Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1 The Matrix Revolution: Matricellular and Restructuring of the

2 Cancer Microenvironment

3

4 Casimiro Gerarduzzi1,2,*,#, Ursula Hartmann3, Andrew Leask4 and Elliot Drobetsky1,2

5

6

7 1Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec H1T 2M4, Canada

8 2Département de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada

9 3Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany

10 4College of Dentistry, University of Saskatchewan, 105 Wiggins Rd, Saskatoon SK S7N 5E4, Canada

11

12

13 *To whom correspondence should be addressed: Casimiro Gerarduzzi, PhD, Maisonneuve-Rosemont

14 Hospital Research Center, Division of Nephrology, 5415, boul. de l'Assomption, Montreal, Quebec,

15 Canada, H1T 2M4. Phone: (514) 252-3400 ext.2813, email: [email protected]

16 #Senior Author

17

18

19 Running title: The Role of Matricellular Proteins in Cancer

20 Keywords: Matricellular Proteins, , Microenvironment, Cancer Progression,

21 Biomarkers/Therapeutics

22 Number of Figures: 1

23 Conflict of interest: The authors declare no potential conflicts of interest.

1

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

24

25 Abstract

26 The extracellular matrix (ECM) surrounding cells is indispensable for regulating their

27 behavior. The dynamics of ECM signaling are tightly controlled throughout growth and

28 development. During tissue remodeling, matricellular proteins (MCPs) are secreted into the

29 ECM. These factors do not serve classical structural roles, but rather regulate matrix proteins

30 and cell-matrix interactions to influence normal cellular functions. In the tumor

31 microenvironment, it is becoming increasingly clear that aberrantly expressed MCPs can

32 support multiple hallmarks of carcinogenesis by interacting with various cellular components

33 that are coupled to an array of downstream signals. Moreover, MCPs also reorganize the

34 biomechanical properties of the ECM to accommodate metastasis and tumor colonization. This

35 realization is stimulating new research on MCPs as reliable and accessible biomarkers in cancer,

36 as well as effective and selective therapeutic targets.

37

2

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

38 Introduction

39 The behavior of individual cells is influenced by a plethora of signals originating from the

40 surrounding microenvironment, which includes the extracellular matrix (ECM). Previously

41 regarded as merely a static scaffold for cell/tissue organization, the ECM is now viewed as a

42 critical niche contributing to the regulation of cellular survival, proliferation and migration. This

43 realization has positioned the ECM at the center stage of normal physiological processes such

44 as development, tissue homeostasis and tissue remodeling.

45 The dynamic nature of ECM signaling is determined by a secreted subset of non-

46 structural matricellular proteins (MCPs) (1), in contrast to the structural roles of “classical” ECM

47 proteins such as and (2). MCP functional versatility is achieved by its

48 multiple domains that either (i) bind ECM proteins and/or cell surface receptors, (ii) bind and

49 regulate the activity or accessibility of extracellular signaling molecules such as growth factors,

50 proteases, chemokines, and cytokines, or (iii) mediate intrinsic enzymatic activities to precisely

51 orchestrate the assembly, degradation, and organization of the ECM. MCPs are tightly

52 controlled, with expression promptly occurring in context-specific scenarios. Typically, they are

53 highly expressed during early development, ultimately subsiding in adult tissues under

54 physiological conditions. However, transient re-expression is observed during injury repair, and

55 can also be sustained in chronic pathologies such as cancer (2-7). Indeed, chronic unscheduled

56 expression of various MCPs, either by tumor cells or the surrounding stroma (8), leads to

57 abnormal ECM remodeling and stimulation of mitogenic pathways essential for cancer

58 progression. This may underlie the correlation between the upregulation of many MCPs and

3

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

59 poor prognosis in cancer patients (9) and, moreover, provide rationale for exploring the utility

60 of MCPs as cancer biomarkers and therapeutic targets.

61 This review will focus on the burgeoning roles of the MCP families SPARC, CCN, SIBLING,

62 and Gla-containing proteins in both cancer development, detection and treatment.

63 Certainly, members of these particular families are aberrantly expressed in various tumor types,

64 and moreover exhibit biochemical, biomechanical and metastatic properties influencing cancer

65 progression.

66

67 Normal physiological roles of MCPs

68 The ever-growing number of newly-discovered MCPs has necessitated their

69 classification into families. Members are grouped based on shared domains, which in turn

70 reflect the functional diversity between families.

71 The SPARC (Secreted Protein Acidic and Rich in Cysteine; hereafter alternative

72 protein names are included in parentheses; BM40, ), one of the original MCPs to be

73 characterized, is considered prototypical due to its simple structure and rich functionality. The

74 subsequent discovery of other MCPs with structural similarity revealed a broader family of

75 SPARC-related proteins (10). Such SPARC family members share follistatin-like and extracellular

76 -binding (EC) domains, and are classified into five distinct groups based on sequence

77 homology of their EC domains (10): SPARCs, SPARCL1, SMOCs, SPOCKs and Follistatin-like

78 protein-1 (FSTL1). SPARC family members were shown to regulate ECM assembly and

79 deposition, influence cytokine activity, inhibit cell adhesion and cell cycle progression, regulate

80 cell differentiation and activate matrix metalloproteinases (10). While most SPARC members

4

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

81 exhibit ubiquitous expression throughout early development, in adults, expression is largely

82 limited to tissues that are diseased or undergoing wound repair/remodeling.

83 The vertebrate CCN (Centralized Coordination Network) family is composed of six

84 homologous cysteine-rich members (11): CCN1 (CYR61), CCN2 (CTGF), CCN3 (NOV), CCN4

85 (WISP-1), CCN5 (WISP-2) and CCN6 (WISP-3). Each is comprised of an N-terminal secretory

86 peptide and four functional domains: insulin-like growth factor-binding protein domain (IGFBP),

87 Von Willebrand factor domain (VWR), thrombospondin type-1 repeat module (TSR) and

88 carboxy-terminal cysteine-knot (CT) motif (11). In response to tissue remodeling, CCN proteins

89 are expressed, principally in mesenchymal cells, during development and in

90 pathologies (12). The postnatal role of CCN proteins is known for promoting collagen stability or

91 organization (13).

92 (TNs) comprise a family of four large extracellular matrix , i.e.,

93 TN-C, -R, -W and -X, which exist as either trimers or hexamers (14). TNs share a characteristic

94 modular structure composed of tandem (EGF)-like domains,

95 fibronectin-type III domains and a C-terminal fibrinogen related domain (FReD). Consequently,

96 TNs share functions in modulating cellular responses to the ECM and growth factors, specifically

97 regulating growth, differentiation, adhesion and migration during tissue remodeling events

98 (15). However, each member has distinct spatial and temporal expression. TN-C expression is

99 typically present in all organs during fetal development and mechanical stress, whereas TN-W

100 expression is restricted to developing/remodeling and certain stem cell niches (14). TN-R

101 is expressed exclusively in the developing and adult nervous system, while TN-X represents a

5

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

102 constitutive ECM component of most connective tissues, being hardly influenced by external

103 factors (14).

104 The SIBLING (Small Integrin-Binding Ligand N-Linked ) family includes Bone

105 Sialoprotein (BSP), Osteopontin (SPP1, aka OPN), Dentin Sialophosphoprotein (DSPP), Matrix

106 Extracellular Phosphoglycoprotein (MEPE) and Dentin Matrix Protein-1 (DMP1). These proteins

107 are primarily implicated in bone morphogenesis and , and were thus thought

108 to be exclusively localized to mineralized tissue such as bone and teeth (16). However, apart

109 from these traditional functions, SIBLING members were also shown to influence cellular

110 proliferation/survival pathways, collagen fibrillogenesis, MMP activities and response to injury

111 (17-20).

112 The Gla-protein family members contain -dependent γ-carboxyglutamic acid

113 residues (21), which have high affinity for calcium ions, thus conferring important roles in

114 coagulation and bone homeostasis (22). Among the 17 Gla-protein members, Periostin (POSTN)

115 and Matrix Gla-Protein (MGP) are known to affect ECM crosslinking and various cellular

116 behaviors, such as migration, adhesion and proliferation in epithelial, endothelial, ,

117 osteoblast and myocyte cells (23-27). POSTN is expressed in osteoblast, mesangial, fibroblast,

118 mesenchymal and vascular smooth muscle cells (22), while MGP is typically secreted and

119 localized in the surrounding ECM of chondrocytes or endothelial cells (28).

120 Considering that MCP expression is context dependent, MCP knockout mouse models

121 generally lack any postnatal phenotype unless challenged by injury or disease, in which case

122 they exhibit an impaired yet subtle response (see references for further details: SPARC family

123 (29-34); CCN family (11,35); TN family (36-38); SIBLING family (39-42); POSTN (43-45)).

6

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

124 However, some MCP mouse knockouts are characterized by severe complications. For example,

125 FSTL-1 and CCN2 null mice die shortly after birth, while CCN1 and CCN5 whole body knockouts

126 are embryonic lethal, showing that these proteins are essential for development (11,46,47). As

127 for MGP knockout mice, they show severe vascular calcification, arteriovenous malformation

128 and craniofacial anomalies, and die within 8 weeks after birth (48-50).

129

130 Expression of MCPs in Cancer

131 MCP overexpression is characteristic of tissue remodeling processes, including those

132 occurring during carcinogenesis, as opposed to low/undetectable levels in normal tissue. Tumor

133 cells and the surrounding activated stromal cells are the major cell types that aberrantly secrete

134 MCPs into the tumor microenvironment, in turn promoting cancer development (5,51).

135 Nonetheless, we note there are certain cases where MCP expression has been shown to oppose

136 cancer development (51,52).

137 SPARC protein is highly expressed in cancer cells and the stroma of certain cancers,

138 including glioma, breast and cervical melanoma (53-56), where it exhibits oncogenic roles in cell

139 growth, invasion and apoptosis. Interestingly, SPARC has also been associated with tumor

140 suppression by influencing these same processes (57). This discrepancy might be explained by

141 cancer type and stage, and/or the concentration of SPARC in the tumor microenvironment (57).

142 Like SPARC, the role of FSTL1 in carcinogenesis has generated significant controversy.

143 Endometrial and ovarian cancers exhibit low FSTL1 levels; moreover, ectopic FSTL1 expression

144 exerts anti-neoplastic activity by inducing apoptosis (58). Among SPOCK isoforms (SPOCK1-3),

145 SPOCK1 is upregulated in different tumor types, and its expression positively correlates with

7

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

146 invasive/metastatic potential and hence poor prognosis (59-61). However, in brain tumors,

147 expression of all SPOCK family members decreases with increasing tumor grade (62). SMOC2

148 was shown to be upregulated in hepatocellular-, endometrial- and colorectal- cancers where it

149 modulated proliferation, chemoresistance and metastasis, respectively (63-65). Very little is

150 known regarding any role for SMOC1 in carcinogenesis, although its expression is increased in

151 brain tumors, where it interacts with TN-C to counteract the chemo-attractive effect of the

152 latter on glioma cells in vitro (66).

153 Among the CCN family, CCN1 and CCN2 are the most studied in cancer (11). Specifically,

154 CCN1 expression is elevated in many tumor types including brain, breast, prostate and pancreas

155 (67-70); similarly, CCN2 upregulation is implicated in proliferation, apoptosis and migration for

156 numerous cancers (71), including gastric (72), pancreatic (73), melanoma (74,75) and breast

157 (76). In addition to cancer cells, a potential origin of these MCPs may be cancer-associated

158 (CAFs) (77), and indeed this cell type was shown to be the source of CCN1/CCN2

159 expression in murine models of skin cancer (78,79). Although unscheduled expression of CCN1

160 and CCN2 are generally associated with tumor promotion, in some cases these proteins were

161 reported to inhibit cancer development (80,81). Like CCN1/2, CCN3 and CCN4 exhibit a mixture

162 of pro- vs anti- tumorigenic effects, whereas CCN5 and CCN6 are predominantly regarded as

163 tumor suppressors (11,82).

164 Each TN family member differs substantially in spatial- (tissue specificity) and temporal-

165 expression patterns (14). In the case of TN-C and TN-W, de novo expression is prominent in

166 tumors versus healthy tissue, where they promote tumor progression on multiple levels, i.e.

167 proliferation, invasion, metastasis and angiogenesis. TN-C is recovered in the stroma of most

8

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

168 solid cancers, while TN-W is primarily restricted to brain, colon, kidney and lung cancers (14). In

169 contrast, TN-R and TN-X are constitutively expressed and largely unaffected by tumorigenic

170 signals, i.e. to date have not been reported to play a substantial role in carcinogenesis (14).

171 Among SIBLING proteins, SPP1 and BSP have been the most extensively studied in the

172 context of cancer (16). Consistent with their roles in osteogenesis, SPP1 and BSP have been

173 implicated in bone malignancy (16). However, while these proteins were initially thought to be

174 expressed only during bone morphogenesis, both were subsequently shown to be broadly

175 expressed in human epithelial carcinomas, including but not limited to breast (83,84), lung

176 (85,86), prostate (87), liver (88), pancreas (89) and colon (87,90), where their

177 pathophysiological roles have recently been thoroughly reviewed (16,91). Furthermore, CAFs

178 have been shown to produce and secrete SPP1, which contributes to melanoma tumor growth

179 (92).

180 POSTN, the most well-characterized Gla-protein family member, was shown to be a

181 major determinant in proliferation for a number of aggressive, advanced solid tumors with poor

182 prognosis (22). MGP is much less known than POSTN, but is gradually emerging as a

183 determinant in cancer progression, exhibiting increased expression in colorectal, glioblastoma,

184 breast, cervical, osteosarcoma and skin cancers with unfavorable prognosis (93-97).

185 In general, MCPs are capable of regulating a variety of mechanisms necessary for

186 tumorigenesis, such as survival, proliferation, migration, matrix stiffness and development of a

187 signal reservoir and metastatic microenvironment. These versatile functions depend on the

188 diverse biochemical, biomechanical and metastatic niche effects induced by MCPs (Fig. 1), as

189 discussed in more detail immediately below.

