancreatic carcinoma cells and isoverexpressed in human pancreatic cancer. J Clin Invest 1998;102:1662–73.

146. Csordas G, Santra M, Reed CC, Eichstetter I, McQuillan DJ, Gross D,et al. Sustained down-regulation of the epidermal growth factorreceptor by decorin. A mechanism for controlling tumor growth invivo. J Biol Chem 2000;275:32879–87.

147. Buraschi S, Neill T, Owens RT, Iniguez LA, Purkins G, Vadigepalli R,et al. Decorin protein core affects theglobal geneexpressionprofile ofthe tumor microenvironment in a triple-negative orthotopic breastcarcinoma xenograft model. PLoS ONE 2012;7:e45559.

148. Matsumine A, Shintani K, Kusuzaki K, Matsubara T, Satonaka H,Wakabayashi T, et al. Expression of decorin, a small leucine-richproteoglycan, as a prognostic factor in soft tissue tumors. J SurgOncol 2007;96:411–8.

149. Coulson-Thomas VJ, Coulson-Thomas YM, Gesteira TF, Andrade dePaulaCA,CarneiroCR,Ortiz V, et al. Lumicanexpression, localizationand antitumor activity in prostate cancer. Exp Cell Res 2013;319:967–81.

150. Chung LW. Prostate Cancer Bone Colonization: Osteomimicry in theBone Niche. In: Bone and Cancer,Bronner F., Farach-Carson C.M.,editors. Springer; 2010.

151. Dunlevy JR, Hassell JR. Heparan Sulfate Proteoglycans in BasementMembranes: Perlecan, Agrin, and Collagen XVIII. In: Proteoglycans:Structure, Biology, and Molecular Interactions,Iozzo R.V., editors.CRC Press: New York; 2000.

152. Cohen IR, Murdoch AD, Naso MF, Marchetti D, Berd D, Iozzo RV.Abnormal expression of perlecan proteoglycan in metastatic mela-nomas. Cancer Res 1994;54:5771–4.

Stromal Paracrine Factors Promote Cancer Progression

www.aacrjournals.org Mol Cancer Res; 12(3) March 2014 309

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 14: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

153. AviezerD,HechtD, SafranM,EisingerM,DavidG,YayonA.Perlecan,basal lamina proteoglycan, promotes basic fibroblast growth factor-receptor binding, mitogenesis, and angiogenesis. Cell 1994;79:1005–13.

154. Mongiat M, Otto J, Oldershaw R, Ferrer F, Sato JD, Iozzo RV.Fibroblast growth factor-binding protein is a novel partner for perle-can protein core. J Biol Chem 2001;276:10263–71.

155. Mongiat M, Taylor K, Otto J, Aho S, Uitto J, Whitelock JM, et al. Theprotein core of the proteoglycan perlecan binds specifically to fibro-blast growth factor-7. J Biol Chem 2000;275:7095–100.

156. Lee B, Clarke D, Al Ahmad A, Kahle M, Parham C, Auckland L, et al.Perlecan domain V is neuroprotective and proangiogenic followingischemic stroke in rodents. J Clin Invest 2011;121:3005–23.

157. Farach-Carson MC, Brown AJ, Lynam M, Safran JB, Carson DD. Anovel peptide sequence in perlecan domain IV supports cell adhe-sion, spreading and FAK activation. Matrix Biol 2008;27:150–60.

158. Kobayashi N, Miyoshi S, Mikami T, Koyama H, Kitazawa M, TakeokaM, et al. Hyaluronan deficiency in tumor stroma impairs macrophagetrafficking and tumor neovascularization. Cancer Res 2010;70:7073–83.

159. AnttilaMA, TammiRH, TammiMI, Syrj€anenKJ, Saarikoski SV, KosmaVM. High levels of stromal hyaluronan predict poor disease outcomein epithelial ovarian cancer. Cancer Res 2000;60:150–5.

160. Auvinen P, Tammi R, Parkkinen J, Tammi M, Agren U, Johansson R,et al. Hyaluronan in peritumoral stroma and malignant cells associ-ates with breast cancer spreading and predicts survival. Am J Pathol2000;156:529–36.

