tissue factor proangiogenic signaling in cancer progression

Upload: iaiadodi

Post on 04-Jun-2018

217 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/13/2019 Tissue Factor Proangiogenic Signaling in Cancer Progression

    1/5

    Thrombosis Research 129, Supplement 1 (2012) S127S131

    PL-27

    Tissue factor proangiogenic signaling in cancer progression

    Florence Schaffner, Naho Yokota, Wolfram Ruf*Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA

    A R T I C L E I N F O A B S T R A C T

    Keywords:Tissue factorProtease activated receptorAngiogenensisCoagulation factor VIIMetastasisProtein C receptor

    Cancer progression from a dormant, non-vascularized benign tumor to metastatic disease is a multiple stepsprocess that critically depends on contributions from the hemostatic system. Tissue factor (TF), protease ac-tivated receptors (PARs), factor VIIa, and the endothelial protein C receptor (EPCR) are expressed by tumorcells as well as the host compartment. These components of the hemostatic system regulate tumor growth,angiogenesis and metastasis. Here we review the evidence that TF-dependent signaling is the major driver of primary tumor growth, whereas TF-initiated coagulation and interactions of procoagulant tumor cells withthe host compartments initiate multiple pathways that support and regulate the efficiency of metastatic tu-mor dissemination.

    2012 Elsevier Ltd. All rights reserved.

    1. Tissue factor procoagulant activity in hematogenousmetastasis

    Coagulation is an important step of tumor dissemination andtissue factor (TF) on tumor cells is the key initiator of the coagula-tion activation in experimental lung metastasis [1]. Reconstitution

    of TF-decient cells with procoagulant TF [24], modulation of TF activity by antibody [1] and inhibitors including tissue factorpathway inhibitor (TFPI) [5,6], or TF knock-down [7] demonstratethat TF is crucial for the survival of metastatic cells arrested in themicrocirculation. The TF-VIIa complex activates coagulation [4] andgenerates thrombin that is a central mediator for successful tumorcell implantation in pulmonary metastasis models [8]. Thrombinactivates platelet through protease activator receptor (PAR) 4 inmice [9]. Activated platelets use surface assembled bronectinand von Willebrand factor (VWF) as an adhesive bridge to tumorcells [10] and thrombin-generated brin further stabilizes tumorcell-associated clots. Platelets and brin protect tumor cells fromnatural killer (NK) cell-mediated attack and thus assure the survivalof metastatic cells after arrest at the endothelial interface [2,11

    13]. Recent studies show that these initial interactions of tumorcells with the endothelium are regulated at multiple levels by thehemostatic system.

    2. Regulation of thrombin thresholds at the endothelialinterface

    Thrombin not only promotes clot formation, but is also cru-cial for the anticoagulant pathway upon binding to endothelial

    * Correspondence to: Wolfram Ruf, Department of Immunology and MicrobialScience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla,CA 92037, USA. Tel.: +1 858-784-2748; fax: +1 858-784-8480.

    E-mail address: [email protected] (W. Ruf).

    0049-3848/$ see front matter 2012 Elsevier Ltd. All rights reserved.

    cell-expressed thrombomodulin (TM). The TM-thrombin complexgenerates activated protein C (aPC) facilitated by binding of PCto the endothelial protein C receptor (EPCR). The anticoagulantsystem, specically TM, aPC, and EPCR, has recently been shownto play important regulatory roles in tumor metastasis [1416].TM mutant mice (TM Pro ) have a single amino acid substitution

    (Glu404 /Pro) and exhibit 1,000-fold decreased PC activation and100-fold reduced binding affinity for thrombin [17]. The resulting

    combined loss of local thrombin neutralization and diminished aPCgeneration results in a dramatically increased metastasis in TM Pro

    mice that is dependent on tumor cell TF, circulating prothrombin,as well as platelets [14].