9

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

190

191 MCP Biochemical Effects

192 Much evidence has shown that MCPs possess biochemical properties essential for

193 regulating various cellular behaviors, including ones implicated in tumor development. These

194 properties mainly pertain to the ability of MCPs to activate a number of cell surface receptors

195 and elicit their downstream signaling (Fig. 1A). Most MCPs are well-known to directly bind

196 integrins, which are αβ heterodimers composed from 18 α subunits and 8 β subunits (98).

197 Integrins are commonly bound by members of the SPARC, CCN, SIBLING, TN and Gla-protein

198 families (99-101), each bound to varying heterodimer combinations. Other than integrins,

199 members of CCN and TN families can also bind syndecans, while SPP1 is reported to also bind

200 CD44 receptors (99,100). In addition, MCPs can act indirectly by binding a variety of ligands (i.e.

201 growth factors and cytokines), thereby limiting ligand distribution and accessibility, or either co-

202 activate or inhibit their function (101).

203 SPARC has been reported to mediate a variety of signaling pathways. For example,

204 SPARC can bind directly to integrin receptors (αvβ1, αvβ3 and αvβ5) resulting in the activation

205 of the proximal intracellular kinases Akt, focal adhesion kinase (FAK), and integrin-linked kinase

206 (ILK) (102-105). These kinases were associated with SPARC-mediated invasion and survival of

207 glioma cells (105). SPARC may also directly interact with the TGFβ1 receptor to mediate Smad

208 signaling, as shown in lung cancer cells (106). Recently, SPARC was reported to bind TGFβ1 to

209 regulate its deposition in the ECM (107). In addition, SPARC may bind other growth factors but

210 with unknown effects (108,109). Interestingly, SPOCK1 was identified as a downstream target

211 of TGFβ1 and a key player in lung cancer metastasis and proliferation (110), as well as in anti-

10

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

212 apoptosis via activation of the PI3K/Akt pathway (111,112). SMOC2 acts to maintain ILK activity

213 during G1 which in turn influences cell cycle progression by modulating cyclin D1 expression

214 and DNA synthesis (113). This possibly involves ILK interaction with integrin β1 and β3

215 cytoplasmic domains, which also leads to inhibition of anoikis and apoptosis through activation

216 of PI3K/Akt signaling (114). Studies by Maier et al suggested that SMOC2 can bind directly to

217 integrins αVβ1 and αVβ6 (115), consistent with recent data showing that SMOC2 binds integrin

218 β1 to activate FAK in kidney fibroblasts (29).

219 CCNs act through multiple mechanisms to regulate a plethora of dynamic cellular

220 processes (11,101,116). In particular, these proteins activate ILK/Akt, MAPK, and associated

221 growth promoting pathways in cancer, with each CCN member exerting distinct effects and

222 temporal expression profiles. For example, CCN1 signals through integrin αVβ3/Sonic hedgehog

223 to promote motility in vitro and tumorigenic growth in vivo (117) as well as integrin α6β1-

224 mediated invasion (118), in pancreatic cancer. In glioma, CCN1 overexpression enhances

225 tumorigenicity through integrin αVβ3- and αVβ1-linked ILK-mediated activation of Akt, β-

226 catenin-TCF/Lef and associated survival and proliferation pathways (119). In breast cancer cells,

227 CCN1 can promote (i) resistance to anoikis, partly via integrin β1 (120), as well as (ii)

228 proliferation, survival and apoptosis resistance through the αvβ3-activated ERK1/2 pathway

229 (121). Similar to CCN1, ectopic expression of CCN2 (i) promotes migration and angiogenesis

230 (122), and (ii) confers apoptosis resistance through integrin αvβ3/ERK1/2 upregulation of anti-

231 apoptotic Bcl-xL and cIAP (76) in breast cancer cells. Although most CCNs act primarily through

232 binding various integrin heterodimer combinations, they also bind several other receptors

11

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

233 (11,101,123,124), for example syndecan-4 and Notch in the case of CCN1/2 and CCN3,

234 respectively. Interestingly, CCN proteins may be activated by proteolytic cleavage (125,126).

235 The opposing effects of CCN3 and CCN4 in different cancers raise the question of which

236 biochemical pathways are responsible for their signaling diversity. In colorectal cancer cells,

237 CCN3 inhibits survival by regulating caspase-3/-8 while inhibiting JNK-mediated migration (127).

238 On the contrary, CCN3 promotes osteoclastogenesis through the FAK/Akt/p38/NF-κB pathway

239 (128). CCN4 also promotes FAK and p38 signaling through αvβ1 integrin in prostate cancer cells;

240 however, this pathway specifically induces migration and vascular cell adhesion molecule-1

241 (VCAM-1) expression by down-regulating miR-126 (129). Furthermore, osteoblast-derived CCN4

242 plays a key role in prostate cancer cell adhesion to bone through VCAM-1/integrin α4β1 (130).

243 CCN4 also promotes lymphangiogenesis in oral squamous cell carcinoma (SCC) via integrin

244 αvβ3/Akt signaling and upregulation of VEGF-C expression, as well as promotes integrin

245 αvβ3/FAK/JNK signaling to induce VEGF-A-activation of angiogenesis in osteosarcoma cells

246 (131,132). Conversely, CCN4 inhibits migration in melanoma and lung cancer cells by

247 inactivating the family of Rho-like GTPases (133,134).

248 TN-C has been shown to stimulate proliferation and survival in a variety of cancers by

249 activating several pathways downstream of integrins and syndecans (135), including integrin

250 α9β1 activation of Akt and MAPK (136) and αvβ3 activation of FAK and paxillin (137). However,

251 a recent study showed that TN-C signaling through integrin α2β1, but not α9β1 or αvβ3,

252 induced autocrine growth in brain tumor cells (138). Through an indirect mechanism of

253 tumorigenesis, TN-C is able to compete with syndecan-4 binding to fibronectin, thereby

254 interfere with fibronectin inhibition of proliferation (139). Instead, the FBG domain of TN-X was

12

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

255 reported to convert latent TGFβ1 into its biologically active form to indirectly control

256 mesenchymal differentiation (140).

257 POSTN is primarily known for binding integrins αvβ3 and αvβ5 to elicit activation of

258 FAK/JNK and PI3-K/Akt signaling pathways controlling cell proliferation, survival, or migration in

259 various cancers (141-143). POSTN may also signal through epidermal growth factor receptor

260 (EGFR) to influence migration in esopahageal SCC (144), potentially through cross-talk with

261 integrin αvβ5. Unlike POSTN, little is known regarding the mechanism of MGP in cancer

262 development, although the latter can influence the TGFβ superfamily, including activation of

263 TGFβ1 receptor and inhibition of the bone morphogenetic proteins BMP-2 and BMP-4 (27,145).

264 The SIBLING family members BSP and SPP1 exhibit similar activities in cancer

265 development. BSP supports adhesion, proliferation and migration through αvβ3 and αvβ5, and

266 the pro-metastatic activity of TGFβ1 in breast cancer cells (146,147). SPP1 can interact with

267 several integrin receptors (αvβ1,3,5, α8β1, α9β1,4 and α4β1) to regulate cell proliferation,

268 angiogenesis, adhesion and migration (116,148). SPP1 can also signal through CD44 (149) to

269 activate HIF-2α-induced stemness in hepatocellular carcinoma and glioblastoma cells (150,151),

270 and Akt-mediated cell survival in mesothelioma and colorectal cancer cells (152,153).

271

272 MCP Biomechanical Effects

273 Remodeling of the ECM is an integral process in cancer development that

274 accommodates the structural architecture of the tumor and provides necessary physical

275 changes such as increased matrix and tissue stiffness to promote and sustain neoplastic

13

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

276 transformation (154). Mechanotransduction is a process in which perturbations in ECM

277 mechanical stiffness are transduced into biochemical signals. ECM stiffness can communicate

278 with cells through mechano-responsive integrins (98). In a normal setting, the ECM forms a

279 structural microenvironment of relaxed non-oriented fibrils that exerts homeostatic stiffness on

280 embedded cells. In cancer, disruption of this local ECM structure can occur through MCP-

281 mediated remodeling (5,8,155-157), which result in structures that are often stiffer, more

282 highly linearized and have a different orientation, relative to normal stroma (7). In response to

283 this, matrix bound integrin structures convert these physical mechanical signals into

284 conventional integrin biochemical signals to influence survival, proliferation and growth (158).

285 Moreover, integrin and their associated intracellular cytoskeleton mature into reinforced focal

286 adhesions and stress fibers, respectively, to compensate for changes in ECM stiffness (Fig. 1B).

287 Stress fibers are formed from the bundling of actin, and generate a counter-force, both of

288 which are regulated by phosphoactivating myosin through the stiffness-induced integrin-RhoA-

289 ROCK axis (159). Some of the biomechanical processes regulated by MCPs that affect ECM

290 stiffness include increased matrix organization, collagen crosslinking and deregulation of

291 enzymatic activity.

292 SPARC is well-known to be implicated in rearranging the matrix through collagen

293 crosslinking. SPARC binds to several fibrillary (I, II, III and V) as well as to collagen IV, a

294 prominent constituent of basement membranes (160,161), and is critical for organization of

295 collagenous extracellular matrices. SPARC knockout mice manifest significant changes in

296 collagen fibril morphology, as well as a substantial decrease in adult tissue concentrations of

297 collagen (for review see Bradshaw and references therein). SPARC also influences the response

14

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

298 of host tissue to implanted tumor cells and a lack of endogenous SPARC engenders decreased

299 capacity to encapsulate the tumor as well as a reduction in the deposition of collagen (162).

300 SPARC exerts at least two roles in collagen fibril assembly, i.e., by modulating interactions of

301 collagen with cell surface receptors and directly regulating collagen incorporation into fibrils

302 (163). Loss of SPARC also disrupts the homeostasis of basement membranes and alters tissue

303 biomechanics and physiological function (164). Finally, SPARC can act as an extracellular

304 chaperone for collagens that enhance the tumorigenic environment (164-166).

305 In a recent study, TN-C significantly co-localized with aligned collagen fibers in breast

306 cancer patients, compared to the wavy and randomly organized layout of collagen (167)

307 typically observed in normal tissue (168). TN-C contains multiple ECM binding partners,

308 including collagen; however, its involvement in collagen alignment may be mediated through

309 binding to fibronectin, which serves to direct collagen organization (169-171). Similarly, POSTN

310 plays a mechanistic role in inter-matrix interactions through formation of a POSTN-BMP-1-LOX

311 complex, where BMP-1 promotes LOX activity for collagen crosslinking (172,173). In fact, POSTN

312 knockout animal models exhibit aberrant collagen fibrillogenesis (174). Furthermore, the

313 mechanotransduction pathways of both ROCK in SCC and the transcription factor TWIST from

314 various mechanical stress models are known to increase POSTN deposition (24,175). The POSTN

315 family member MGP was recently shown to be incorporated into crosslinked multimers of

316 fibronectin, which enhanced cancer cell attachment to fibronectin (23). As for the CCN family,

317 recent studies have shown CCN1, CCN2 and CCN4 to promote alignment and stability of

318 collagen fibers (13,157,176).

319

15

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

320 MCP Influence on the Metastatic Niche

321 The matrix environment needs to achieve a level of plasticity for cellular displacement

322 during metastasis. In order to disseminate, cancer cells require a local ECM niche to support

323 cellular differentiation and intravasation, and an ECM at the secondary metastatic site to

324 permit invasion and colonization (Fig. 1C). There are various ways in which MCPs are able to

325 establish a metastatic niche by influencing the ECM and its embedded cells. First, MCPs induce

326 cancer cells to undergo an epithelial-to-mesenchymal transition (EMT), a genetic program that

327 promotes metastatic dissemination of cancer cells from primary epithelial tumors (177).

328 Second, MCPs reorganize the ECM architecture and integrity to promote cancer cell

329 accessibility into intact structures, i.e. (178). MCPs can also affect physical

330 properties of the ECM, including spatial arrangement, orientation, rigidity, permeability and

331 solubility, in such a way as to alter anchorage sites and create motility tracks suitable for

332 metastasis (178).

333 Normally, epithelial cells maintain their polarity, intercellular tight-junctions and

334 adherence to the basement membrane necessary for a proper tissue architecture and function

335 (179). During EMT, epithelial cells undergo reorganization of adhesion and cytoskeletal

336 structures to acquire a mesenchymal morphology. This allows cells to detach which, in

337 conjunction with enhanced migratory capacity associated with the mesenchymal phenotype,

338 stimulates metastasis (179).

339 SPARC family members promote EMT in a variety of cancers (Fig. 1C). Recently, SMOC2

340 was shown to participate in a pro-metastatic secretome mediated by the ARNTL2 transcription

16

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

341 factor in lung adenocarcinoma (180), and SMOC2 induction is required for colon cancer

342 invasion by stimulating EMT (65). Several studies also show that SPOCK1 promotes EMT

343 (110,181). Among SPARC family members, SPARC is the most characterized for influencing EMT

344 (106,182,183): (i) In lung cancer cell lines, TGFβ1 activation of migration and EMT is in part

345 through SPARC (106), (ii) In head and neck cancer cells, SPARC enhances EMT signaling via

346 activation of Akt (182), and (iii) Overexpression of SPARC in melanoma cells increases

347 invasiveness mediated by phosphorylation of FAK and Snail repression of E-cadherin promoter

348 activity (184).

349 The CCN family exerts varying effects on EMT. An early study using pancreatic cancer

350 cells reported that CCN1 promotes EMT and stemness, and that silencing this MCP forestalled

351 aggressive tumor cell behavior by reversing the EMT phenotype (67). Recent studies have

352 continued to dissect CCN1 signaling leading to EMT. In osteosarcoma, pharmacological or

353 knockdown of integrin αvβ5/Raf-1/MEK/ERK signaling components inhibited CCN1-induced

354 EMT (185), as well as CCN1-mediated expression of EMT markers and cell spreading through an

355 IGF1Rβ-JNK-dependent pathway (186). In contrast, CCN5 and CCN6 exert opposing effects on

356 EMT. In triple negative breast cancer cells, CCN5 activates the Bcl-2/Bax apoptotic pathway and

357 inhibits both EMT and migration (187), while activation of the JAK/Akt/STAT pathway reverses

358 such CCN5-mediated events (188). Similarly, CCN6 reversed the EMT features and inhibited

359 metastasis of breast cancer cells in vivo, but through a Slug signaling axis that regulates Notch1

360 activation (189). Another mechanism involves CCN6-BMP-4 binding in breast cancer cells, which

361 reduces BMP-4 signaling through p38/TAK1 and subsequent downstream activation of invasion

362 and migration (190).