161. Lipponen P, Aaltomaa S, Tammi R, Tammi M, Agren U, Kosma VM.High stromal hyaluronan level is associated with poor differenti-ation and metastasis in prostate cancer. Eur J Cancer 2001;37:849–56.

162. Pirinen R, Tammi R, Tammi M, Hirvikoski P, Parkkinen JJ, JohanssonR, et al. Prognostic value of hyaluronan expression in non-small-celllung cancer: increased stromal expression indicates unfavorableoutcome in patients with adenocarcinoma. Int J Cancer 2001;95:12–7.

163. Miles FL, Pruitt FL, van Golen KL, Cooper CR. Stepping out of theflow: capillary extravasation in cancer metastasis. Clin Exp Metas-tasis 2008;25:305–24.

164. Wang KK, Liu N, Radulovich N, Wigle DA, Johnston MR, ShepherdFA, et al. Novel candidate tumor marker genes for lung adenocarci-noma. Oncogene 2002;21:7598–604.

165. Cesselli D, Beltrami AP, PozA,Marzinotto S,Comisso E, BergaminN,et al. Role of tumor associatedfibroblasts in human liver regeneration,cirrhosis, and cancer. Int J Hepatol 2011;2011:120925.

166. Plow EF, Haas TA, Zhang L, Loftus J, Smith JW. Ligand binding tointegrins. J Biol Chem 2000;275:21785–8.

167. Velling T, Risteli J, Wennerberg K, Mosher DF, Johansson S. Poly-merization of type I and III collagens is dependent on fibronectin andenhanced by integrins alpha 11beta 1 and alpha 2beta 1. J Biol Chem2002;277:37377–81.

168. Wipff PJ, Hinz B. Integrins and the activation of latent transforminggrowth factor beta1 - an intimate relationship. Eur J Cell Biol2008;87:601–15.

169. Orimo A, Weinberg RA. Stromal fibroblasts in cancer: a novel tumor-promoting cell type. Cell Cycle 2006;5:1597–601.

170. Chen H, Yang WW, Wen QT, Xu L, Chen M. TGF-beta inducesfibroblast activation protein expression; fibroblast activation proteinexpression increases the proliferation, adhesion, and migration ofHO-8910PM [corrected]. Exp Mol Pathol 2009;87:189–94.

171. Goodman JD, Rozypal TL, Kelly T. Seprase, a membrane-boundprotease, alleviates the serum growth requirement of human breastcancer cells. Clin Exp Metastasis 2003;20:459–70.

172. Huang Y,Wang S, Kelly T. Seprase promotes rapid tumor growth andincreased microvessel density in a mouse model of human breastcancer. Cancer Res 2004;64:2712–6.

173. Huber MA, Kraut N, Park JE, Schubert RD, Rettig WJ, Peter RU, et al.Fibroblast activation protein: differential expression and serine pro-tease activity in reactive stromal fibroblasts of melanocytic skintumors. J Invest Dermatol 2003;120:182–8.

174. Ramirez-Montagut T, Blachere NE, Sviderskaya EV, Bennett DC,Rettig WJ, Garin-Chesa P, et al. FAPalpha, a surface peptidaseexpressed during wound healing, is a tumor suppressor. Oncogene2004;23:5435–46.

175. Cheng JD, Valianou M, Canutescu AA, Jaffe EK, Lee HO, Wang H,et al. Abrogation of fibroblast activation protein enzymatic activityattenuates tumor growth. Mol Cancer Ther 2005;4:351–60.

176. Abbas O, Richards JE, Mahalingam M. Fibroblast-activation protein:a single marker that confidently differentiates morpheaform/infiltra-tive basal cell carcinoma from desmoplastic trichoepithelioma. ModPathol 2010;23:1535–43.

177. WenY,WangCT, Ma TT, Li ZY, Zhou LN,Mu B, et al. Immunotherapytargeting fibroblast activation protein inhibits tumor growth andincreases survival in a murine colon cancer model. Cancer Sci2010;101:2325–32.

178. Kraman M, Bambrough PJ, Arnold JN, Roberts EW, Magiera L,Jones JO, et al. Suppression of antitumor immunity by stromal cellsexpressing fibroblast activation protein-alpha. Science 2010;330:827–30.

179. Zhi K, Shen X, Zhang H, Bi J. Cancer-associated fibroblasts arepositively correlated with metastatic potential of human gastric can-cers. J Exp Clin Cancer Res 2010;29:66.

