Skip to main content
Top
Published in: Tumor Biology 9/2014

01-09-2014 | Review

Metastasis review: from bench to bedside

Authors: Ali Mohammad Alizadeh, Sadaf Shiri, Sadaf Farsinejad

Published in: Tumor Biology | Issue 9/2014

Login to get access

Abstract

Cancer is the final result of uninhibited cell growth that involves an enormous group of associated diseases. One major aspect of cancer is when cells attack adjacent components of the body and spread to other organs, named metastasis, which is the major cause of cancer-related mortality. In developing this process, metastatic cells must successfully negotiate a series of complex steps, including dissociation, invasion, intravasation, extravasation, and dormancy regulated by various signaling pathways. In this review, we will focus on the recent studies and collect a comprehensive encyclopedia in molecular basis of metastasis, and then we will discuss some new potential therapeutics which target the metastasis pathways. Understanding the new aspects on molecular mechanisms and signaling pathways controlling tumor cell metastasis is critical for the development of therapeutic strategies for cancer patients that would be valuable for researchers in both fields of molecular and clinical oncology.
Literature
1.
go back to reference Jemal A et al. Global cancer statistics. CA: Cancer J Clin. 2011;61(2):69–90. Jemal A et al. Global cancer statistics. CA: Cancer J Clin. 2011;61(2):69–90.
2.
go back to reference Jonsson B, Wilking N. A global comparison regarding patient access to cancer drugs. Ann Oncol. 2007;18:1–78. Jonsson B, Wilking N. A global comparison regarding patient access to cancer drugs. Ann Oncol. 2007;18:1–78.
3.
go back to reference Miovic M, Block S. Psychiatric disorders in advanced cancer. Cancer. 2007;110(8):1665–76.PubMed Miovic M, Block S. Psychiatric disorders in advanced cancer. Cancer. 2007;110(8):1665–76.PubMed
4.
go back to reference Khan N, Mukhtar H. Cancer and metastasis: prevention and treatment by green tea. Cancer Metastasis Rev. 2010;29(3):435–45.PubMedCentralPubMed Khan N, Mukhtar H. Cancer and metastasis: prevention and treatment by green tea. Cancer Metastasis Rev. 2010;29(3):435–45.PubMedCentralPubMed
5.
go back to reference Weigelt B, Peterse JL, Van’t Veer LJ. Breast cancer metastasis: markers and models. Nat Rev Cancer. 2005;5(8):591–602.PubMed Weigelt B, Peterse JL, Van’t Veer LJ. Breast cancer metastasis: markers and models. Nat Rev Cancer. 2005;5(8):591–602.PubMed
6.
go back to reference Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat Med. 2004;10(8):789–99.PubMed Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat Med. 2004;10(8):789–99.PubMed
7.
go back to reference Ferlay J et al. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127(12):2893–917.PubMed Ferlay J et al. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127(12):2893–917.PubMed
8.
go back to reference Liotta LA, Kohn EC. Cancer’s deadly signature. Nat Genet. 2003;33(1):10–1.PubMed Liotta LA, Kohn EC. Cancer’s deadly signature. Nat Genet. 2003;33(1):10–1.PubMed
9.
go back to reference Fidler IJ. The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat Rev Cancer. 2003;3(6):453–8.PubMed Fidler IJ. The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat Rev Cancer. 2003;3(6):453–8.PubMed
10.
go back to reference Thiery JP, Sleeman JP. Complex networks orchestrate epithelial–mesenchymal transitions. Nat Rev Mol Cell Biol. 2006;7(2):131–42.PubMed Thiery JP, Sleeman JP. Complex networks orchestrate epithelial–mesenchymal transitions. Nat Rev Mol Cell Biol. 2006;7(2):131–42.PubMed
11.
go back to reference Nguyen DX, Bos PD, Massagué J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 2009;9(4):274–84.PubMed Nguyen DX, Bos PD, Massagué J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 2009;9(4):274–84.PubMed
12.