17

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

363 TN-C and POSTN have also been associated with metastasis (191-195). While the

364 influence of TN-C (196,197) and POSTN (198,199) can be exerted through the EMT process,

365 interestingly, these MCPs are also capable of remodeling the ECM to form migratory tracks that

366 support rapid dissemination of cancer cells (Fig. 1C). TN-C is frequently observed to be

367 expressed along the border of matrix tracks in skin (200), pulmonary (201), colorectal (202) and

368 breast (203) cancers. In fact, TN-C assembles into matrix tracks with ECM molecules such as

369 fibronectin, and several collagens (200,204), which are also linked to metastatic

370 potential (200,205,206). Evidence reveals that these TN-C matrix tracks have a functional

371 purpose in metastasis. In co-culture experiments, leading fibroblasts were able to create matrix

372 tracks composed of TN-C and fibronectin, which were left behind for the movement of SCC cells

373 (207). For fibronectin and TN-C to co-assemble into such tracks, POSTN is responsible for

374 incorporating TN-C into the meshwork architecture (208). While integrating TN-C, it is possible

375 that POSTN could also serve as a scaffold for BMP-1, LOX-1 and collagen to accelerate collagen

376 crosslinking into migratory tracks during metastasis (172). Mechanistically, track mobility

377 involves TN-C competing for syndecan-4 binding to fibronectin, which blocks integrin α5β1–

378 mediated cell adhesion for detachment (139), followed by TN-C promotion of migration

379 through integrin α9β1 following YAP inactivation (209). As previously discussed, TN-C and

380 POSTN can bind multiple ECM proteins (i.e. fibronectin and various collagens) and enzymes (i.e.

381 LOX) to serve as scaffolds of collagen crosslinking needed for cancer cell proliferation and

382 survival. This is a similar process that comes into play when TN-C and POSTN interact to build

383 ECM scaffolds for migration tracks (24,167,204,208,210).

18

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

384 Finally, for metastatic cells to exit the embedded state for intravasation, and then

385 return to the ECM for colonization after invasion at a distant site, a degree of matrix plasticity is

386 required. Such ECM remodeling is achieved by the degradative activity of extracellular

387 proteases (Fig. 1C), in particular metalloproteinases (MMPs). Like several other MCPs, SIBLINGs

388 bind and activate MMPs to promote metastasis. SPP1 binds CD44 to activate MMP-3 while BSP

389 binds to integrin αvβ3 to activate MMP-2 to increase invasiveness in various cancer cell types

390 (20,211). Furthermore, SPP1 and BSP bind and activate MMP-3 and MMP-2, respectively (20).

391 Early studies reported that SPARC upregulates the expression and activity of MMP-2 and MMP-

392 14 in glioma cells and MMP-2 in breast cancer cells (212,213). On the other hand, the SPARC

393 family member SPOCK2 was recently shown to inhibit the expression of MMP-2 and MMP14,

394 and activation of MMP-2 in endometrial cancer cells (214,215). TN-C may also influence the

395 invasion of chondrosarcoma, colon cancer and glioma cells by interacting with and upregulating

396 MMP-1, -2/9 and -12, respectively (216,217).

397 MCPs can also serve as a substrate to MMPs (218), i.e., SPARC and SPP1 in the case of

398 MMP-2, -3, -7, -9, -12 and -14, and MMP-3, -7, -9 and -12, respectively. From the SPARC family,

399 SPARCL1 and SPARC are cleaved by MMP-3 in gliomas (219) and cathepsin K in bone cancer

400 (220), respectively, whose fragments could affect SPARC activity. Recently, MMP-9 cleavage of

401 SPARC was reported to enhance SPARC-collagen binding, preventing collagen degradation by

402 MMPs in lung cancer (166). As for SPP1, thrombin and plasmin can cleave its C-terminal, which

403 increases adhesion of melanoma cells (221) and migration of breast cancer cells (222), while

404 cleavage of SPP1 by MMP-9 is essential for hepatocellular carcinoma invasion, which correlates

405 with metastatic potential (223).

19

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

406 Apart from targeting MMPs, the role of TN-C in influencing the ECM to promote invasion

407 is multifaceted. In Ewing sarcoma, TN-C expression and Src activation cooperate to promote

408 invadopodia formation, an actin-rich protrusion of the plasma membrane involved in

409 degradation of the ECM during cancer cell extravasation (224). In order for distant sites to

410 accommodate disseminated tumor cells, MCPs are also required at the secondary target tissue

411 to prime the metastatic niche for colonization. TN-C has been shown to be involved in

412 metastatic colonization since loss of this MCP in breast cancer, melanoma or metastatic niche

413 stromal cells inhibited colonization in the lungs (225-227). Gla-containing proteins have also

414 been implicated in establishing a metastatic niche. Tumor-derived POSTN was reported to form

415 a microenvironmental niche supportive of breast cancer stem cells via the integrin αvβ3/ERK

416 pathway (228). In various mouse models, POSTN was responsible for metastatic colonization of

417 the lung by breast and melanoma cells as evidenced by POSTN neutralizing antibodies,

418 antisense oligonucleotides and KO mice, all independently inhibiting metastasis (229-231).

419 Given their significance for breast cancer cell dissemination to the lungs, it remains possible

420 that both POSTN and TN-C are interdependent in promoting colonization of the metastatic

421 niche, since POSTN anchors TN-C to the ECM (208). MGP was also recently shown to influence

422 the metastatic niche by promoting osteosarcoma adhesion, extravasation and MMP activities in

423 murine lung endothelium in vitro (94).

424

425 Future Clinical Applications

426 MCPs are generally expressed at low levels in adult tissues but highly upregulated in

427 various pathologies or injuries (4-6). This has prompted researchers to elucidate the potential

20

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

428 functions of different MCPs in diseases such as cancer. As discussed throughout this review,

429 numerous studies have shown that MCPs play critical roles in cancer development. In addition,

430 the presence of certain MCPs in circulation as well as diseased tissue indicate their utility as

431 non-invasive diagnostic and prognostic cancer biomarkers. Furthermore, their extracellular

432 location and involvement in cancer pathology indicate that MCPs represent accessible and

433 potentially effective therapeutic targets. In the following sections, we discuss various pre-

434 clinical studies and clinical trials exploring the above possibilities.

435 MCPs as Cancer Biomarkers

436 SPARC has been suggested as a prognostic biomarker for certain cancers such as soft

437 tissue sarcoma, esophageal SCC and glioblastoma since its expression correlates with poor

438 survival (232-234). In addition, SPP1 may be prognostic for breast, lung, gastric, liver and colon

439 cancers since it associated with tumor progression and decreased patient survival (235-238).

440 Subsequently, a number of ongoing clinical trials have been established to validate their

441 application. Recently, SPARC has been the subject of a clinical study probing its utility as a

442 diagnostic marker for brain cancer (registered number clinical trial (NCT) 01012609), given prior

443 investigations correlating increased tumor vascular SPARC expression with decreased brain

444 cancer patient survival (239). Several groups have also reported that high plasma SPP1

445 concentrations might be predictive of poor outcome for several cancers, including breast

446 cancer (240). Consequently, one clinical study is currently probing the relevance of SPP1 serum

447 levels for diagnosis of breast cancer (NCT 02895178). Other MCP families await successful

448 clinical trials since the expression of several CCN family members in pancreatic, breast, oral,

449 esophageal and brain cancers (241-245), TN-C in colorectal, glioma, pancreatic and bladder

21

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

450 cancers (196,246-248), and POSTN in various solid cancers (249) have all been touted as

451 potential diagnostic and prognostic biomarkers.

452 MCPs as Therapeutic Targets

453 Targeting MCPs for therapeutic purposes has received relatively little attention,

454 primarily due to limited data concerning mechanisms of action. The fact that MCPs are located

455 in the extracellular space during cancer development renders them attractive as accessible

456 targets for drug delivery; moreover, their context-specific expression implies that targeting

457 these proteins would result in minimum pleiotropic side-effects. Neutralizing antibodies against

458 MCPs have shown success in various preclinical settings; however, translation to the clinic has

459 been difficult. One group showed that an SPP1 monoclonal antibody (AOM1) significantly

460 inhibited tumor growth and metastasis in a mouse model of non-small lung cancer (250). In

461 addition, a commonly-used monoclonal antibody for antagonizing CCN2 (FG-3019) has

462 reportedly been used in preclinical models with success in both monotherapy and combination

463 therapy for different tumor types, including pancreatic and melanoma (251-256). With such

464 progress, neutralizing-antibodies targeting MCPs have advanced to registered clinical trials. For

465 example, FG-3019 is currently in phase III for CCN2-targeted treatment of pancreatic cancer

466 (LAPIS, NCT03941093).

467 Alternatively, MCPs could be targeted by inhibiting in patients. In fact,

468 one of the first studies using RNA interference (RNAi) to treat cancer with promising results

469 involved targeting TN-C in 11 glioma patients (257). This was followed up with an investigation

470 of a larger cohort of 46 patients, which reported significant improvement in overall survival

471 (258). Other promising MCP targets for posttranscriptional gene silencing include SPP1, POSTN,

22

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

472 CCN1 and CCN2, where inhibition of RNA expression was shown to reduce cancer progression in

473 various animal models (68,73,229,235). Another therapeutic approach involves exploiting the

474 high expression of MCP within the tumor environment as a strategy to deliver therapeutic

475 molecules. Using a high affinity antibody to deliver radiotherapy (monoclonal antibody 81C6),

476 TN-C was targeted for treatment of glioma and lymphoma (259,260), showing safe and

477 promising anti-tumor benefit.

478

479 Conclusion and Future Perspective

480 Upon perturbation of tissue homeostasis during multistage carcinogenesis, MCPs are

481 upregulated in the tumor microenvironment to become key mediators of cell-ECM

482 communication that in turn promotes cellular proliferation, survival and metastasis. The

483 functional diversity of MCPs stems from their ability to interact with a variety of extracellular

484 signaling molecules such as ECM components and growth factors. Moreover, as emphasized in

485 this review, many MCPs have been implicated in cancer development, and thus may certainly

486 exert additive and/or antagonistic effects in this process. Our current understanding of MCP

487 pathways gives an impression of redundancy, and so a primary aim in the ECM field is to

488 elucidate the precise manner in which MCPs mechanistically converge, both functionally and

489 temporally, to remodel the tumor microenvironment and orchestrate critical neoplastic

490 processes.

491 This overarching goal highlights a major challenge, that of developing experimental

492 systems which better model the physical state of the native interstitial ECM. The usefulness of

23

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

493 various existing models, including 2D monolayers (261), 3D matrigels (262) and tissue-extracted

494 ECM (263), is limited since these models fall well short of fully recapitulating the complexities of

495 tissue ECM in vivo. This inadequacy may underlie some of the discrepancies in the literature

496 regarding MCP functionality in cancer. Furthermore, the identification of naturally occurring

497 protein-protein interactions and post-translational modifications among MCPs in the ECM have

498 been difficult to characterize. Overall, as concisely reviewed elsewhere (264), new approaches

499 are clearly needed to dissect the daunting complexity of the ECM environment and its role in

500 carcinogenesis.

501 While confronting the above challenges it remains important to concomitantly work

502 towards characterizing particular MCPs, alone or in combination, as impactful

503 diagnostic/prognostic cancer biomarkers and therapeutic targets. In fact, given their

504 burgeoning roles in cancer development and extracellular accessibility, MCPs have long been

505 regarded as potentially useful for diagnosing and treating various pathologies such as

506 and cancer; nonetheless clinical data supporting this notion have been relatively scant. Towards

507 addressing this knowledge gap, over the past decade, progress has been made in defining

508 better the fundamental mechanisms of MCPs, opening new questions that entice the

509 generation of the next needed tools to understand sufficient detail for optimal therapeutic

510 design.

511 Herein we have summarized some important ways in which misregulation of MCP

512 expression promotes cancer development, including perturbation of intracellular signaling and

513 aberrant coordination of ECM remodeling. Although we focused on MCP families with the most

514 well-characterized roles, others are emerging as potentially important players, such as the

24

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

515 EMILIN and R-Spondin families. Recently, R-Spondin-1 and 2 were shown to promote liver,

516 glioblastoma and ovarian cancer through their well-defined influence on Wnt/β-catenin

517 signaling (265-267). In addition, EMILIN2 promotes the formation of tumor-associated vessels

518 in melanoma (268), and EMILIN1 exerts an oncosuppressive role in colon and skin (269,270).

519 Clearly, there is still much to be discovered regarding the exquisite spatiotemporal regulation of

520 MCP expression patterns and functions in the extracellular space during cancer tissue

521 remodeling, similar to the approach taken in fibrosis (17). In this respect, as more and more

522 knowledge accumulates, it should be possible to design appropriate clinical studies that could

523 firmly establish MCPs as useful biomarkers and therapeutic targets in cancer.

524

525 Acknowledgements

526 This work was supported by the Operating Grant Funding Program 24347 (co-funded by Cancer

527 Research Society and the Kidney Cancer Research Network of Canada to C. Gerarduzzi), and

528 start-up funds from Hôpital Maisonneuve-Rosemont Foundation (to C. Gerarduzzi). C.