180. Mueller SC, Ghersi G, Akiyama SK, Sang QX, Howard L, Pineiro-Sanchez M, et al. A novel protease-docking function of integrin atinvadopodia. J Biol Chem 1999;274:24947–52.

181. Christian S, Ahorn H, Koehler A, Eisenhaber F, Rodi HP, Garin-ChesaP, et al.Molecular cloning andcharacterization of endosialin, aC-typelectin-like cell surface receptor of tumor endothelium. J Biol Chem2001;276:7408–14.

182. MacFadyen JR, Haworth O, Roberston D, Hardie D, Webster MT,Morris HR, et al. Endosialin (TEM1, CD248) is a marker of stromalfibroblasts and is not selectively expressed on tumour endothelium.FEBS Lett 2005;579:2569–75.

183. Tomkowicz B, Rybinski K, Foley B, Ebel W, Kline B, Routhier E, et al.Interaction of endosialin/TEM1 with extracellular matrix proteinsmediates cell adhesion and migration. Proc Natl Acad Sci U S A2007;104:17965–70.

184. Maia M, DeVriese A, Janssens T, Moons M, Lories RJ, Tavernier J,et al. CD248 facilitates tumor growth via its cytoplasmic domain.BMC Cancer 2011;11:162.

185. Kahn HJ, Marks A. A new monoclonal antibody, D2–40, for detec-tion of lymphatic invasion in primary tumors. Lab Invest 2002;82:1255–7.

186. Hoshino A, Ishii G, Ito T, Aoyagi K, Ohtaki Y, Nagai K, et al. Podo-planin-positive fibroblasts enhance lung adenocarcinoma tumor for-mation: podoplanin in fibroblast functions for tumor progression.Cancer Res 2011;71:4769–79.

187. PulaB, JethonA,PiotrowskaA,GomulkiewiczA,Owczarek T,Calik J,et al. Podoplanin expression by cancer-associated fibroblasts pre-dicts poor outcome in invasive ductal breast carcinoma. Histopa-thology 2011;59:1249–60.

188. Schoppmann SF, Berghoff A, Dinhof C, Jakesz R, Gnant M, DubskyP, et al. Podoplanin-expressing cancer-associated fibroblasts areassociated with poor prognosis in invasive breast cancer. BreastCancer Res Treat 2012;134:237–44.

189. Kawase A, Ishii G, Nagai K, Ito T, Nagano T, Murata Y, et al.Podoplanin expression by cancer associated fibroblasts predictspoor prognosis of lung adenocarcinoma. Int J Cancer 2008;123:1053–9.

190. Schoppmann SF, Jesch B, Riegler MF, Maroske F, Schwameis K,Jomrich G, et al. Podoplanin expressing cancer associated fibro-blasts are associated with unfavourable prognosis in adenocarcino-ma of the esophagus. Clin Exp Metastasis 2013;30:441–6.

191. Yamanashi T, Nakanishi Y, Fujii G, Akishima-Fukasawa Y, Moriya Y,Kanai Y, et al. Podoplanin expression identified in stromal fibroblastsas a favorable prognostic marker in patients with colorectal carcino-ma. Oncology 2009;77:53–62.

192. Zhang X, Wang W, True LD, Vessella RL, Takayama TK. Protease-activated receptor-1 is upregulated in reactive stroma of primaryprostate cancer and bone metastasis. Prostate 2009;69:727–36.

Miles and Sikes

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research310

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 15: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

193. Wang W, Mize GJ, Zhang X, Takayama TK. Kallikrein-related pepti-dase-4 initiates tumor-stroma interactions in prostate cancer throughprotease-activated receptor-1. Int J Cancer 2010;126:599–610.

194. Infante JR, Matsubayashi H, Sato N, Tonascia J, Klein AP, Riall TA,et al. Peritumoral fibroblast SPARC expression and patient outcomewith resectable pancreatic adenocarcinoma. J Clin Oncol 2007;25:319–25.

195. Jacob K, Webber M, Benayahu D, Kleinman HK. Osteonectin pro-motes prostate cancer cell migration and invasion: a possible mech-anism for metastasis to bone. Cancer Res 1999;59:4453–7.