    Diminished activation of the anticoagulant pathway is at leastin part responsible for the enhanced metastasis in TM Pro mice.Although EPCR is expressed by tumor cells [1820], transgenicoverexpression of endothelial cell EPCR in the host compartmentor treatment with recombinant aPC markedly reduce experimentalmetastasis in liver and lung [16], whereas inhibition of endogenousaPC increases metastasis [15]. Interestingly, when the anticoagulantactivity of the endogenous aPC is blocked, brin depletion has

    only a minimal effect on metastasis [13], indicating that a hyper-thrombotic state diminishes the contributions of certain thrombintargets to successful tumor cell implantation at metastatic sites.EPCR-aPC-PAR1 signaling promotes endothelial barrier stabilizationthrough sphingosine 1 phosphate receptor 1 cross-activation andthrombin-PAR1 signaling can disrupt endothelial barrier integrity[21,22]. Therefore, regulation of barrier function may contributeto the anti-metastatic effects of aPC [15,23]. However, PAR1-deciency, which in the mouse circulatory system mainly affectsendothelial thrombin signaling, does not alter metastasis [9]. Fur-ther studies are needed to fully understand the precise mechanismsby which a prothombotic state enhances metastasis.

  • 8/13/2019 Tissue Factor Proangiogenic Signaling in Cancer Progression

    2/5

    S128 F. Schaffner et al. / Thrombosis Research 129, Supplement 1 (2012) S127S131

    3. Multicellular interactions contribute to successful tumor cellmetastasis

    Substantial evidence indicates that the initial recruitment of platelets by tumor cells has broader effects on multiple cell typesin the metastatic niche. Coagulation activation not only generatesa protective barrier against immune surveillance, but facilitatesthe initial steps of tumor cell extravasation [24,25]. Plateletsdirectly stimulate tumor growth and inuence angiogenesis byreleasing a variety of growth factors and chemokines stored in theirgranules [26,27]. The aggregates of activated platelets and tumorcells arrested at the endothelium furthermore recruit leukocytesinvolving mechanisms of tethering and adhesion typically employedin inammation. These interactions are supported by selectins,integrins, platelet receptors, and soluble and extracellular matrixproteins.

    Selectins, a family of transmembrane cell adhesion moleculesexpressed by platelets (P-selectin), endothelial cells (P- and E-selectin), and leukocytes (L-selectin), mediate the initial attachmentof circulating tumor cells on the endothelium and the subsequentrecruitment of leukocytes [28]. Tumor cells can express selectinligands, such as P-selectin glycoprotein (PSGL)-1, mucins, and CD44that further contribute to tumor cell-platelet interaction and alsodirectly trigger clot formation [29]. Experimental metastasis issignicantly reduced in the absence of P- or L-selectin on hostcells, while no attenuation of metastasis was observed in E-selectindecient mice [3036]. However, mice overexpressing E selectin inliver redirects metastasis from lungs to liver [37]. The combinedabsence of P- and L- selectin further reduced metastasis, indicatingbroad roles of blood- and endothelial cell-expressed selectins insupporting multicellular aggregate formation at sites of metastasis[33].

    Selectins mediate initial tethering that is followed by integrin-mediated rm cell adhesion. Key integrins supporting multicellularaggregates in the metastatic process are IIb3 on platelets [38,39],v3 on tumor cells [40], and M2 on leukocytes. Platelet P-

    selectin ligation of PSGL-1 on leukocyte activate integrin M2which plays an important role in mediating leukocyte interactionswith tumor cells involving brinogen and brin bridging to in-tegrin v3 and ICAM-1 [41,42]. Interaction between ICAM-1 onmelanoma cells and 2 integrins on leukocyte facilitates adhesionto the endothelium and subsequent extravasation in in vitro models[43].

    Selectin-dependent interaction between tumor cells, platelets,and leukocytes induces the activation of endothelial cells to re-lease the C-C motif chemokine 5 (CCL5), which recruits monocytesto support metastasis [44]. The important role of leukocyte re-cruitment to sites of metastasis is further demonstrated by thepro-metastatic effect of the chemokine CCL2 (MCP-1) that canbe derived from either stromal or tumor cells. CCL2 leads to

    the recruitment of inammatory monocytes, which express thechemokine CCL2 receptor CCR2, as well as to the association of macrophages with metastasizing tumor cells [45]. These stud-ies provide initial evidence that tumor cell-initiated formation of micro-thrombi supports complex heterotypic cellular interactionscontributing to successful metastatic tumor cell dissemination.