529 Gerarduzzi is a recipient of the Kidney Research Scientist Core Education and National Training

530 (KRESCENT) Program New Investigator Award KRES180003 (co-funded by the Kidney

531 Foundation of Canada, Canadian Society of Nephrology, and Canadian Institutes of Health

532 Research) and the Cole Foundation Early Career Transition Award.

533

25

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

534 References

535 1. Bornstein P. Matricellular proteins: an overview. Matrix biology : journal of the 536 International Society for Matrix Biology 2000;19:555-6 537 2. Bornstein P. Matricellular proteins: an overview. J Cell Commun Signal 2009;3:163-5 538 3. Roberts DD. Emerging functions of matricellular proteins. Cell Mol Life Sci 2011;68:3133- 539 6 540 4. Matsui Y, Morimoto J, Uede T. Role of matricellular proteins in cardiac tissue remodeling 541 after myocardial infarction. World J Biol Chem 2010;1:69-80 542 5. Wong GS, Rustgi AK. Matricellular proteins: priming the tumour microenvironment for 543 cancer development and metastasis. Br J Cancer 2013;108:755-61 544 6. Frangogiannis NG. Matricellular proteins in cardiac adaptation and disease. Physiol Rev 545 2012;92:635-88 546 7. Lu P, Weaver VM, Werb Z. The extracellular matrix: a dynamic niche in cancer 547 progression. The Journal of cell biology 2012;196:395-406 548 8. Chiodoni C, Colombo MP, Sangaletti S. Matricellular proteins: from homeostasis to 549 inflammation, cancer, and metastasis. Cancer Metastasis Rev 2010;29:295-307 550 9. Sawyer AJ, Kyriakides TR. Matricellular proteins in drug delivery: Therapeutic targets, 551 active agents, and therapeutic localization. Adv Drug Deliv Rev 2016;97:56-68 552 10. Bradshaw AD. Diverse biological functions of the SPARC family of proteins. The 553 international journal of biochemistry & cell biology 2012;44:480-8 554 11. Jun JI, Lau LF. Taking aim at the extracellular matrix: CCN proteins as emerging 555 therapeutic targets. Nat Rev Drug Discov 2011;10:945-63 556 12. Lau LF. Cell surface receptors for CCN proteins. J Cell Commun Signal 2016;10:121-7 557 13. Quensel K, Shi-wen X, Hutchenreuther J, Xiao Y, Liu S, Peidl A, et al. CCN1 expression by 558 fibroblasts is required for bleomycin-induced skin fibrosis. Matrix Biology Plus 559 2019;3:100009 560 14. Chiovaro F, Chiquet-Ehrismann R, Chiquet M. Transcriptional regulation of tenascin 561 . Cell Adh Migr 2015;9:34-47 562 15. Chiquet-Ehrismann R, Tucker RP. Tenascins and the importance of adhesion modulation. 563 Cold Spring Harb Perspect Biol 2011;3 564 16. Kruger TE, Miller AH, Godwin AK, Wang J. Bone sialoprotein and osteopontin in bone 565 metastasis of osteotropic cancers. Crit Rev Oncol Hematol 2014;89:330-41 566 17. Feng D, Ngov C, Henley N, Boufaied N, Gerarduzzi C. Characterization of Matricellular 567 Protein Expression Signatures in Mechanistically Diverse Mouse Models of Kidney Injury. 568 Sci Rep 2019;9:16736 569 18. Jiang C, Zurick K, Qin C, Bernards MT. Probing the influence of SIBLING proteins on 570 collagen-I fibrillogenesis and denaturation. Connect Tissue Res 2018;59:274-86 571 19. Luo X, Ruhland MK, Pazolli E, Lind AC, Stewart SA. Osteopontin stimulates preneoplastic 572 cellular proliferation through activation of the MAPK pathway. Mol Cancer Res 573 2011;9:1018-29

26

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

574 20. Fedarko NS, Jain A, Karadag A, Fisher LW. Three small integrin binding ligand N-linked 575 glycoproteins (SIBLINGs) bind and activate specific matrix metalloproteinases. FASEB J 576 2004;18:734-6 577 21. Vermeer C. Vitamin K: the effect on health beyond coagulation - an overview. Food Nutr 578 Res 2012;56 579 22. Gonzalez-Gonzalez L, Alonso J. Periostin: A Matricellular Protein With Multiple Functions 580 in Cancer Development and Progression. Front Oncol 2018;8:225 581 23. Nishimoto SK, Nishimoto M. binds to fibronectin and enhances cell 582 attachment and spreading on fibronectin. Int J Cell Biol 2014;2014:807013 583 24. Kudo A. Periostin in fibrillogenesis for tissue regeneration: periostin actions inside and 584 outside the cell. Cell Mol Life Sci 2011;68:3201-7 585 25. Zhu S, Barbe MF, Liu C, Hadjiargyrou M, Popoff SN, Rani S, et al. Periostin-like-factor in 586 osteogenesis. J Cell Physiol 2009;218:584-92 587 26. Kuhn B, del Monte F, Hajjar RJ, Chang YS, Lebeche D, Arab S, et al. Periostin induces 588 proliferation of differentiated cardiomyocytes and promotes cardiac repair. Nat Med 589 2007;13:962-9 590 27. Bostrom K, Zebboudj AF, Yao Y, Lin TS, Torres A. Matrix GLA protein stimulates VEGF 591 expression through increased transforming growth factor-beta1 activity in endothelial 592 cells. The Journal of biological chemistry 2004;279:52904-13 593 28. Gheorghe SR, Craciun AM. Matrix Gla protein in tumoral pathology. Clujul Med 594 2016;89:319-21 595 29. Gerarduzzi C, Kumar RK, Trivedi P, Ajay AK, Iyer A, Boswell S, et al. Silencing SMOC2 596 ameliorates kidney fibrosis by inhibiting fibroblast to transformation. JCI 597 Insight 2017;2 598 30. Hartmann U, Hulsmann H, Seul J, Roll S, Midani H, Breloy I, et al. Testican-3: a brain- 599 specific proteoglycan member of the BM-40/SPARC/osteonectin family. J Neurochem 600 2013;125:399-409 601 31. Roll S, Seul J, Paulsson M, Hartmann U. Testican-1 is dispensable for mouse 602 development. Matrix Biol 2006;25:373-81 603 32. Bradshaw AD, Puolakkainen P, Dasgupta J, Davidson JM, Wight TN, Helene Sage E. 604 SPARC-null mice display abnormalities in the characterized by decreased 605 collagen fibril diameter and reduced tensile strength. J Invest Dermatol 2003;120:949- 606 55 607 33. Bradshaw AD, Sage EH. SPARC, a matricellular protein that functions in cellular 608 differentiation and tissue response to injury. J Clin Invest 2001;107:1049-54 609 34. McKinnon PJ, McLaughlin SK, Kapsetaki M, Margolskee RF. Extracellular matrix- 610 associated protein Sc1 is not essential for mouse development. Mol Cell Biol 611 2000;20:656-60 612 35. Maeda A, Ono M, Holmbeck K, Li L, Kilts TM, Kram V, et al. WNT1-induced Secreted 613 Protein-1 (WISP1), a Novel Regulator of Bone Turnover and Wnt Signaling. J Biol Chem 614 2015;290:14004-18 615 36. Midwood KS, Hussenet T, Langlois B, Orend G. Advances in tenascin-C biology. Cell Mol 616 Life Sci 2011;68:3175-99

27

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

617 37. Forsberg E, Hirsch E, Frohlich L, Meyer M, Ekblom P, Aszodi A, et al. Skin wounds and 618 severed nerves heal normally in mice lacking tenascin-C. Proc Natl Acad Sci U S A 619 1996;93:6594-9 620 38. Saga Y, Yagi T, Ikawa Y, Sakakura T, Aizawa S. Mice develop normally without tenascin. 621 Genes Dev 1992;6:1821-31 622 39. Nagao T, Okura T, Irita J, Jotoku M, Enomoto D, Desilva VR, et al. Osteopontin plays a 623 critical role in interstitial fibrosis but not glomerular sclerosis in diabetic nephropathy. 624 Nephron Extra 2012;2:87-103 625 40. Franzen A, Hultenby K, Reinholt FP, Onnerfjord P, Heinegard D. Altered osteoclast 626 development and function in osteopontin deficient mice. J Orthop Res 2008;26:721-8 627 41. Rittling SR, Matsumoto HN, McKee MD, Nanci A, An XR, Novick KE, et al. Mice lacking 628 osteopontin show normal development and bone structure but display altered 629 osteoclast formation in vitro. J Bone Miner Res 1998;13:1101-11 630 42. Liaw L, Birk DE, Ballas CB, Whitsitt JS, Davidson JM, Hogan BL. Altered wound healing in 631 mice lacking a functional osteopontin gene (spp1). J Clin Invest 1998;101:1468-78 632 43. Bozyk PD, Bentley JK, Popova AP, Anyanwu AC, Linn MD, Goldsmith AM, et al. Neonatal 633 periostin knockout mice are protected from hyperoxia-induced alveolar simplication. 634 PLoS One 2012;7:e31336 635 44. Elliott CG, Wang J, Guo X, Xu SW, Eastwood M, Guan J, et al. Periostin modulates 636 myofibroblast differentiation during full-thickness cutaneous wound repair. J Cell Sci 637 2012;125:121-32 638 45. Rios H, Koushik SV, Wang H, Wang J, Zhou HM, Lindsley A, et al. periostin null mice 639 exhibit dwarfism, incisor enamel defects, and an early-onset periodontal disease-like 640 phenotype. Mol Cell Biol 2005;25:11131-44 641 46. Ivkovic S, Yoon BS, Popoff SN, Safadi FF, Libuda DE, Stephenson RC, et al. Connective 642 tissue growth factor coordinates chondrogenesis and angiogenesis during skeletal 643 development. Development 2003;130:2779-91 644 47. Mo FE, Muntean AG, Chen CC, Stolz DB, Watkins SC, Lau LF. CYR61 (CCN1) is essential 645 for placental development and vascular integrity. Mol Cell Biol 2002;22:8709-20 646 48. Marulanda J, Eimar H, McKee MD, Berkvens M, Nelea V, Roman H, et al. Matrix Gla 647 protein deficiency impairs nasal septum growth, causing midface hypoplasia. J Biol 648 Chem 2017;292:11400-12 649 49. Yao Y, Yao J, Radparvar M, Blazquez-Medela AM, Guihard PJ, Jumabay M, et al. Reducing 650 Jagged 1 and 2 levels prevents cerebral arteriovenous malformations in matrix Gla 651 protein deficiency. Proc Natl Acad Sci U S A 2013;110:19071-6 652 50. Luo G, Ducy P, McKee MD, Pinero GJ, Loyer E, Behringer RR, et al. Spontaneous 653 calcification of and in mice lacking matrix GLA protein. Nature 654 1997;386:78-81 655 51. Viloria K, Hill NJ. Embracing the complexity of matricellular proteins: the functional and 656 clinical significance of splice variation. Biomol Concepts 2016;7:117-32 657 52. Wu T, Ouyang G. Matricellular proteins: multifaceted extracellular regulators in tumor 658 dormancy. Protein Cell 2014;5:249-52

28

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

659 53. Shi D, Jiang K, Fu Y, Fang R, Liu XI, Chen J. Overexpression of SPARC correlates with poor 660 prognosis in patients with cervical carcinoma and regulates cancer cell epithelial- 661 mesenchymal transition. Oncol Lett 2016;11:3251-8 662 54. Botti G, Scognamiglio G, Marra L, Collina F, Di Bonito M, Cerrone M, et al. 663 SPARC/osteonectin is involved in metastatic process to the lung during melanoma 664 progression. Virchows Arch 2014;465:331-8 665 55. Hsiao YH, Lien HC, Hwa HL, Kuo WH, Chang KJ, Hsieh FJ. SPARC (osteonectin) in breast 666 tumors of different histologic types and its role in the outcome of invasive ductal 667 carcinoma. Breast J 2010;16:305-8 668 56. Seno T, Harada H, Kohno S, Teraoka M, Inoue A, Ohnishi T. Downregulation of SPARC 669 expression inhibits cell migration and invasion in malignant gliomas. Int J Oncol 670 2009;34:707-15 671 57. Nagaraju GP, Dontula R, El-Rayes BF, Lakka SS. Molecular mechanisms underlying the 672 divergent roles of SPARC in human carcinogenesis. Carcinogenesis 2014;35:967-73 673 58. Chan QK, Ngan HY, Ip PP, Liu VW, Xue WC, Cheung AN. Tumor suppressor effect of 674 follistatin-like 1 in ovarian and endometrial carcinogenesis: a differential expression and 675 functional analysis. Carcinogenesis 2009;30:114-21 676 59. Singh M, Venugopal C, Tokar T, Brown KR, McFarlane N, Bakhshinyan D, et al. RNAi 677 screen identifies essential regulators of human brain metastasis-initiating cells. Acta 678 Neuropathol 2017;134:923-40 679 60. Perurena N, Zandueta C, Martinez-Canarias S, Moreno H, Vicent S, Almeida AS, et al. 680 EPCR promotes breast cancer progression by altering SPOCK1/testican 1-mediated 3D 681 growth. J Hematol Oncol 2017;10:23 682 61. Ma LJ, Wu WJ, Wang YH, Wu TF, Liang PI, Chang IW, et al. SPOCK1 Overexpression 683 Confers a Poor Prognosis in Urothelial Carcinoma. J Cancer 2016;7:467-76 684 62. Nakada M, Miyamori H, Yamashita J, Sato H. Testican 2 abrogates inhibition of 685 membrane-type matrix metalloproteinases by other testican family proteins. Cancer Res 686 2003;63:3364-9 687 63. Lu H, Ju DD, Yang GD, Zhu LY, Yang XM, Li J, et al. Targeting cancer stem cell signature 688 gene SMOC-2 Overcomes chemoresistance and inhibits cell proliferation of endometrial 689 carcinoma. EBioMedicine 2019;40:276-89 690 64. Su JR, Kuai JH, Li YQ. Smoc2 potentiates proliferation of hepatocellular carcinoma cells 691 via promotion of cell cycle progression. World J Gastroenterol 2016;22:10053-63 692 65. Shvab A, Haase G, Ben-Shmuel A, Gavert N, Brabletz T, Dedhar S, et al. Induction of the 693 intestinal stem cell signature gene SMOC-2 is required for L1-mediated colon cancer 694 progression. Oncogene 2016;35:549-57 695 66. Brellier F, Ruggiero S, Zwolanek D, Martina E, Hess D, Brown-Luedi M, et al. SMOC1 is a 696 tenascin-C interacting protein over-expressed in brain tumors. Matrix Biol 2011;30:225- 697 33 698 67. Haque I, Mehta S, Majumder M, Dhar K, De A, McGregor D, et al. Cyr61/CCN1 signaling 699 is critical for epithelial-mesenchymal transition and stemness and promotes pancreatic 700 carcinogenesis. Mol Cancer 2011;10:8