196. Lau CP, Poon RT, Cheung ST, Yu WC, Fan ST. SPARC and Hevinexpression correlate with tumour angiogenesis in hepatocellularcarcinoma. J Pathol 2006;210:459–68.

197. Le Bail B, Faouzi S, Boussarie L, Guirouilh J, Blanc JF, Carles J, et al.Osteonectin/SPARC is overexpressed in human hepatocellular car-cinoma. J Pathol 1999;189:46–52.

198. Koblinski JE, Kaplan-Singer BR, VanOsdol SJ, Wu M, Engbring JA,Wang S, et al. Endogenous osteonectin/SPARC/BM-40 expressioninhibits MDA-MB-231 breast cancer cell metastasis. Cancer Res2005;65:7370–7.

199. Witkiewicz AK, Freydin B, Chervoneva I, PotoczekM, RizzoW, Rui H,et al. Stromal CD10 and SPARC expression in ductal carcinoma insitu (DCIS) patients predicts disease recurrence. Cancer Biol Ther2010;10:391–6.

200. Said N, Frierson HF, Sanchez-CarbayoM, Brekken RA, TheodorescuD. Loss of SPARC in bladder cancer enhances carcinogenesis andprogression. J Clin Invest 2013;123:751–66.

201. Bull PhelpsSL,Carbon J,Miller A, Castro-Rivera E, Arnold S, BrekkenRA, et al. Secreted protein acidic and rich in cysteine as a regulator ofmurine ovarian cancer growth and chemosensitivity. Am J ObstetGynecol 2009;200:180 e1–7.

202. CheethamS, TangMJ,MesakF, KenneckeH,OwenD, Tai IT. SPARCpromoter hypermethylation in colorectal cancers can be reversed by5-Aza-20deoxycytidine to increase SPARC expression and improvetherapy response. Br J Cancer 2008;98:1810–9.

203. Coppola D, Szabo M, Boulware D, Muraca P, Alsarraj M, ChambersAF, et al. Correlation of osteopontin protein expression and patho-logical stage across a wide variety of tumor histologies. Clin CancerRes 2004;10(1 Pt 1):184–90.

204. Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Zhao H,et al. Stromal gene expression predicts clinical outcome in breastcancer. Nat Med 2008;14:518–27.

205. Luo X, Ruhland MK, Pazolli E, Lind AC, Stewart SA. Osteopontinstimulates preneoplastic cellular proliferation through activation ofthe MAPK pathway. Mol Cancer Res 2011;9:1018–29.

206. Pazolli E, Luo X, Brehm S, Carbery K, Chung JJ, Prior JL, et al.Senescent stromal-derived osteopontin promotes preneoplastic cellgrowth. Cancer Res 2009;69:1230–9.

207. Derosa CA, Furusato B, Shaheduzzaman S, Srikantan V, Wang Z,Chen Y, et al. Elevated osteonectin/SPARC expression in primaryprostate cancer predicts metastatic progression. Prostate CancerProstatic Dis 2012;15:150–6.

208. Thalmann GN, Sikes RA, Devoll RE, Kiefer JA, Markwalder R, Klima I,et al. Osteopontin: possible role in prostate cancer progression. ClinCancer Res 1999;5:2271–7.

209. Wang X, Chao L, Ma G, Chen L, Tian B, Zang Y, et al. Increasedexpression of osteopontin in patients with triple-negative breastcancer. Eur J Clin Invest 2008;38:438–46.

210. Bao S, Ouyang G, Bai X, Huang Z, Ma C, Liu M, et al. Periostinpotently promotes metastatic growth of colon cancer by augment-ing cell survival via the Akt/PKB pathway. Cancer Cell 2004;5:329–39.

211. ErkanM,Kleeff J,GorbachevskiA,ReiserC,MitkusT, Esposito I, et al.Periostin creates a tumor-supportive microenvironment in the pan-creas by sustaining fibrogenic stellate cell activity. Gastroenterology2007;132:1447–64.

212. Tilman G, Mattiussi M, Brasseur F, van Baren N, Decottignies A.Human periostin gene expression in normal tissues, tumors andmelanoma: evidences for periostin production by both stromal andmelanoma cells. Mol Cancer 2007;6:80.