    4. Hemostatic mechanisms triggering early events in metastasis

    The hemostatic system is not only pivotal for the nal stagesof metastatic homing, but plays critical roles in the initiation of metastasis. TF is upregulated in tumor cells selected for increasedintravasation into the blood stream from the highly vascularizedchicken chorioallantoic membrane (CAM) [46]. In these cells se-lected for more efficient metastasis in the CAM model, blockingTF by either RNA silencing or antibody inhibition of specically

    TFs coagulation activity reduced tumor cell intravasation withoutimpairing primary tumor growth. The tumor microenvironment isconstantly exposed to components of the hemostatic systems due tothe abnormal architecture and hyperpermeability of tumor vessels.A recent study uncovered an intriguing role for platelet interactionswith tumor cells to promote early stages of metastasis. Activatedplatelets release large amounts of TGF which synergizes withdirect platelet-tumor cell contacts to activate TGF /Smad and NF-B pathways in cancer cells [47]. These signaling events promoteepithelial to mesenchymal transition of cancer cells, a key featureof more metastatic tumor cell populations. These studies documenta wide range of effects mediated by distinct components of thehemostatic system on early and late stages of tumor progressionand metastasis.

    5. TF signaling in tumor angiogenesis

    TF also supports primary tumor growth [4851]. Unlike the pro-coagulant roles of TF in metastasis, our studies show that the tumorgrowth promoting activities of TF are predominantly dependent ondirect cell signaling [52]. Tumor cells can ectopically synthesize VIIa,either constitutively or in response to hypoxia [53,54] and onco-genic growth factor receptors upregulate the entire complement of the upstream TF signaling complex [55]. The tumor cell-expressedTF-VIIa complex cleaves PAR2 to inuence important aspects of tumor progression, including survival, immune modulation andangiogenesis. TF-VIIa-PAR2 G protein-coupled receptor signaling in-duces pro-angiogenic factors, such as IL-8, CXCL1, or VEGF [5659]and growth factors for myeloid cells and macrophages [57].

    TF-VIIa-PAR2 signaling is not necessarily restricted to autocrinetumor cell signaling. For example, glioblastoma cells express highlevels of TF that drives tumor progression [60,61]. In the hypoxictumor microenvironment, glioblastoma cells also release TF + microparticles that elicit TF-VIIa signaling in trans by targeting hypoxicendothelial cells that upregulate PAR2 [62]. Microparticle-inducedtrans-signaling specically induces endothelial cell expression of

    HB-EGF that supports angiogenesis and activates the MET receptoron tumor cells to enhance their survival, proliferation and migration[63] and potentially induce a prothrombotic state [64]. Thus, TF-VIIa-PAR2 signaling contributes to tumor initiation and progressionunder hypoxia.

    The critical role of TF-VIIa-PAR2 signaling in tumor angiogenesisis supported by genetic and pharmacological evidence. In thepolyoma middle T (PyMT) oncogene-driven model of spontaneousbreast cancer development, tumor progression occurs in the contextof an immune competent host and the angiogenic switch isdependent on the recruitment of immune cells and macrophagesto the tumor stroma [65]. PAR2-deciency results in delayed PyMTtumor development, low levels of the chemokine CXCL1 (KC) in thetumor stroma, and reduced counts of F4/80 positive macrophages

    in early tumors compared to wild-type mice [66]. Reconstitutionexperiments of PAR2-decient tumor cells derived from these miceshowed that tumor cell autonomous TF-PAR2 signaling supportedCXCL1 induction in vitro and a more aggressive tumor growthphenotype in vivo [67].