29

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

701 68. Goodwin CR, Lal B, Zhou X, Ho S, Xia S, Taeger A, et al. Cyr61 mediates hepatocyte 702 growth factor-dependent tumor cell growth, migration, and Akt activation. Cancer Res 703 2010;70:2932-41 704 69. Lv H, Fan E, Sun S, Ma X, Zhang X, Han DM, et al. Cyr61 is up-regulated in prostate 705 cancer and associated with the p53 gene status. J Cell Biochem 2009;106:738-44 706 70. Tsai MS, Bogart DF, Castaneda JM, Li P, Lupu R. Cyr61 promotes breast tumorigenesis 707 and cancer progression. Oncogene 2002;21:8178-85 708 71. Wells JE, Howlett M, Cole CH, Kees UR. Deregulated expression of connective tissue 709 growth factor (CTGF/CCN2) is linked to poor outcome in human cancer. Int J Cancer 710 2015;137:504-11 711 72. Mao Z, Ma X, Rong Y, Cui L, Wang X, Wu W, et al. Connective tissue growth factor 712 enhances the migration of gastric cancer through downregulation of E-cadherin via the 713 NF-kappaB pathway. Cancer Sci 2011;102:104-10 714 73. Bennewith KL, Huang X, Ham CM, Graves EE, Erler JT, Kambham N, et al. The role of 715 tumor cell-derived connective tissue growth factor (CTGF/CCN2) in pancreatic tumor 716 growth. Cancer Res 2009;69:775-84 717 74. Hutchenreuther J, Vincent KM, Carter DE, Postovit LM, Leask A. CCN2 Expression by 718 Tumor Stroma Is Required for Melanoma Metastasis. J Invest Dermatol 2015;135:2805- 719 13 720 75. Nallet-Staub F, Marsaud V, Li L, Gilbert C, Dodier S, Bataille V, et al. Pro-invasive activity 721 of the Hippo pathway effectors YAP and TAZ in cutaneous melanoma. J Invest Dermatol 722 2014;134:123-32 723 76. Wang MY, Chen PS, Prakash E, Hsu HC, Huang HY, Lin MT, et al. Connective tissue 724 growth factor confers drug resistance in breast cancer through concomitant up- 725 regulation of Bcl-xL and cIAP1. Cancer Res 2009;69:3482-91 726 77. Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, et al. A 727 framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev 728 Cancer 2020 729 78. Hutchenreuther J, Vincent K, Norley C, Racanelli M, Gruber SB, Johnson TM, et al. 730 Activation of cancer-associated fibroblasts is required for tumor neovascularization in a 731 murine model of melanoma. Matrix Biol 2018;74:52-61 732 79. Erez N, Truitt M, Olson P, Arron ST, Hanahan D. Cancer-Associated Fibroblasts Are 733 Activated in Incipient Neoplasia to Orchestrate Tumor-Promoting Inflammation in an 734 NF-kappaB-Dependent Manner. Cancer Cell 2010;17:135-47 735 80. Chen CC, Kim KH, Lau LF. The matricellular protein CCN1 suppresses 736 hepatocarcinogenesis by inhibiting compensatory proliferation. Oncogene 737 2016;35:1314-23 738 81. Chang CC, Yang MH, Lin BR, Chen ST, Pan SH, Hsiao M, et al. CCN2 inhibits lung cancer 739 metastasis through promoting DAPK-dependent anoikis and inducing EGFR degradation. 740 Cell Death Differ 2013;20:443-55 741 82. Li J, Ye L, Owen S, Weeks HP, Zhang Z, Jiang WG. Emerging role of CCN family proteins in 742 tumorigenesis and cancer metastasis (Review). Int J Mol Med 2015;36:1451-63

30

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

743 83. Sharon Y, Raz Y, Cohen N, Ben-Shmuel A, Schwartz H, Geiger T, et al. Tumor-derived 744 osteopontin reprograms normal mammary fibroblasts to promote inflammation and 745 tumor growth in breast cancer. Cancer Res 2015;75:963-73 746 84. Wang L, Song L, Li J, Wang Y, Yang C, Kou X, et al. Bone sialoprotein-alphavbeta3 integrin 747 axis promotes breast cancer metastasis to the bone. Cancer Sci 2019;110:3157-72 748 85. Cho WY, Hong SH, Singh B, Islam MA, Lee S, Lee AY, et al. Suppression of tumor growth 749 in lung cancer xenograft model mice by poly(sorbitol-co-PEI)-mediated delivery of 750 osteopontin siRNA. Eur J Pharm Biopharm 2015;94:450-62 751 86. Zhang L, Pu D, Liu D, Wang Y, Luo W, Tang H, et al. Identification and validation of novel 752 circulating biomarkers for early diagnosis of lung cancer. Lung Cancer 2019;135:130-7 753 87. Fedarko NS, Jain A, Karadag A, Van Eman MR, Fisher LW. Elevated serum bone 754 sialoprotein and osteopontin in colon, breast, prostate, and lung cancer. Clin Cancer Res 755 2001;7:4060-6 756 88. Sun T, Li P, Sun D, Bu Q, Li G. Prognostic value of osteopontin in patients with 757 hepatocellular carcinoma: A systematic review and meta-analysis. Medicine (Baltimore) 758 2018;97:e12954 759 89. Loosen SH, Hoening P, Puethe N, Luedde M, Spehlmann M, Ulmer TF, et al. Elevated 760 serum levels of bone sialoprotein (BSP) predict long-term mortality in patients with 761 pancreatic adenocarcinoma. Sci Rep 2019;9:1489 762 90. Ng L, Wan T, Chow A, Iyer D, Man J, Chen G, et al. Osteopontin Overexpression Induced 763 Tumor Progression and Chemoresistance to Oxaliplatin through Induction of Stem-Like 764 Properties in Human Colorectal Cancer. Stem Cells Int 2015;2015:247892 765 91. Weber GF, Lett GS, Haubein NC. Categorical meta-analysis of Osteopontin as a clinical 766 cancer marker. Oncol Rep 2011;25:433-41 767 92. Anderberg C, Li H, Fredriksson L, Andrae J, Betsholtz C, Li X, et al. Paracrine signaling by 768 platelet-derived growth factor-CC promotes tumor growth by recruitment of cancer- 769 associated fibroblasts. Cancer Res 2009;69:369-78 770 93. Caiado H, Conceicao N, Tiago D, Marreiros A, Vicente S, Enriquez JL, et al. Evaluation of 771 MGP gene expression in colorectal cancer. Gene 2020;723:144120 772 94. Zandueta C, Ormazabal C, Perurena N, Martinez-Canarias S, Zalacain M, Julian MS, et al. 773 Matrix-Gla protein promotes osteosarcoma lung metastasis and associates with poor 774 prognosis. J Pathol 2016;239:438-49 775 95. Mertsch S, Schurgers LJ, Weber K, Paulus W, Senner V. Matrix gla protein (MGP): an 776 overexpressed and migration-promoting mesenchymal component in glioblastoma. 777 BMC Cancer 2009;9:302 778 96. Yoshimura K, Takeuchi K, Nagasaki K, Ogishima S, Tanaka H, Iwase T, et al. Prognostic 779 value of matrix Gla protein in breast cancer. Mol Med Rep 2009;2:549-53 780 97. Chen Y, Miller C, Mosher R, Zhao X, Deeds J, Morrissey M, et al. Identification of cervical 781 cancer markers by cDNA and tissue microarrays. Cancer Res 2003;63:1927-35 782 98. Takada Y, Ye X, Simon S. The integrins. Genome Biol 2007;8:215 783 99. Chong HC, Tan CK, Huang RL, Tan NS. Matricellular proteins: a sticky affair with cancers. 784 J Oncol 2012;2012:351089

31

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

785 100. Bellahcene A, Castronovo V, Ogbureke KU, Fisher LW, Fedarko NS. Small integrin-binding 786 ligand N-linked glycoproteins (SIBLINGs): multifunctional proteins in cancer. Nat Rev 787 Cancer 2008;8:212-26 788 101. Murphy-Ullrich JE, Sage EH. Revisiting the matricellular concept. Matrix Biol 2014;37:1- 789 14 790 102. Tseng C, Kolonin MG. Proteolytic Isoforms of SPARC Induce Adipose Stromal Cell 791 Mobilization in Obesity. Stem Cells 2016;34:174-90 792 103. Nie J, Chang B, Traktuev DO, Sun J, March K, Chan L, et al. IFATS collection: 793 Combinatorial peptides identify alpha5beta1 integrin as a receptor for the matricellular 794 protein SPARC on adipose stromal cells. Stem Cells 2008;26:2735-45 795 104. Shi Q, Bao S, Song L, Wu Q, Bigner DD, Hjelmeland AB, et al. Targeting SPARC expression 796 decreases glioma cellular survival and invasion associated with reduced activities of FAK 797 and ILK kinases. Oncogene 2007;26:4084-94 798 105. De S, Chen J, Narizhneva NV, Heston W, Brainard J, Sage EH, et al. Molecular pathway 799 for cancer metastasis to bone. The Journal of biological chemistry 2003;278:39044-50 800 106. Sun W, Feng J, Yi Q, Xu X, Chen Y, Tang L. SPARC acts as a mediator of TGF-beta1 in 801 promoting epithelial-to-mesenchymal transition in A549 and H1299 lung cancer cells. 802 Biofactors 2018;44:453-64 803 107. Tumbarello DA, Andrews MR, Brenton JD. SPARC Regulates Transforming Growth Factor 804 Beta Induced (TGFBI) Extracellular Matrix Deposition and Paclitaxel Response in Ovarian 805 Cancer Cells. PLoS One 2016;11:e0162698 806 108. Chandrasekaran V, Ambati J, Ambati BK, Taylor EW. Molecular docking and analysis of 807 interactions between vascular endothelial growth factor (VEGF) and SPARC protein. J 808 Mol Graph Model 2007;26:775-82 809 109. Raines EW, Lane TF, Iruela-Arispe ML, Ross R, Sage EH. The extracellular glycoprotein 810 SPARC interacts with platelet-derived growth factor (PDGF)-AB and -BB and inhibits the 811 binding of PDGF to its receptors. Proc Natl Acad Sci U S A 1992;89:1281-5 812 110. Miao L, Wang Y, Xia H, Yao C, Cai H, Song Y. SPOCK1 is a novel transforming growth 813 factor-beta target gene that regulates lung cancer cell epithelial-mesenchymal 814 transition. Biochem Biophys Res Commun 2013;440:792-7 815 111. Zhao P, Guan HT, Dai ZJ, Ma YG, Liu XX, Wang XJ. Knockdown of SPOCK1 Inhibits the 816 Proliferation and Invasion in Colorectal Cancer Cells by Suppressing the PI3K/Akt 817 Pathway. Oncol Res 2016;24:437-45 818 112. Shu YJ, Weng H, Ye YY, Hu YP, Bao RF, Cao Y, et al. SPOCK1 as a potential cancer 819 prognostic marker promotes the proliferation and metastasis of gallbladder cancer cells 820 by activating the PI3K/AKT pathway. Mol Cancer 2015;14:12 821 113. Liu P, Lu J, Cardoso WV, Vaziri C. The SPARC-related factor SMOC-2 promotes growth 822 factor-induced cyclin D1 expression and DNA synthesis via integrin-linked kinase. Mol 823 Biol Cell 2008;19:248-61 824 114. Zheng CC, Hu HF, Hong P, Zhang QH, Xu WW, He QY, et al. Significance of integrin-linked 825 kinase (ILK) in tumorigenesis and its potential implication as a biomarker and 826 therapeutic target for human cancer. Am J Cancer Res 2019;9:186-97

32

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

827 115. Maier S, Paulsson M, Hartmann U. The widely expressed extracellular matrix protein 828 SMOC-2 promotes keratinocyte attachment and migration. Experimental cell research 829 2008;314:2477-87 830 116. Thakur R, Mishra DP. Matrix reloaded: CCN, tenascin and SIBLING group of matricellular 831 proteins in orchestrating cancer hallmark capabilities. Pharmacol Ther 2016;168:61-74 832 117. Haque I, De A, Majumder M, Mehta S, McGregor D, Banerjee SK, et al. The matricellular 833 protein CCN1/Cyr61 is a critical regulator of Sonic Hedgehog in pancreatic 834 carcinogenesis. J Biol Chem 2012;287:38569-79 835 118. Huang YT, Lan Q, Ponsonnet L, Blanquet M, Christofori G, Zaric J, et al. The matricellular 836 protein CYR61 interferes with normal pancreatic islets architecture and promotes 837 pancreatic neuroendocrine tumor progression. Oncotarget 2016;7:1663-74 838 119. Xie D, Yin D, Tong X, O'Kelly J, Mori A, Miller C, et al. Cyr61 is overexpressed in gliomas 839 and involved in integrin-linked kinase-mediated Akt and beta-catenin-TCF/Lef signaling 840 pathways. Cancer Res 2004;64:1987-96 841 120. Huang YT, Lan Q, Lorusso G, Duffey N, Ruegg C. The matricellular protein CYR61 842 promotes breast cancer lung metastasis by facilitating tumor cell extravasation and 843 suppressing anoikis. Oncotarget 2017;8:9200-15 844 121. Menendez JA, Vellon L, Mehmi I, Teng PK, Griggs DW, Lupu R. A novel CYR61-triggered 845 'CYR61-alphavbeta3 integrin loop' regulates breast cancer cell survival and 846 chemosensitivity through activation of ERK1/ERK2 MAPK signaling pathway. Oncogene 847 2005;24:761-79 848 122. Chien W, O'Kelly J, Lu D, Leiter A, Sohn J, Yin D, et al. Expression of connective tissue 849 growth factor (CTGF/CCN2) in breast cancer cells is associated with increased migration 850 and angiogenesis. Int J Oncol 2011;38:1741-7 851 123. Stephens S, Palmer J, Konstantinova I, Pearce A, Jarai G, Day E. A functional analysis of 852 Wnt inducible signalling pathway protein -1 (WISP-1/CCN4). J Cell Commun Signal 853 2015;9:63-72 854 124. Haque I, Banerjee S, De A, Maity G, Sarkar S, Majumdar M, et al. CCN5/WISP-2 promotes 855 growth arrest of triple-negative breast cancer cells through accumulation and trafficking 856 of p27(Kip1) via Skp2 and FOXO3a regulation. Oncogene 2015;34:3152-63 857 125. Kaasboll OJ, Gadicherla AK, Wang JH, Monsen VT, Hagelin EMV, Dong MQ, et al. 858 Connective tissue growth factor (CCN2) is a matricellular preproprotein controlled by 859 proteolytic activation. J Biol Chem 2018;293:17953-70 860 126. Brigstock DR, Steffen CL, Kim GY, Vegunta RK, Diehl JR, Harding PA. Purification and 861 characterization of novel heparin-binding growth factors in uterine secretory fluids. 862 Identification as heparin-regulated Mr 10,000 forms of connective tissue growth factor. 863 J Biol Chem 1997;272:20275-82 864 127. Li J, Ye L, Sun PH, Zheng F, Ruge F, Satherley LK, et al. Reduced NOV expression 865 correlates with disease progression in colorectal cancer and is associated with survival, 866 invasion and chemoresistance of cancer cells. Oncotarget 2017;8:26231-44 867 128. Chen PC, Cheng HC, Tang CH. CCN3 promotes prostate cancer bone metastasis by 868 modulating the tumor-bone microenvironment through RANKL-dependent pathway. 869 Carcinogenesis 2013;34:1669-79