go back to reference Chung LW et al. Molecular insights into prostate cancer progression: the missing link of tumor microenvironment. J Urol. 2005;173(1):10–20.PubMed Chung LW et al. Molecular insights into prostate cancer progression: the missing link of tumor microenvironment. J Urol. 2005;173(1):10–20.PubMed
13.
go back to reference Wang J, Loberg R, Taichman RS. The pivotal role of CXCL12 (SDF-1)/CXCR4 axis in bone metastasis. Cancer Metastasis Rev. 2006;25(4):573–87.PubMed Wang J, Loberg R, Taichman RS. The pivotal role of CXCL12 (SDF-1)/CXCR4 axis in bone metastasis. Cancer Metastasis Rev. 2006;25(4):573–87.PubMed
14.
go back to reference Weinberg RA. Is metastasis predetermined? Mol Oncol. 2007;1(3):263–4.PubMed Weinberg RA. Is metastasis predetermined? Mol Oncol. 2007;1(3):263–4.PubMed
15.
go back to reference Hyoudou K et al. Inhibition of metastatic tumor growth in mouse lung by repeated administration of polyethylene glycol-conjugated catalase quantitative analysis with firefly luciferase-expressing melanoma cells. Clin Cancer Res. 2004;10(22):7685–91.PubMed Hyoudou K et al. Inhibition of metastatic tumor growth in mouse lung by repeated administration of polyethylene glycol-conjugated catalase quantitative analysis with firefly luciferase-expressing melanoma cells. Clin Cancer Res. 2004;10(22):7685–91.PubMed
16.
go back to reference Folkman J, Klagsbrun M. Angiogenic factors. Science. 1987;235(4787):442–7.PubMed Folkman J, Klagsbrun M. Angiogenic factors. Science. 1987;235(4787):442–7.PubMed
17.
go back to reference Leong SP et al. Clinical patterns of metastasis. Cancer Metastasis Rev. 2006;25(2):221–32.PubMed Leong SP et al. Clinical patterns of metastasis. Cancer Metastasis Rev. 2006;25(2):221–32.PubMed
18.
go back to reference Tuttle TM. Technical advances in sentinel lymph node biopsy for breast cancer. Am Surg. 2004;70(5):407–13.PubMed Tuttle TM. Technical advances in sentinel lymph node biopsy for breast cancer. Am Surg. 2004;70(5):407–13.PubMed
19.
go back to reference Pepper MS et al. Lymphangiogenesis and tumor metastasis. Cell Tissue Res. 2003;314(1):167–77.PubMed Pepper MS et al. Lymphangiogenesis and tumor metastasis. Cell Tissue Res. 2003;314(1):167–77.PubMed
20.
go back to reference Bacac M, Stamenkovic I. Metastatic cancer cell. Annu Rev Pathmechdis Mech Dis. 2008;3:221–47. Bacac M, Stamenkovic I. Metastatic cancer cell. Annu Rev Pathmechdis Mech Dis. 2008;3:221–47.
21.
go back to reference Brooks SA et al. Molecular interactions in cancer cell metastasis. Acta Histochemica. 2010;112(1):3–25.PubMed Brooks SA et al. Molecular interactions in cancer cell metastasis. Acta Histochemica. 2010;112(1):3–25.PubMed
22.
go back to reference Spano D, et al. Molecular networks that regulate cancer metastasis. in Seminars in cancer biology. 2012. Elsevier. Spano D, et al. Molecular networks that regulate cancer metastasis. in Seminars in cancer biology. 2012. Elsevier.
23.
24.
go back to reference Chambers AF, Groom AC, MacDonald IC. Metastasis: dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2(8):563–72.PubMed Chambers AF, Groom AC, MacDonald IC. Metastasis: dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2(8):563–72.PubMed
25.
go back to reference Meighan CM, Schwarzbauer JE. Temporal and spatial regulation of integrins during development. Curr Opin Cell Biol. 2008;20(5):520–4.PubMedCentralPubMed Meighan CM, Schwarzbauer JE. Temporal and spatial regulation of integrins during development. Curr Opin Cell Biol. 2008;20(5):520–4.PubMedCentralPubMed
26.
go back to reference van Zijl F, Krupitza G, Mikulits W. Initial steps of metastasis: cell invasion and endothelial transmigration. Mutat Res/Rev Mutat Res. 2011;728(1):23–34. van Zijl F, Krupitza G, Mikulits W. Initial steps of metastasis: cell invasion and endothelial transmigration. Mutat Res/Rev Mutat Res. 2011;728(1):23–34.
27.
go back to reference Cavallaro U, Christofori G. Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer. 2004;4(2):118–32.PubMed Cavallaro U, Christofori G. Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer. 2004;4(2):118–32.PubMed