    In clinical breast cancer biopsies, upregulation of PAR2 andTF was associated with a marked phosphorylation of the TFcytoplasmic domain [68]. Only patients with phosphorylated TFhad a cancer relapse and genetic studies in mice showed thatPAR2 signaling was important for tumor cell TF cytoplasmic domainphosphorylation in vivo. Conrming that the TF cytoplasmic domainplayed an important role in tumor progression, PyMT tumordevelopment was delayed in mice lacking the TF cytoplasmicdomain similar to PAR2-decient animals [67]. Late stage tumors of TF cytoplasmic domain-deleted mice also displayed altered vesselarchitecture and reduced macrophages numbers in the tumor

  • 8/13/2019 Tissue Factor Proangiogenic Signaling in Cancer Progression

    3/5

  • 8/13/2019 Tissue Factor Proangiogenic Signaling in Cancer Progression

    4/5

    S130 F. Schaffner et al. / Thrombosis Research 129, Supplement 1 (2012) S127S131

    Conict of interest statement

    Wolfram Ruf has pending patent applications on the use of antibodies described in this review.

    References

    [1] Mueller BM, Reisfeld RA, Edgington TS, Ruf W. Expression of tissue factor bymelanoma cells promotes efficient hematogenous metastasis. Proc Natl AcadSci USA 1992;89(24):118326.

    [2] Palumbo JS, Talmage KE, Massari JV, La Jeunesse CM, Flick MJ, KombrinckKW, et al. Tumor cell-associated tissue factor and circulating hemostaticfactors cooperate to increase metastatic potential through natural killer cell-dependent and -independent mechanisms. Blood 2007;110:13341.

    [3] Bromberg ME, Konigsberg WH, Madison JF, Pawashe A, Garen A. Tissuefactor promotes melanoma metastasis by a pathway independent of bloodcoagulation. Proc Natl Acad Sci USA 1995;92:82059.

    [4] Mueller BM, Ruf W. Requirement for binding of catalytically active fac-tor VIIa in tissue factor dependent experimental metastasis. J Clin Invest1998;101(7):13728.

    [5] Amirkhosravi A, Meyer T, Chang JY, Amaya M, Siddiqui F, Desai H, et al.Tissue factor pathway inhibitor reduces experimental lung metastasis of B16melanoma. Thromb Haemost 2002;87(6):9306.

    [6] Zhao J, Aguilar G, Palencia S, Newton E, Abo A. rNAPc2 inhibits colorectalcancer in mice through tissue factor. Clin Cancer Res 2009;15(1):20816.

    [7] Wang X, Wang M, Amarzguioui M, Liu F, Fodstad O, Prydz H. Downregulationof tissue factor by RNA interference in human melanoma LOX-L cells reducespulmonary metastasis in nude mice. Int J Cancer 2004;112(6):9941002.

    [8] Esumi N, Fan D, Fidler IJ. Inhibition of murine melanoma experimental meta-stasis by recombinant desulfatohirudin, a highly specic thrombin inhibitor.Cancer Res 1991;51:454956.

    [9] Camerer E, Qazi AA, Duong DN, Cornelissen I, Advincula R, Coughlin SR.Platelets, protease-activated receptors, and brinogen in hematogenous meta-stasis. Blood 2004;104(2):397401.

    [10] Nierodzik ML, Karpatkin S. Thrombin induces tumor growth, metastasis, andangiogenesis: Evidence for a thrombin-regulated dormant tumor phenotype.Cancer Cell 2006;10(5):35562.

    [11] Nieswandt B, Hafner M, Echtenacher B, Mannel DN. Lysis of tumorcells by natural killer cells in mice is impeded by platelets. Cancer Res1999;59(6):1295300.

    [12] Palumbo JS, Talmage KE, Massari JV, La Jeunesse CM, Flick MJ, KombrinckKW, et al. Platelets and brin(ogen) increase metastatic potential by impeding

    natural killer-mediated elimination of tumor cells. Blood 2004;105(1):17885.[13] Van Sluis GL, Bruggemann LW, Esmon CT, Kamphuisen PW, Richel DJ, Buller

    HR, et al. Endogenous activated protein C is essential for immune-mediatedcancer cell elimination from the circulation. Cancer Lett 2011;306(1):10610.