33

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

870 129. Tai HC, Chang AC, Yu HJ, Huang CY, Tsai YC, Lai YW, et al. Osteoblast-derived WNT- 871 induced secreted protein 1 increases VCAM-1 expression and enhances prostate cancer 872 metastasis by down-regulating miR-126. Oncotarget 2014;5:7589-98 873 130. Chang AC, Chen PC, Lin YF, Su CM, Liu JF, Lin TH, et al. Osteoblast-secreted WISP-1 874 promotes adherence of prostate cancer cells to bone via the VCAM-1/integrin 875 alpha4beta1 system. Cancer Lett 2018;426:47-56 876 131. Tsai HC, Tzeng HE, Huang CY, Huang YL, Tsai CH, Wang SW, et al. WISP-1 positively 877 regulates angiogenesis by controlling VEGF-A expression in human osteosarcoma. Cell 878 Death Dis 2017;8:e2750 879 132. Lin CC, Chen PC, Lein MY, Tsao CW, Huang CC, Wang SW, et al. WISP-1 promotes VEGF- 880 C-dependent lymphangiogenesis by inhibiting miR-300 in human oral squamous cell 881 carcinoma cells. Oncotarget 2016;7:9993-10005 882 133. Shao H, Cai L, Moller M, Issac B, Zhang L, Owyong M, et al. Notch1-WISP-1 axis 883 determines the regulatory role of mesenchymal stem cell-derived stromal fibroblasts in 884 melanoma metastasis. Oncotarget 2016;7:79262-73 885 134. Soon LL, Yie TA, Shvarts A, Levine AJ, Su F, Tchou-Wong KM. Overexpression of WISP-1 886 down-regulated motility and invasion of lung cancer cells through inhibition of Rac 887 activation. J Biol Chem 2003;278:11465-70 888 135. Orend G, Chiquet-Ehrismann R. Tenascin-C induced signaling in cancer. Cancer Lett 889 2006;244:143-63 890 136. Fiorilli P, Partridge D, Staniszewska I, Wang JY, Grabacka M, So K, et al. Integrins mediate 891 adhesion of medulloblastoma cells to tenascin and activate pathways associated with 892 survival and proliferation. Lab Invest 2008;88:1143-56 893 137. Yokosaki Y, Monis H, Chen J, Sheppard D. Differential effects of the integrins 894 alpha9beta1, alphavbeta3, and alphavbeta6 on cell proliferative responses to tenascin. 895 Roles of the beta subunit extracellular and cytoplasmic domains. J Biol Chem 896 1996;271:24144-50 897 138. San Martin R, Pathak R, Jain A, Jung SY, Hilsenbeck SG, Pina-Barba MC, et al. Tenascin-C 898 and Integrin alpha9 Mediate Interactions of Prostate Cancer with the Bone 899 Microenvironment. Cancer Res 2017;77:5977-88 900 139. Huang W, Chiquet-Ehrismann R, Moyano JV, Garcia-Pardo A, Orend G. Interference of 901 tenascin-C with syndecan-4 binding to fibronectin blocks cell adhesion and stimulates 902 tumor cell proliferation. Cancer Res 2001;61:8586-94 903 140. Alcaraz LB, Exposito JY, Chuvin N, Pommier RM, Cluzel C, Martel S, et al. Tenascin-X 904 promotes epithelial-to-mesenchymal transition by activating latent TGF-beta. The 905 Journal of cell biology 2014;205:409-28 906 141. Chuanyu S, Yuqing Z, Chong X, Guowei X, Xiaojun Z. Periostin promotes migration and 907 invasion of renal cell carcinoma through the integrin/focal adhesion kinase/c-Jun N- 908 terminal kinase pathway. Tumour Biol 2017;39:1010428317694549 909 142. Orecchia P, Conte R, Balza E, Castellani P, Borsi L, Zardi L, et al. Identification of a novel 910 cell binding site of periostin involved in tumour growth. Eur J Cancer 2011;47:2221-9 911 143. Bao S, Ouyang G, Bai X, Huang Z, Ma C, Liu M, et al. Periostin potently promotes 912 metastatic growth of colon cancer by augmenting cell survival via the Akt/PKB pathway. 913 Cancer Cell 2004;5:329-39

34

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

914 144. Michaylira CZ, Wong GS, Miller CG, Gutierrez CM, Nakagawa H, Hammond R, et al. 915 Periostin, a cell adhesion molecule, facilitates invasion in the tumor microenvironment 916 and annotates a novel tumor-invasive signature in esophageal cancer. Cancer Res 917 2010;70:5281-92 918 145. Yao Y, Zebboudj AF, Shao E, Perez M, Bostrom K. Regulation of bone morphogenetic 919 protein-4 by matrix GLA protein in vascular endothelial cells involves activin-like kinase 920 receptor 1. The Journal of biological chemistry 2006;281:33921-30 921 146. Nam JS, Suchar AM, Kang MJ, Stuelten CH, Tang B, Michalowska AM, et al. Bone 922 sialoprotein mediates the tumor cell-targeted prometastatic activity of transforming 923 growth factor beta in a mouse model of breast cancer. Cancer Res 2006;66:6327-35 924 147. Sung V, Stubbs JT, 3rd, Fisher L, Aaron AD, Thompson EW. Bone sialoprotein supports 925 breast cancer cell adhesion proliferation and migration through differential usage of the 926 alpha(v)beta3 and alpha(v)beta5 integrins. J Cell Physiol 1998;176:482-94 927 148. Zhao H, Chen Q, Alam A, Cui J, Suen KC, Soo AP, et al. The role of osteopontin in the 928 progression of solid organ tumour. Cell Death Dis 2018;9:356 929 149. Gimba ERP, Brum MCM, Nestal De Moraes G. Full-length osteopontin and its splice 930 variants as modulators of chemoresistance and radioresistance (Review). Int J Oncol 931 2019;54:420-30 932 150. Shirasaki T, Honda M, Yamashita T, Nio K, Shimakami T, Shimizu R, et al. The 933 osteopontin-CD44 axis in hepatic cancer stem cells regulates IFN signaling and HCV 934 replication. Sci Rep 2018;8:13143 935 151. Pietras A, Katz AM, Ekstrom EJ, Wee B, Halliday JJ, Pitter KL, et al. Osteopontin-CD44 936 signaling in the glioma perivascular niche enhances cancer stem cell phenotypes and 937 promotes aggressive tumor growth. Cell Stem Cell 2014;14:357-69 938 152. Rao G, Wang H, Li B, Huang L, Xue D, Wang X, et al. Reciprocal interactions between 939 tumor-associated and CD44-positive cancer cells via osteopontin/CD44 940 promote tumorigenicity in colorectal cancer. Clin Cancer Res 2013;19:785-97 941 153. Tajima K, Ohashi R, Sekido Y, Hida T, Nara T, Hashimoto M, et al. Osteopontin-mediated 942 enhanced hyaluronan binding induces multidrug resistance in mesothelioma cells. 943 Oncogene 2010;29:1941-51 944 154. Pickup MW, Mouw JK, Weaver VM. The extracellular matrix modulates the hallmarks of 945 cancer. EMBO Rep 2014;15:1243-53 946 155. Fiorino S, Di Saverio S, Leandri P, Tura A, Birtolo C, Silingardi M, et al. The role of 947 matricellular proteins and tissue stiffness in breast cancer: a systematic review. Future 948 Oncol 2018;14:1601-27 949 156. Sangaletti S, Colombo MP. Matricellular proteins at the crossroad of inflammation and 950 cancer. Cancer Lett 2008;267:245-53 951 157. Zhu X, Zhong J, Zhao Z, Sheng J, Wang J, Liu J, et al. Epithelial derived CTGF promotes 952 breast tumor progression via inducing EMT and collagen I fibers deposition. Oncotarget 953 2015;6:25320-38 954 158. Legate KR, Wickstrom SA, Fassler R. Genetic and cell biological analysis of integrin 955 outside-in signaling. Genes Dev 2009;23:397-418 956 159. Lee S, Kumar S. Actomyosin stress fiber mechanosensing in 2D and 3D. F1000Res 2016;5

35

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

957 160. Bradshaw AD. The role of SPARC in extracellular matrix assembly. J Cell Commun Signal 958 2009;3:239-46 959 161. Martinek N, Shahab J, Sodek J, Ringuette M. Is SPARC an evolutionarily conserved 960 collagen chaperone? Journal of dental research 2007;86:296-305 961 162. Brekken RA, Puolakkainen P, Graves DC, Workman G, Lubkin SR, Sage EH. Enhanced 962 growth of tumors in SPARC null mice is associated with changes in the ECM. J Clin Invest 963 2003;111:487-95 964 163. Bradshaw AD, Baicu CF, Rentz TJ, Van Laer AO, Boggs J, Lacy JM, et al. Pressure 965 overload-induced alterations in fibrillar collagen content and myocardial diastolic 966 function: role of secreted protein acidic and rich in cysteine (SPARC) in post-synthetic 967 procollagen processing. Circulation 2009;119:269-80 968 164. Chioran A, Duncan S, Catalano A, Brown TJ, Ringuette MJ. Collagen IV trafficking: The 969 inside-out and beyond story. Dev Biol 2017;431:124-33 970 165. Chlenski A, Cohn SL. Modulation of matrix remodeling by SPARC in neoplastic 971 progression. Semin Cell Dev Biol 2010;21:55-65 972 166. Kehlet SN, Manon-Jensen T, Sun S, Brix S, Leeming DJ, Karsdal MA, et al. A fragment of 973 SPARC reflecting increased collagen affinity shows pathological relevance in lung cancer 974 - implications of a new collagen chaperone function of SPARC. Cancer Biol Ther 975 2018;19:904-12 976 167. Tomko LA, Hill RC, Barrett A, Szulczewski JM, Conklin MW, Eliceiri KW, et al. Targeted 977 matrisome analysis identifies thrombospondin-2 and tenascin-C in aligned collagen 978 stroma from invasive breast carcinoma. Sci Rep 2018;8:12941 979 168. Provenzano PP, Eliceiri KW, Campbell JM, Inman DR, White JG, Keely PJ. Collagen 980 reorganization at the tumor-stromal interface facilitates local invasion. BMC Med 981 2006;4:38 982 169. Kubow KE, Vukmirovic R, Zhe L, Klotzsch E, Smith ML, Gourdon D, et al. Mechanical 983 forces regulate the interactions of fibronectin and collagen I in extracellular matrix. Nat 984 Commun 2015;6:8026 985 170. Kadler KE, Hill A, Canty-Laird EG. Collagen fibrillogenesis: fibronectin, integrins, and 986 minor collagens as organizers and nucleators. Curr Opin Cell Biol 2008;20:495-501 987 171. Sottile J, Shi F, Rublyevska I, Chiang HY, Lust J, Chandler J. Fibronectin-dependent 988 collagen I deposition modulates the cell response to fibronectin. Am J Physiol Cell 989 Physiol 2007;293:C1934-46 990 172. Maruhashi T, Kii I, Saito M, Kudo A. Interaction between periostin and BMP-1 promotes 991 proteolytic activation of . J Biol Chem 2010;285:13294-303 992 173. Garnero P. The contribution of collagen crosslinks to bone strength. Bonekey Rep 993 2012;1:182 994 174. Gineyts E, Bonnet N, Ferrari S, Garnero P. PERIOSTIN, A MATRICELLULAR GLUTAMIC- 995 ACID PROTEIN INFLUENCES CROSSLINKING OF BONE COLLAGEN. 2011. 43- p. 996 175. Ibbetson SJ, Pyne NT, Pollard AN, Olson MF, Samuel MS. Mechanotransduction 997 pathways promoting tumor progression are activated in invasive human squamous cell 998 carcinoma. Am J Pathol 2013;183:930-7