213. Fukushima N, Kikuchi Y, Nishiyama T, Kudo A, Fukayama M. Peri-ostin deposition in the stroma of invasive and intraductal neoplasmsof the pancreas. Mod Pathol 2008;21:1044–53.

214. Sun C, Zhao X, Xu K, Gong J, Liu W, Ding W, et al. Periostin: apromising target of therapeutical intervention for prostate cancer. JTransl Med 2011;9:99.

215. Dolznig H, Walzl A, Kramer N, Rosner M, Garin-Chesa P,Hengstschl€ager M. Organotypic spheroid cultures to study tumor-stroma interaction during cancer development. Drug DiscoveryToday: Disease Models 2011;8:113–9.

216. Cukierman E. Cell migration analyses within fibroblast-derived 3-Dmatrices. Methods Mol Biol 2005;294:79–93.

217. Campbell JJ, Davidenko N, Caffarel MM, Cameron RE, Watson CJ.A multifunctional 3D co-culture system for studies of mammarytissue morphogenesis and stem cell biology. PLoS ONE 2011;6:e25661.

218. Liang Y, Jeong J, DeVolder RJ, ChaC,Wang F, Tong YW, et al. A cell-instructive hydrogel to regulate malignancy of 3D tumor spheroidswith matrix rigidity. Biomaterials 2011;32:9308–15.

219. Beacham DA, Cukierman E. Stromagenesis: the changing face offibroblastic microenvironments during tumor progression. SeminCancer Biol 2005;15:329–41.

220. KosticA, LynchCD,SheetzMP.Differentialmatrix rigidity response inbreast cancer cell lines correlates with the tissue tropism. PLoS ONE2009;4:e6361.

221. Barcus CE, Keely PJ, Eliceiri KW, Schuler LA. Stiff collagen matricesincrease tumorigenic prolactin signaling in breast cancer cells. J BiolChem 2013;288:12722–32.

222. Wozniak MA, Modzelewska K, Kwong L, Keely PJ. Focal adhesionregulation of cell behavior. BiochimBiophys Acta 2004;1692:103–19.

223. Cukierman E, Pankov R, Yamada KM. Cell interactions with three-dimensional matrices. Curr Opin Cell Biol 2002;14:633–9.

224. Grinnell F. Fibroblast-collagen-matrix contraction: growth-factor sig-nalling and mechanical loading. Trends Cell Biol 2000;10:362–5.

225. Hinz B, Mastrangelo D, Iselin CE, Chaponnier C, Gabbiani G.Mechanical tension controls granulation tissue contractile activityand myofibroblast differentiation. Am J Pathol 2001;159:1009–20.

226. Lindahl GE, Chambers RC, Papakrivopoulou J, Dawson SJ, Jacob-senMC,BishopJE, et al. Activationoffibroblast procollagenalpha1(I)transcription by mechanical strain is transforming growth factor-beta-dependent and involves increased binding of CCAAT-bindingfactor (CBF/NF-Y) at the proximal promoter. J Biol Chem 2002;277:6153–61.

227. Klein EA,Yin L, Kothapalli D,CastagninoP,ByfieldFJ, XuT, et al. Cell-cycle control by physiological matrix elasticity and in vivo tissuestiffening. Curr Biol 2009;19:1511–8.

228. Winer JP, Janmey PA, McCormick ME, Funaki M. Bone marrow-derived human mesenchymal stem cells become quiescent on softsubstrates but remain responsive to chemical or mechanical stimuli.Tissue Eng Part A 2009;15:147–54.

229. Menon S, Beningo KA. Cancer cell invasion is enhanced by appliedmechanical stimulation. PLoS ONE 2011;6:e17277.

230. Friedland JC, Lee MH, Boettiger D. Mechanically activated integrinswitch controls alpha5beta1 function. Science 2009;323:642–4.

231. Garcia AJ, Schwarzbauer JE, Boettiger D. Distinct activation states ofalpha5beta1 integrin show differential binding to RGD and synergydomains of fibronectin. Biochemistry 2002;41:9063–9.

232. Garcia AJ, Takagi J, Boettiger D. Two-stage activation for alpha5-beta1 integrin binding to surface-adsorbed fibronectin. J Biol Chem1998;273:34710–5.