    [14] Horowitz NA, Blevins EA, Miller WM, Perry AR, Talmage KE, Mullins ES, etal. Thrombomodulin is a determinant of metastasis through a mechanismlinked to the thrombin binding domain but not the lectin-like domain. Blood2011;118(10):288995.

    [15] Van Sluis GL, Niers TM, Esmon CT, Tigchelaar W, Richel DJ, Buller HR, etal. Endogenous activated protein C limits cancer cell extravasation throughsphingosine-1-phosphate receptor 1-mediated vascular endothelial barrierenhancement. Blood 2009;114(9):196873.

    [16] Bezuhly M, Cullen R, Esmon CT, Morris SF, West KA, Johnston B, et al. Role of activated protein C and its receptor in inhibition of tumor metastasis. Blood2009;113(14):33714.

    [17] Weiler-Guettler H, Christie PD, Beeler DL, Healy AM, Hancock WW, RayburnH, et al. A targeted point mutation in thrombomodulin generates viable mice

    with a prethrombotic state. J Clin Invest 1998;101(9):198391.[18] Shipitsin M, Campbell LL, Argani P, Weremowicz S, Bloushtain-Qimron N,Yao J, et al. Molecular denition of breast tumor heterogeneity. Cancer Cell2007;11(3):25973.

    [19] Hwang-Verslues WW, Kuo WH, Chang PH, Pan CC, Wang HH, Tsai ST, et al.Multiple lineages of human breast cancer stem/progenitor cells identied byproling with stem cell markers. PLoS ONE 2009;4(12):e8377.

    [20] Beaulieu LM, Church FC. Activated protein C promotes breast cancercell migration through interactions with EPCR and PAR-1. Exp Cell Res2007;313(4):67787.

    [21] Feistritzer C, Riewald M. Endothelial barrier protection by activated protein Cthrough PAR1-dependent sphingosine 1-phosphate receptor-1 crossactivation.Blood 2005;105(8):317884.

    [22] Finigan JH, Dudek SM, Singleton PA, Chiang ET, Jacobson JR, Camp SM, etal. Activated protein C mediates novel lung endothelial barrier enhance-ment: role of sphingosine 1-phosphate receptor transactivation. J Biol Chem2005;280(17):1728693.

    [23] Van Sluis GL, Buller HR, Spek CA. The role of activated protein C in cancerprogression. Thromb Res 2010;125(Suppl 2):S13842.

    [24] Im JH, Fu W, Wang H, Bhatia SK, Hammer DA, Kowalska MA, et al.Coagulation facilitates tumor cell spreading in the pulmonary vasculatureduring early metastatic colony formation. Cancer Res 2004;64(23):86139.

    [25] Wang H, Fu W, Im JH, Zhou Z, Santoro SA, Iyer V, et al. Tumor cellalpha3beta1 integrin and vascular laminin-5 mediate pulmonary arrest andmetastasis. J Cell Biol 2004;164(6):93541.

    [26] Gay LJ, Felding-Habermann B. Contribution of platelets to tumour metastasis.Nat Rev Cancer 2011;11(2):12334.

    [27] Sabrkhany S, Griffioen AW, oude Egbrink MG. The role of blood platelets in

    tumor angiogenesis. Biochim Biophys Acta 2011;1815(2):18996.[28] St Hill CA. Interactions between endothelial selectins and cancer cells regulatemetastasis. Front Biosci 2011;17:323351.

    [29] Shao B, Wahrenbrock MG, Yao L, David T, Coughlin SR, Xia L, et al. Carcinomamucins trigger reciprocal activation of platelets and neutrophils in a murinemodel of Trousseau syndrome. Blood 2011;118(15):401523.

    [30] Barthel SR, Gavino JD, Descheny L, Dimitroff CJ. Targeting selectins andselectin ligands in inammation and cancer. Expert Opin Ther Targets2007;11(11):147391.

    [31] Chen M, Geng JG. P-selectin mediates adhesion of leukocytes, platelets, andcancer cells in inammation, thrombosis, and cancer growth and metastasis.Arch Immunol Ther Exp (Warsz ) 2006;54(2):7584.