36

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

999 176. Jia H, Janjanam J, Wu SC, Wang R, Pano G, Celestine M, et al. The tumor cell-secreted 1000 matricellular protein WISP1 drives pro-metastatic collagen linearization. EMBO J 1001 2019;38:e101302 1002 177. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in 1003 development and disease. Cell 2009;139:871-90 1004 178. Walker C, Mojares E, Del Rio Hernandez A. Role of Extracellular Matrix in Development 1005 and Cancer Progression. Int J Mol Sci 2018;19 1006 179. Lu W, Kang Y. Epithelial-Mesenchymal Plasticity in Cancer Progression and Metastasis. 1007 Dev Cell 2019;49:361-74 1008 180. Brady JJ, Chuang CH, Greenside PG, Rogers ZN, Murray CW, Caswell DR, et al. An Arntl2- 1009 Driven Secretome Enables Lung Adenocarcinoma Metastatic Self-Sufficiency. Cancer Cell 1010 2016;29:697-710 1011 181. Kim HP, Han SW, Song SH, Jeong EG, Lee MY, Hwang D, et al. Testican-1-mediated 1012 epithelial-mesenchymal transition signaling confers acquired resistance to lapatinib in 1013 HER2-positive gastric cancer. Oncogene 2014;33:3334-41 1014 182. Chang CH, Yen MC, Liao SH, Hsu YL, Lai CS, Chang KP, et al. Secreted Protein Acidic and 1015 Rich in Cysteine (SPARC) Enhances Cell Proliferation, Migration, and Epithelial 1016 Mesenchymal Transition, and SPARC Expression is Associated with Tumor Grade in Head 1017 and Neck Cancer. Int J Mol Sci 2017;18 1018 183. Sangaletti S, Di Carlo E, Gariboldi S, Miotti S, Cappetti B, Parenza M, et al. - 1019 derived SPARC bridges tumor cell-extracellular matrix interactions toward metastasis. 1020 Cancer Res 2008;68:9050-9 1021 184. Robert G, Gaggioli C, Bailet O, Chavey C, Abbe P, Aberdam E, et al. SPARC represses E- 1022 cadherin and induces mesenchymal transition during melanoma development. Cancer 1023 Res 2006;66:7516-23 1024 185. Hou CH, Lin FL, Hou SM, Liu JF. Cyr61 promotes epithelial-mesenchymal transition and 1025 tumor metastasis of osteosarcoma by Raf-1/MEK/ERK/Elk-1/TWIST-1 signaling pathway. 1026 Mol Cancer 2014;13:236 1027 186. Habel N, Stefanovska B, Carene D, Patino-Garcia A, Lecanda F, Fromigue O. CYR61 1028 triggers osteosarcoma metastatic spreading via an IGF1Rbeta-dependent EMT-like 1029 process. BMC Cancer 2019;19:62 1030 187. Das A, Dhar K, Maity G, Sarkar S, Ghosh A, Haque I, et al. Deficiency of CCN5/WISP-2- 1031 Driven Program in breast cancer Promotes Cancer Epithelial cells to mesenchymal stem 1032 cells and Breast Cancer growth. Sci Rep 2017;7:1220 1033 188. Haque I, Ghosh A, Acup S, Banerjee S, Dhar K, Ray A, et al. Leptin-induced ER-alpha- 1034 positive breast cancer cell viability and migration is mediated by suppressing CCN5- 1035 signaling via activating JAK/AKT/STAT-pathway. BMC Cancer 2018;18:99 1036 189. Huang W, Martin EE, Burman B, Gonzalez ME, Kleer CG. The matricellular protein CCN6 1037 (WISP3) decreases Notch1 and suppresses breast cancer initiating cells. Oncotarget 1038 2016;7:25180-93 1039 190. Tran MN, Kleer CG. Matricellular CCN6 (WISP3) protein: a tumor suppressor for 1040 mammary metaplastic carcinomas. J Cell Commun Signal 2018;12:13-9

37

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1041 191. Qin X, Yan M, Zhang J, Wang X, Shen Z, Lv Z, et al. TGFbeta3-mediated induction of 1042 Periostin facilitates head and neck cancer growth and is associated with metastasis. Sci 1043 Rep 2016;6:20587 1044 192. Wu G, Wang X, Zhang X. Clinical implications of periostin in the liver metastasis of 1045 colorectal cancer. Cancer Biother Radiopharm 2013;28:298-302 1046 193. Kudo Y, Ogawa I, Kitajima S, Kitagawa M, Kawai H, Gaffney PM, et al. Periostin promotes 1047 invasion and anchorage-independent growth in the metastatic process of head and neck 1048 cancer. Cancer Res 2006;66:6928-35 1049 194. Insua-Rodriguez J, Pein M, Hongu T, Meier J, Descot A, Lowy CM, et al. Stress signaling in 1050 breast cancer cells induces matrix components that promote chemoresistant 1051 metastasis. EMBO Mol Med 2018;10 1052 195. Murakami T, Kikuchi H, Ishimatsu H, Iino I, Hirotsu A, Matsumoto T, et al. in 1053 colorectal cancer stroma is a predictive marker for liver metastasis and is a potent target 1054 of miR-198 as identified by microRNA analysis. Br J Cancer 2017;117:1360-70 1055 196. Yang Z, Zhang C, Qi W, Cui C, Cui Y, Xuan Y. Tenascin-C as a prognostic determinant of 1056 colorectal cancer through induction of epithelial-to-mesenchymal transition and 1057 proliferation. Exp Mol Pathol 2018;105:216-22 1058 197. Yoneura N, Takano S, Yoshitomi H, Nakata Y, Shimazaki R, Kagawa S, et al. Expression of 1059 II and stromal tenascin C promotes epithelial to mesenchymal transition and 1060 correlates with distant metastasis in pancreatic cancer. Int J Mol Med 2018;42:821-30 1061 198. Chen L, Tian X, Gong W, Sun B, Li G, Liu D, et al. Periostin mediates epithelial- 1062 mesenchymal transition through the MAPK/ERK pathway in hepatoblastoma. Cancer 1063 Biol Med 2019;16:89-100 1064 199. Hu WW, Chen PC, Chen JM, Wu YM, Liu PY, Lu CH, et al. Periostin promotes epithelial- 1065 mesenchymal transition via the MAPK/miR-381 axis in lung cancer. Oncotarget 1066 2017;8:62248-60 1067 200. Kaariainen E, Nummela P, Soikkeli J, Yin M, Lukk M, Jahkola T, et al. Switch to an invasive 1068 growth phase in melanoma is associated with tenascin-C, fibronectin, and procollagen-I 1069 forming specific channel structures for invasion. J Pathol 2006;210:181-91 1070 201. Sun Z, Velazquez-Quesada I, Murdamoothoo D, Ahowesso C, Yilmaz A, Spenle C, et al. 1071 Tenascin-C increases lung metastasis by impacting invasions. Matrix Biol 1072 2019 1073 202. Degen M, Brellier F, Schenk S, Driscoll R, Zaman K, Stupp R, et al. Tenascin-W, a new 1074 marker of cancer stroma, is elevated in sera of colon and breast cancer patients. Int J 1075 Cancer 2008;122:2454-61 1076 203. Shoji T, Kamiya T, Tsubura A, Hatano T, Sakakura T, Yamamoto M, et al. 1077 Immunohistochemical staining patterns of tenascin in invasive breast carcinomas. 1078 Virchows Arch A Pathol Anat Histopathol 1992;421:53-6 1079 204. Spenle C, Gasser I, Saupe F, Janssen KP, Arnold C, Klein A, et al. Spatial organization of 1080 the tenascin-C microenvironment in experimental and human cancer. Cell Adh Migr 1081 2015;9:4-13 1082 205. Saupe F, Schwenzer A, Jia Y, Gasser I, Spenle C, Langlois B, et al. Tenascin-C 1083 downregulates wnt inhibitor dickkopf-1, promoting tumorigenesis in a neuroendocrine 1084 tumor model. Cell Rep 2013;5:482-92

38

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1085 206. Midwood KS, Orend G. The role of tenascin-C in tissue injury and tumorigenesis. J Cell 1086 Commun Signal 2009;3:287-310 1087 207. Gaggioli C, Hooper S, Hidalgo-Carcedo C, Grosse R, Marshall JF, Harrington K, et al. 1088 Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases 1089 in leading and following cells. Nat Cell Biol 2007;9:1392-400 1090 208. Kii I, Nishiyama T, Li M, Matsumoto K, Saito M, Amizuka N, et al. Incorporation of 1091 tenascin-C into the extracellular matrix by periostin underlies an extracellular meshwork 1092 architecture. J Biol Chem 2010;285:2028-39 1093 209. Sun Z, Schwenzer A, Rupp T, Murdamoothoo D, Vegliante R, Lefebvre O, et al. Tenascin- 1094 C Promotes Tumor Cell Migration and Metastasis through Integrin alpha9beta1- 1095 Mediated YAP Inhibition. Cancer Res 2018;78:950-61 1096 210. Kii I. Periostin Functions as a Scaffold for Assembly of Extracellular Proteins. Adv Exp 1097 Med Biol 2019;1132:23-32 1098 211. Karadag A, Ogbureke KU, Fedarko NS, Fisher LW. Bone sialoprotein, matrix 1099 metalloproteinase 2, and alpha(v)beta3 integrin in osteotropic cancer cell invasion. J 1100 Natl Cancer Inst 2004;96:956-65 1101 212. McClung HM, Thomas SL, Osenkowski P, Toth M, Menon P, Raz A, et al. SPARC 1102 upregulates MT1-MMP expression, MMP-2 activation, and the secretion and cleavage of 1103 galectin-3 in U87MG glioma cells. Neurosci Lett 2007;419:172-7 1104 213. Gilles C, Bassuk JA, Pulyaeva H, Sage EH, Foidart JM, Thompson EW. SPARC/osteonectin 1105 induces matrix metalloproteinase 2 activation in human breast cancer cell lines. Cancer 1106 Res 1998;58:5529-36 1107 214. Liu G, Ren F, Song Y. Upregulation of SPOCK2 inhibits the invasion and migration of 1108 prostate cancer cells by regulating the MT1-MMP/MMP2 pathway. PeerJ 2019;7:e7163 1109 215. Ren F, Wang D, Wang Y, Chen P, Guo C. SPOCK2 Affects the Biological Behavior of 1110 Endometrial Cancer Cells by Regulation of MT1-MMP and MMP2. Reprod Sci 1111 2019:1933719119834341 1112 216. Suzuki H, Sasada M, Kamiya S, Ito Y, Watanabe H, Okada Y, et al. The Promoting Effect of 1113 the Extracellular Matrix Peptide TNIIIA2 Derived from Tenascin-C in Colon Cancer Cell 1114 Infiltration. Int J Mol Sci 2017;18 1115 217. Sarkar S, Nuttall RK, Liu S, Edwards DR, Yong VW. Tenascin-C stimulates glioma cell 1116 invasion through matrix metalloproteinase-12. Cancer Res 2006;66:11771-80 1117 218. Khalil RA. Matrix Metalloproteinases and Tissue Remodeling in Health and Disease: 1118 Target Tissues and Therapy. Academic Press; 2017. 1119 219. Weaver M, Workman G, Schultz CR, Lemke N, Rempel SA, Sage EH. Proteolysis of the 1120 matricellular protein hevin by matrix metalloproteinase-3 produces a SPARC-like 1121 fragment (SLF) associated with neovasculature in a murine glioma model. J Cell Biochem 1122 2011;112:3093-102 1123 220. Podgorski I, Linebaugh BE, Koblinski JE, Rudy DL, Herroon MK, Olive MB, et al. Bone 1124 marrow-derived cathepsin K cleaves SPARC in bone metastasis. Am J Pathol 1125 2009;175:1255-69 1126 221. Christensen B, Klaning E, Nielsen MS, Andersen MH, Sorensen ES. C-terminal 1127 modification of osteopontin inhibits interaction with the alphaVbeta3-integrin. J Biol 1128 Chem 2012;287:3788-97

39

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1129 222. Mi Z, Oliver T, Guo H, Gao C, Kuo PC. Thrombin-cleaved COOH(-) terminal osteopontin 1130 peptide binds with cyclophilin C to CD147 in murine breast cancer cells. Cancer Res 1131 2007;67:4088-97 1132 223. Takafuji V, Forgues M, Unsworth E, Goldsmith P, Wang XW. An osteopontin fragment is 1133 essential for tumor cell invasion in hepatocellular carcinoma. Oncogene 2007;26:6361- 1134 71 1135 224. Hawkins AG, Julian CM, Konzen S, Treichel S, Lawlor ER, Bailey KM. Microenvironmental 1136 Factors Drive Tenascin C and Src Cooperation to Promote Invadopodia Formation in 1137 Ewing Sarcoma. Neoplasia 2019;21:1063-72 1138 225. O'Connell JT, Sugimoto H, Cooke VG, MacDonald BA, Mehta AI, LeBleu VS, et al. VEGF-A 1139 and Tenascin-C produced by S100A4+ stromal cells are important for metastatic 1140 colonization. Proc Natl Acad Sci U S A 2011;108:16002-7 1141 226. Oskarsson T, Acharyya S, Zhang XH, Vanharanta S, Tavazoie SF, Morris PG, et al. Breast 1142 cancer cells produce tenascin C as a metastatic niche component to colonize the lungs. 1143 Nature medicine 2011;17:867-74 1144 227. Fukunaga-Kalabis M, Martinez G, Nguyen TK, Kim D, Santiago-Walker A, Roesch A, et al. 1145 Tenascin-C promotes melanoma progression by maintaining the ABCB5-positive side 1146 population. Oncogene 2010;29:6115-24 1147 228. Lambert AW, Wong CK, Ozturk S, Papageorgis P, Raghunathan R, Alekseyev Y, et al. 1148 Tumor Cell-Derived Periostin Regulates Cytokines That Maintain Breast Cancer Stem 1149 Cells. Mol Cancer Res 2016;14:103-13 1150 229. Semba T, Sugihara E, Kamoshita N, Ueno S, Fukuda K, Yoshino M, et al. Periostin 1151 antisense oligonucleotide suppresses bleomycin-induced formation of a lung 1152 premetastatic niche for melanoma. Cancer Sci 2018;109:1447-54 1153 230. Wang Z, Xiong S, Mao Y, Chen M, Ma X, Zhou X, et al. Periostin promotes 1154 immunosuppressive premetastatic niche formation to facilitate breast tumour 1155 metastasis. J Pathol 2016;239:484-95 1156 231. Malanchi I, Santamaria-Martinez A, Susanto E, Peng H, Lehr HA, Delaloye JF, et al. 1157 Interactions between cancer stem cells and their niche govern metastatic colonization. 1158 Nature 2011;481:85-9 1159 232. Chen Y, Zhang Y, Tan Y, Liu Z. Clinical significance of SPARC in esophageal squamous cell 1160 carcinoma. Biochem Biophys Res Commun 2017;492:184-91 1161 233. Davis EJ, Zhao L, Lucas DR, Schuetze SM, Baker LH, Zalupski MM, et al. SPARC expression 1162 in patients with high-risk localized sarcoma treated on a randomized phase II 1163 trial of neo/adjuvant chemotherapy. BMC Cancer 2016;16:663 1164 234. Rich JN, Hans C, Jones B, Iversen ES, McLendon RE, Rasheed BK, et al. Gene expression 1165 profiling and genetic markers in glioblastoma survival. Cancer Res 2005;65:4051-8 1166 235. Cao DX, Li ZJ, Jiang XO, Lum YL, Khin E, Lee NP, et al. Osteopontin as potential biomarker 1167 and therapeutic target in gastric and liver cancers. World J Gastroenterol 2012;18:3923- 1168 30 1169 236. Feng YH, Su YC, Lin SF, Lin PR, Wu CL, Tung CL, et al. Oct4 upregulates osteopontin via 1170 Egr1 and is associated with poor outcome in human lung cancer. BMC Cancer 1171 2019;19:791