233. Paszek MJ, Zahir N, Johnson KR, Lakins JN, Rozenberg GI, Gefen A,et al. Tensional homeostasis and the malignant phenotype. CancerCell 2005;8:241–54.

234. Provenzano PP, Inman DR, Eliceiri KW, Keely PJ. Matrix density-induced mechanoregulation of breast cell phenotype, signaling andgene expression through a FAK-ERK linkage. Oncogene 2009;28:4326–43.

235. Croft DR, Sahai E, Mavria G, Li S, Tsai J, Lee WM, et al. ConditionalROCK activation in vivo induces tumor cell dissemination and angio-genesis. Cancer Res 2004;64:8994–9001.

Stromal Paracrine Factors Promote Cancer Progression

www.aacrjournals.org Mol Cancer Res; 12(3) March 2014 311

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 16: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

236. Provenzano PP, Eliceiri KW, Campbell JM, Inman DR, White JG,Keely PJ. Collagen reorganization at the tumor-stromal interfacefacilitates local invasion. BMC Med 2006;4:38.

237. Chiquet-Ehrismann R, Chiquet M. Tenascins: regulation and puta-tive functions during pathological stress. J Pathol 2003;200:488–99.

238. Brown RA, Sethi KK, Gwanmesia I, Raemdonck D, Eastwood M,Mudera V. Enhanced fibroblast contraction of 3D collagenlattices and integrin expression by TGF-beta1 and -beta3:mechanoregulatory growth factors? Exp Cell Res 2002;274:310–22.

239. Grinnell F, Ho CH. Transforming growth factor beta stimulates fibro-blast-collagen matrix contraction by different mechanisms inmechanically loaded and unloaded matrices. Exp Cell Res2002;273:248–55.

240. Hinz B. The myofibroblast: paradigm for a mechanically active cell.J Biomech 2010;43:146–55.

241. Micke P, Ostman A. Exploring the tumour environment: cancer-associated fibroblasts as targets in cancer therapy. Expert Opin TherTargets 2005;9:1217–33.

242. Hofheinz RD, al-Batran SE, Hartmann F, Hartung G, J€ager D, RennerC, et al. Stromal antigen targeting by a humanised monoclonalantibody: an early phase II trial of sibrotuzumab in patients withmetastatic colorectal cancer. Onkologie 2003;26:44–8.

243. Sung SY, Hsieh CL, Law A, Zhau HE, Pathak S, Multani AS, et al.Coevolution of prostate cancer and bone stroma in three-dimension-al coculture: implications for cancer growth and metastasis. CancerRes 2008;68:9996–10003.

244. Farach-CarsonMC, Carson DD. Perlecan–amultifunctional extracel-lular proteoglycan scaffold. Glycobiology 2007;17:897–905.

245. Savore C, Zhang C, Muir C, Liu R, Wyrwa J, Shu J, et al. Perlecanknockdown in metastatic prostate cancer cells reduces heparin-binding growth factor responses in vitro and tumor growth in vivo.Clin Exp Metastasis 2005;22:377–90.

Miles and Sikes

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research312

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535

Page 17: InsidiousChangesinStromalMatrixFuelCancerProgression...Review InsidiousChangesinStromalMatrixFuelCancerProgression Fayth L. Miles1,2 and Robert A. Sikes2,3 Abstract Reciprocal interactions

2014;12:297-312. Published OnlineFirst January 22, 2014.Mol Cancer Res   Fayth L. Miles and Robert A. Sikes  Insidious Changes in Stromal Matrix Fuel Cancer Progression

  Updated version

  10.1158/1541-7786.MCR-13-0535doi:

Access the most recent version of this article at:

   

  Overview

Visual 

http://mcr.aacrjournals.org/content/12/3/297/F1.large.jpgA diagrammatic summary of the major findings and biological implications:

   

  Cited articles

  http://mcr.aacrjournals.org/content/12/3/297.full#ref-list-1

This article cites 241 articles, 73 of which you can access for free at:

  Citing articles

  http://mcr.aacrjournals.org/content/12/3/297.full#related-urls

This article has been cited by 11 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://mcr.aacrjournals.org/content/12/3/297To request permission to re-use all or part of this article, use this link

on July 24, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 22, 2014; DOI: 10.1158/1541-7786.MCR-13-0535