    [32] Kim YJ, Borsig L, Varki NM, Varki A. P-selectin deciency attenuates tumorgrowth and metastasis. Proc Natl Acad Sci U S A 1998;95(16):932530.

    [33] Borsig L, Wong R, Hynes RO, Varki NM, Varki A. Synergistic effects of L- andP-selectin in facilitating tumor metastasis can involve non-mucin ligands andimplicate leukocytes as enhancers of metastasis. Proc Natl Acad Sci U S A2002;99(4):21938.

    [34] Monzavi-Karbassi B, Whitehead TL, Jousheghany F, Artaud C, Hennings L,Shaaf S, et al. Deciency in surface expression of E-selectin ligand pro-motes lung colonization in a mouse model of breast cancer. Int J Cancer2005;117(3):398408.

    [35] Laubli H, Stevenson JL, Varki A, Varki NM, Borsig L. L-selectin facilitation of metastasis involves temporal induction of Fut7-dependent ligands at sites of tumor cell arrest. Cancer Res 2006;66(3):153642.

    [36] Laubli H, Borsig L. Selectins as mediators of lung metastasis. Cancer Microen-viron 2010;3(1):97105.

    [37] Biancone L, Araki M, Araki K, Vassalli P, Stamenkovic I. Redirection of tumormetastasis by expression of E-selectin in vivo. J Exp Med 1996;183(2):5817.

    [38] Karpatkin S, Pearlstein E, Ambrogio C, Coller BS. Role of adhesive protein inplatelet tumor interaction in vitro and metastasis formation in vivo. J ClinInvest 1988;81:10129.

    [39] Amirkhosravi A, Mousa SA, Amaya M, Blaydes S, Desai H, Meyer T, et al.Inhibition of tumor cell-induced platelet aggregation and lung metastasis bythe oral GpIIb/IIIa antagonist XV454. Thromb Haemost 2003;90(3):54954.

    [40] Felding-Habermann B, OToole TE, Smith JW, Fransvea E, Ruggeri ZM, GinsbergMH, et al. Integrin activation controls metastasis in human breast cancer. ProcNatl Acad Sci U S A 2001;98(4):18538.

    [41] Cerletti C, Tamburrelli C, Izzi B, Gianfagna F, de GG. Platelet-leukocyteinteractions in thrombosis. Thromb Res 2011;.(.):.

    [42] Zhang P, Ozdemir T, Chung CY, Robertson GP, Dong C. Sequential binding of alphaVbeta3 and ICAM-1 determines brin-mediated melanoma capture andstable adhesion to CD11b/CD18 on neutrophils. J Immunol 2011;186(1):24254.

    [43] Liang S, Hoskins M, Dong C. Tumor cell extravasation mediated by leukocyteadhesion is shear rate dependent on IL-8 signaling. Mol Cell Biomech2010;7(2):7791.

    [44] Laubli H, Spanaus KS, Borsig L. Selectin-mediated activation of endothelialcells induces expression of CCL5 and promotes metastasis through recruit-ment of monocytes. Blood 2009;114(20):458391.

    [45] Qian BZ, Li J, Zhang H, Kitamura T, Zhang J, Campion LR, et al. CCL2 re-cruits inammatory monocytes to facilitate breast-tumour metastasis. Nature2011;475(7355):2225.

    [46] Conn EM, Madsen MA, Cravatt BF, Ruf W, Deryugina EI, Quigley JP. Cellsurface proteomics identies molecules functionally linked to tumor cellintravasation. J Biol Chem 2008;283(39):2651827.

    [47] Labelle M, Begum S, Hynes RO. Direct Signaling between Platelets andCancer Cells Induces an Epithelial-Mesenchymal-Like Transition and PromotesMetastasis. Cancer Cell 2011;20(5):57690.

    [48] Zhang Y, Deng Y, Luther T, Mller M, Ziegler R, Waldherr R, et al. Tissuefactor controls the balance of angiogenic and antiangiogenic properties of tumor cells in mice. J Clin Invest 1994;94:13207.