40

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1172 237. Wei R, Wong JPC, Lyu P, Xi X, Tong O, Zhang SD, et al. In vitro and clinical data analysis of 1173 Osteopontin as a prognostic indicator in colorectal cancer. J Cell Mol Med 2018;22:4097- 1174 105 1175 238. Bramwell VH, Doig GS, Tuck AB, Wilson SM, Tonkin KS, Tomiak A, et al. Serial plasma 1176 osteopontin levels have prognostic value in . Clin Cancer Res 1177 2006;12:3337-43 1178 239. Capper D, Mittelbronn M, Goeppert B, Meyermann R, Schittenhelm J. Secreted protein, 1179 acidic and rich in cysteine (SPARC) expression in astrocytic tumour cells negatively 1180 correlates with proliferation, while vascular SPARC expression is associated with patient 1181 survival. Neuropathol Appl Neurobiol 2010;36:183-97 1182 240. Rittling SR, Chambers AF. Role of osteopontin in tumour progression. Br J Cancer 1183 2004;90:1877-81 1184 241. Yang JY, Yang MW, Huo YM, Liu W, Liu DJ, Li J, et al. High expression of WISP-1 1185 correlates with poor prognosis in pancreatic ductal adenocarcinoma. Am J Transl Res 1186 2015;7:1621-8 1187 242. Xiao G, Tang Z, Yuan X, Yuan J, Zhao J, Zhang Z, et al. The expression of Wnt-1 inducible 1188 signaling pathway protein-2 in astrocytoma: Correlation between pathological grade 1189 and clinical outcome. Oncol Lett 2015;9:235-40 1190 243. Xie JJ, Xu LY, Wu ZY, Li LY, Xu XE, Wu JY, et al. Expression of cysteine-rich 61 is correlated 1191 with poor prognosis in patients with esophageal squamous cell carcinoma. Eur J Surg 1192 Oncol 2011;37:669-74 1193 244. Kok SH, Chang HH, Tsai JY, Hung HC, Lin CY, Chiang CP, et al. Expression of Cyr61 (CCN1) 1194 in human oral squamous cell carcinoma: An independent marker for poor prognosis. 1195 Head Neck 2010;32:1665-73 1196 245. Davies SR, Watkins G, Mansel RE, Jiang WG. Differential expression and prognostic 1197 implications of the CCN family members WISP-1, WISP-2, and WISP-3 in human breast 1198 cancer. Ann Surg Oncol 2007;14:1909-18 1199 246. Qi J, Esfahani DR, Huang T, Ozark P, Bartom E, Hashizume R, et al. Tenascin-C expression 1200 contributes to pediatric brainstem glioma tumor phenotype and represents a novel 1201 biomarker of disease. Acta Neuropathol Commun 2019;7:75 1202 247. Guan Z, Zeng J, Wang Z, Xie H, Lv C, Ma Z, et al. Urine tenascinC is an independent risk 1203 factor for bladder cancer patients. Mol Med Rep 2014;9:961-6 1204 248. Balasenthil S, Chen N, Lott ST, Chen J, Carter J, Grizzle WE, et al. A migration signature 1205 and plasma biomarker panel for pancreatic adenocarcinoma. Cancer Prev Res (Phila) 1206 2011;4:137-49 1207 249. Yang T, Deng Z, Pan Z, Qian Y, Yao W, Wang J. Prognostic value of periostin in multiple 1208 solid cancers: A systematic review with meta-analysis. J Cell Physiol 2019 1209 250. Shojaei F, Scott N, Kang X, Lappin PB, Fitzgerald AA, Karlicek S, et al. Osteopontin 1210 induces growth of metastatic tumors in a preclinical model of non-small lung cancer. J 1211 Exp Clin Cancer Res 2012;31:26 1212 251. Ohara Y, Chew SH, Misawa N, Wang S, Somiya D, Nakamura K, et al. Connective tissue 1213 growth factor-specific monoclonal antibody inhibits growth of malignant mesothelioma 1214 in an orthotopic mouse model. Oncotarget 2018;9:18494-509

41

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1215 252. Moran-Jones K, Gloss BS, Murali R, Chang DK, Colvin EK, Jones MD, et al. Connective 1216 tissue growth factor as a novel therapeutic target in high grade serous ovarian cancer. 1217 Oncotarget 2015;6:44551-62 1218 253. Neesse A, Frese KK, Bapiro TE, Nakagawa T, Sternlicht MD, Seeley TW, et al. CTGF 1219 antagonism with mAb FG-3019 enhances chemotherapy response without increasing 1220 drug delivery in murine ductal pancreas cancer. Proc Natl Acad Sci U S A 1221 2013;110:12325-30 1222 254. Makino Y, Hikita H, Kodama T, Shigekawa M, Yamada R, Sakamori R, et al. CTGF 1223 Mediates Tumor-Stroma Interactions between Hepatoma Cells and Hepatic Stellate Cells 1224 to Accelerate HCC Progression. Cancer Res 2018;78:4902-14 1225 255. Dornhofer N, Spong S, Bennewith K, Salim A, Klaus S, Kambham N, et al. Connective 1226 tissue growth factor-specific monoclonal antibody therapy inhibits pancreatic tumor 1227 growth and metastasis. Cancer Res 2006;66:5816-27 1228 256. Aikawa T, Gunn J, Spong SM, Klaus SJ, Korc M. Connective tissue growth factor-specific 1229 antibody attenuates tumor growth, metastasis, and angiogenesis in an orthotopic 1230 mouse model of pancreatic cancer. Mol Cancer Ther 2006;5:1108-16 1231 257. Zukiel R, Nowak S, Wyszko E, Rolle K, Gawronska I, Barciszewska MZ, et al. Suppression 1232 of human brain tumor with interference RNA specific for tenascin-C. Cancer Biol Ther 1233 2006;5:1002-7 1234 258. Wyszko E, Rolle K, Nowak S, Zukiel R, Nowak M, Piestrzeniewicz R, et al. A multivariate 1235 analysis of patients with brain tumors treated with ATN-RNA. Acta Pol Pharm 1236 2008;65:677-84 1237 259. Zalutsky MR, Reardon DA, Akabani G, Coleman RE, Friedman AH, Friedman HS, et al. 1238 Clinical experience with alpha-particle emitting 211At: treatment of recurrent brain 1239 tumor patients with 211At-labeled chimeric antitenascin monoclonal antibody 81C6. J 1240 Nucl Med 2008;49:30-8 1241 260. Rizzieri DA, Akabani G, Zalutsky MR, Coleman RE, Metzler SD, Bowsher JE, et al. Phase 1 1242 trial study of 131I-labeled chimeric 81C6 monoclonal antibody for the treatment of 1243 patients with non-Hodgkin lymphoma. Blood 2004;104:642-8 1244 261. Kuschel C, Steuer H, Maurer AN, Kanzok B, Stoop R, Angres B. Cell adhesion profiling 1245 using extracellular matrix protein microarrays. Biotechniques 2006;40:523-31 1246 262. Kleinman HK, Martin GR. Matrigel: basement membrane matrix with biological activity. 1247 Semin Cancer Biol 2005;15:378-86 1248 263. Rosso F, Marino G, Giordano A, Barbarisi M, Parmeggiani D, Barbarisi A. Smart materials 1249 as scaffolds for tissue engineering. J Cell Physiol 2005;203:465-70 1250 264. Madhusoodanan J. Matrix mimics shape cell studies. Nature 2019;566:563-5 1251 265. Conboy CB, Velez-Reyes GL, Tschida BR, Hu H, Kaufmann G, Koes N, et al. R-spondin 2 1252 Drives Liver Tumor Development in a Yes-Associated Protein-Dependent Manner. 1253 Hepatol Commun 2019;3:1496-509 1254 266. Liu Q, Zhao Y, Xing H, Li L, Li R, Dai J, et al. The role of R-spondin 1 through activating 1255 Wnt/beta-catenin in the growth, survival and migration of ovarian cancer cells. Gene 1256 2019;689:124-30 1257 267. Liu S, U KP, Zhang J, Tsang LL, Huang J, Tu SP, et al. R-spodin2 enhances canonical Wnt 1258 signaling to maintain the stemness of glioblastoma cells. Cancer Cell Int 2018;18:156

42

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1259 268. Paulitti A, Andreuzzi E, Bizzotto D, Pellicani R, Tarticchio G, Marastoni S, et al. The 1260 ablation of the matricellular protein EMILIN2 causes defective vascularization due to 1261 impaired EGFR-dependent IL-8 production affecting tumor growth. Oncogene 1262 2018;37:3399-414 1263 269. Capuano A, Pivetta E, Sartori G, Bosisio G, Favero A, Cover E, et al. Abrogation of 1264 EMILIN1-beta1 integrin interaction promotes experimental colitis and colon 1265 carcinogenesis. Matrix Biol 2019;83:97-115 1266 270. Danussi C, Petrucco A, Wassermann B, Modica TM, Pivetta E, Del Bel Belluz L, et al. An 1267 EMILIN1-negative microenvironment promotes tumor cell proliferation and lymph node 1268 invasion. Cancer Prev Res (Phila) 2012;5:1131-43 1269

1270

43

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1271 Figure 1. Activation of the biochemical, biomechanical and metastatic effects by Matricellular

1272 Proteins. Tumor cells and the surrounding activated stromal cells are the major cell types that

1273 abnormally secrete MCPs into the microenvironment to affect cellular behavior and

1274 extracellular matrix (ECM) remodeling. A) Biochemical pathways - Matricellular Proteins (MCPs)

1275 can activate an array of cell surface receptors. Most MCPs can bind and signal through

1276 integrins, with a specific heterodimer signature accounting for signaling diversity (See text for

1277 details). In addition, it has been shown that CCN and tenascin-C (TN-C) can bind and signal

1278 through syndecans, while osteopontin (SPP1) mediates its effects through CD44. B)

1279 Biomechanical pathways – MCPs are able to increase the stiffness of the normal ECM tension

1280 by influencing matrix organization and collagen crosslinking, as well as deregulating enzymatic

1281 activity. Stiffness is converted by integrins into biochemical signals that can influence pathways

1282 in A). In addition, matrix stiffness can lead to the maturation of integrin and the actin

1283 cytoskeleton into focal adhesions and stress fibers, respectively. This occurs by activating the

1284 integrin-RhoA-ROCK-myosin axis, which is reviewed in detail elsewhere(158),(159). C)

1285 Metastatic niche – Various MCPs prepare cancer cells and the local and secondary tumor sites

1286 for metastasis through numerous steps. MCPs stimulate cancer cells into a motile phenotype

1287 through the epithelial-to-mesenchymal transformation (EMT) but also to promote invadopodia

1288 formation at the invasion site. For metastatic cells to exit the embedded state for intravasation,

1289 MCPs can break down the ECM basement membrane through MMPs and guide cells out of

1290 their embedded state by crosslinking collagens into migration tracks. At the secondary site,

1291 MCPs once again activate MMPs to remodel the ECM for colonization after invasion. At the

1292 distant site, MCPs also prime the ECM for colonization in order to accommodate disseminated

44

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1293 tumor cells in the new environment. Figure was produced using Servier Medical Art

1294 (http://smart.servier.com/).

45

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

A) BIOCHEMICAL B) BIOMECHANICAL SMOC2 ECM CCN1 SPARC BSP BSP MGP CCN2 CCN6 MGP INCREASE IN ECM STIFFNESS ECM SPARC TN-X SPARC POSTN Increased matrix organization CCN4 TN-C Collagen crosslinking SPP1 Deregulation of Enzymes (i.e. LOX)

TN-C POSTN TGFβ1 BMPs CCN3 TN-C SPP1 CCN TGFβ1 Receptor ECM Syndecan-4 Integrin Family CD44 Matrix β α Assembly β α β α & Signaling Paxillin ILK FAK Focal FAK Src ILK ILK P FAK FAK ILK ILK Adhesion P FAK FAK Force Maturation Filamentous Recruited RhoA Crk Ras Crk ILK Actin Fibers RhoA Proteins

ROCK Rac1 ERK Jnk Akt ROCK P Myosin Contraction Force NFκB HIF-2α P P Myosin MMP2 AP1 P

MIGRATION INVASION SURVIVAL and PROLIFERATION STEMNESS P Formation MMP9 Myosin Actin of Stress bundling Fibers

SPARC Migration Tracks MGP C) METASTATIC NICHE POSTN Collagen POSTN Crosslinking TN-C MGP TN-C POSTN MGP SPARC TN-C Colonization

MMP MMP POSTN MGP TN-C

SPARC POSTN POSTN SPARC TN-C MGP Epithelial-to- Enzymatic MMP MMP Invasion BSP Activity MMP MMP Mesenchymal TN-C SPP1 TN-C Transition CCN1

SMOC2 Intravasation SPOCK1 Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

The Matrix Revolution: Matricellular Proteins and Restructuring of the Cancer Microenvironment

Casimiro Gerarduzzi, Ursula Hartmann, Andrew Leask, et al.

Cancer Res Published OnlineFirst March 19, 2020.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-18-2098

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet been Manuscript edited.

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

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/early/2020/03/19/0008-5472.CAN-18-2098. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2020 American Association for Cancer Research.