    [49] Yu JL, May L, Lhotak V, Shahrzad S, Shirasawa S, Weitz JI, et al. Oncogenicevents regulate tissue factor expression in colorectal cancer cells: implicationsfor tumor progression and angiogenesis. Blood 2005;105(4):173441.

    [50] Kakkar AK, Chinswangwatanakul V, Lemoine NR, Tebbutt S, Williamson RCN.Role of tissue factor expression on tumour cell invasion and growth of experimental pancreatic adenocarcinoma. Br J Surg 1999;86:8904.

    [51] Liu Y, Jiang P, Capkova K, Xue D, Ye L, Sinha SC, et al. Tissue factor-activatedcoagulation cascade in the tumor microenvironment is critical for tumor

    progression and an effective target for therapy. Cancer Res 2011;71(20):6492502.

  • 8/13/2019 Tissue Factor Proangiogenic Signaling in Cancer Progression

    5/5

    F. Schaffner et al. / Thrombosis Research 129, Supplement 1 (2012) S127S131 S131

    [52] Ruf W, Disse J, Carneiro-Lobo TC, Yokota N, Schaffner F. Tissue factor andcell signalling in cancer progression and thrombosis. J Thromb Haemost2011;9(Suppl 1):30615.

    [53] Koizume S, Jin M-S, Miyagi E, Hirahara F, Nakamura Y, Piao J-H, et al.Activation of cancer cell migration and invasion by ectopic synthesis of coagulation factor VII. Cancer Res 2006;66(19):945360.

    [54] Koizume S, Yokota N, Miyagi E, Hirahara F, Nakamura Y, Sakuma Y, etal. Hepatocyte nuclear factor-4-independent synthesis of coagulation factorVII in breast cancer cells and its inhibition by targeting selective histone

    acetyltransferases. Mol Cancer Res 2009;7(12):192836.[55] Magnus N, Garnier D, Rak J. Oncogenic epidermal growth factor receptor up-regulates multiple elements of the tissue factor signaling pathway in humanglioma cells. Blood 2010;116(5):8158.

    [56] Hjortoe GM, Petersen LC, Albrektsen T, Sorensen BB, Norby PL, Mandal SK, etal. Tissue factor-factor VIIa specic up-regulation of IL-8 expression in MDA-MB-231 cells is mediated via PAR-2 and results in increased cell migration.Blood 2004;103(8):302937.

    [57] Albrektsen T, Sorensen BB, Hjortoe GM, Fleckner J, Rao LVM, Petersen LC.Transcriptional program induced by factor VIIa-tissue factor, PAR1 and PAR2in MDA-MB-231 cells. J Thromb Haemost 2007;5:158897.

    [58] Versteeg HH, Schaffner F, Kerver M, Petersen HH, Ahamed J, Felding-Habermann B, et al. Inhibition of tissue factor signaling suppresses tumorgrowth. Blood 2008;111(1):1909.

    [59] Liu Y, Mueller BM. Protease-activated receptor-2 regulates vascular endothe-lial growth factor expression in MDA-MB-231 cells via MAPK pathways.Biochem Biophys Res Commun 2006;344(4):126370.

    [60] Carneiro-Lobo TC, Konig S, Machado DE, Nasciutti LE, Forni MF, FrancischettiIM, et al. Ixolaris, a tissue factor inhibitor, blocks primary tumor growth andangiogenesis in a glioblastoma model. J Thromb Haemost 2009;7(11):185564.

    [61] Gessler F, Voss V, Dutzmann S, Seifert V, Gerlach R, Kogel D. Inhibition of tis-sue factor/protease-activated receptor-2 signaling limits proliferation, migra-tion and invasion of malignant glioma cells. Neuroscience 2010;165(4):131222.

    [62] Svensson KJ, Kucharzewska P, Christianson HC, Skold S, Lofstedt T, JohanssonMC, et al. Hypoxia triggers a pro-angiogenic pathway involving cancer cellmicrovesicles and PAR-2 mediated HB-EGF signaling in endothelial cells. ProcNatl Acad Sci USA 2011;108:1314752.

    [63] Trusolino L, Bertotti A, Comoglio PM. MET signalling: principles and functionsin development, organ regeneration and cancer. Nat Rev Mol Cell Biol2010;11(12):83448.

    [64] Boccaccio C, Sabatino G, Medico E, Girolami F, Follenzi A, Reato G, et al. TheMET oncogene drives a genetic programme linking cancer to haemostasis.Nature 2005;434(7031):396400.

    [65] Lin EY, Li JF, Gnatovskiy L, Deng Y, Zhu L, Grzesik DA, et al. Macrophagesregulate the angiogenic switch in a mouse model of breast cancer. Cancer Res2006;66(23):1123846.

    [66] Versteeg HH, Schaffner F, Kerver M, Ellies LG, Andrade-Gordon P, Mueller BM,et al. Protease activated receptor (PAR)2, but not PAR1 signaling promotesthe development of mammary adenocarcinoma in PyMT mice. Cancer Res2008;68(17):721927.

    [67] Schaffner F, Versteeg HH, Schillert A, Yokota N, Petersen LC, Mueller BM, etal. Cooperation of tissue factor cytoplasmic domain and PAR2 signaling in

    breast cancer development. Blood 2010;116(26):610613.[68] Ryden L, Grabau D, Schaffner F, Jonsson PE, Ruf W, Belting M. Evidencefor tissue factor phosphorylation and its correlation with protease activatedreceptor expression and the prognosis of primary breast cancer. Int J Cancer2010;126(10):233040.

    [69] Belting M, Dorrell MI, Sandgren S, Aguilar E, Ahamed J, Doreutner A, etal. Regulation of angiogenesis by tissue factor cytoplasmic domain signaling.Nature Med 2004;10(5):5029.

    [70] Uusitalo-Jarvinen H, Kurokawa T, Mueller BM, Andrade-Gordon P, FriedlanderM, Ruf W. Role of Protease Activated Receptor 1 and 2 Signaling in Hypoxia-Induced Angiogenesis. Arterioscler Thromb Vasc Biol 2007;27(6):145662.

    [71] Ahamed J, Versteeg HH, Kerver M, Chen VM, Mueller BM, Hogg PJ, etal. Disulde isomerization switches tissue factor from coagulation to cellsignaling. Proc Natl Acad Sci USA 2006;103(38):139327.

    [72] Gimbrone MA, Jr., Leapman SB, Cotran RS, Folkman J. Tumor dormancy invivo by prevention of neovascularization. J Exp Med 1972;136(2):26176.

    [73] Fischer C, Jonckx B, Mazzone M, Zacchigna S, Loges S, Pattarini L, et al. Anti-PlGF inhibits growth of VEGF(R)-inhibitor-resistant tumors without affectinghealthy vessels. Cell 2007;131(3):46375.

    [74] Witte L, Hicklin DJ, Zhu Z, Pytowski B, Kotanides H, Rockwell P, et al.Monoclonal antibodies targeting the VEGF receptor-2 (Flk1/KDR) as an anti-angiogenic therapeutic strategy. Cancer Metastasis Rev 1998;17(2):15561.

    [75] Srinivasan R, Ozhegov E, van den Berg YW, Aronow BJ, Franco RS, PalascakMB, et al. Splice Variants of Tissue Factor Promote Monocyte-EndothelialInteractions by Triggering the Expression of Cell Adhesion Molecules viaIntegrin-Mediated Signaling. J Thromb Haemost 2011; in press.

    [76] van den Berg YW, van den Hengel LG, Myers HR, Ayachi O, Jordanova E,Ruf W, et al. Alternatively spliced tissue factor induces angiogenesis throughintegrin ligation. Proc Natl Acad Sci U S A 2009;106(46):19497502.

    [77] Saito Y, Hashimoto Y, Kuroda J, Yasunaga M, Koga Y, Takahashi A, et al. Theinhibition of pancreatic cancer invasion-metastasis cascade in both cellularsignal and blood coagulation cascade of tissue factor by its neutralisationantibody. Eur J Cancer 2011;47(14):22309.