28.
go back to reference Engers R, Gabbert HE. Mechanisms of tumor metastasis: cell biological aspects and clinical implications. J Cancer Res Clin Oncol. 2000;126(12):682–92.PubMed Engers R, Gabbert HE. Mechanisms of tumor metastasis: cell biological aspects and clinical implications. J Cancer Res Clin Oncol. 2000;126(12):682–92.PubMed
29.
go back to reference Stoecklein NH, Klein CA. Genetic disparity between primary tumours, disseminated tumour cells, and manifest metastasis. Int J Cancer. 2010;126(3):589–98.PubMed Stoecklein NH, Klein CA. Genetic disparity between primary tumours, disseminated tumour cells, and manifest metastasis. Int J Cancer. 2010;126(3):589–98.PubMed
30.
31.
go back to reference Hood JD, Cheresh DA. Role of integrins in cell invasion and migration. Nat Rev Cancer. 2002;2(2):91–100.PubMed Hood JD, Cheresh DA. Role of integrins in cell invasion and migration. Nat Rev Cancer. 2002;2(2):91–100.PubMed
32.
go back to reference Angst BD, Marcozzi C, Magee AI. The cadherin superfamily: diversity in form and function. J Cell Sci. 2001;114(4):629–41.PubMed Angst BD, Marcozzi C, Magee AI. The cadherin superfamily: diversity in form and function. J Cell Sci. 2001;114(4):629–41.PubMed
33.
go back to reference Gumbiner BM. Regulation of cadherin-mediated adhesion in morphogenesis. Nat Rev Mol Cell Biol. 2005;6(8):622–34.PubMed Gumbiner BM. Regulation of cadherin-mediated adhesion in morphogenesis. Nat Rev Mol Cell Biol. 2005;6(8):622–34.PubMed
34.
go back to reference Takeichi M. Cadherin cell adhesion receptors as a morphogenetic regulator. Science. 1991;251(5000):1451–5.PubMed Takeichi M. Cadherin cell adhesion receptors as a morphogenetic regulator. Science. 1991;251(5000):1451–5.PubMed
35.
go back to reference Kashima T et al. Overexpression of cadherins suppresses pulmonary metastasis of osteosarcoma in vivo. Int J Cancer. 2003;104(2):147–54.PubMed Kashima T et al. Overexpression of cadherins suppresses pulmonary metastasis of osteosarcoma in vivo. Int J Cancer. 2003;104(2):147–54.PubMed
36.
go back to reference Shapiro L, Weis WI. Structure and biochemistry of cadherins and catenins. Cold Spring Harbor Perspect Biol. 2009;1(3):a003053. Shapiro L, Weis WI. Structure and biochemistry of cadherins and catenins. Cold Spring Harbor Perspect Biol. 2009;1(3):a003053.
37.
go back to reference Mohamet L.Hawkins K. and Ward C.M. Loss of function of e-cadherin in embryonic stem cells and the relevance to models of tumorigenesis. Journal of oncology, 2010. 2011. Mohamet L.Hawkins K. and Ward C.M. Loss of function of e-cadherin in embryonic stem cells and the relevance to models of tumorigenesis. Journal of oncology, 2010. 2011.
38.
go back to reference Li L, Bennett S, Wang L. Role of E-cadherin and other cell adhesion molecules in survival and differentiation of human pluripotent stem cells. Cell Adh Migr. 2012;6(1):59–70.PubMedCentralPubMed Li L, Bennett S, Wang L. Role of E-cadherin and other cell adhesion molecules in survival and differentiation of human pluripotent stem cells. Cell Adh Migr. 2012;6(1):59–70.PubMedCentralPubMed
39.
go back to reference Beavon I. The E-cadherin–catenin complex in tumour metastasis: structure, function and regulation. Eur J Cancer. 2000;36(13):1607–20.PubMed Beavon I. The E-cadherin–catenin complex in tumour metastasis: structure, function and regulation. Eur J Cancer. 2000;36(13):1607–20.PubMed
40.
go back to reference Leber MF, Efferth T. Molecular principles of cancer invasion and metastasis (review). Int J Oncol. 2009;34(4):881–95.PubMed Leber MF, Efferth T. Molecular principles of cancer invasion and metastasis (review). Int J Oncol. 2009;34(4):881–95.PubMed
41.
go back to reference Takeichi M. Morphogenetic roles of classic cadherins. Curr Opin Cell Biol. 1995;7(5):619–27.PubMed Takeichi M. Morphogenetic roles of classic cadherins. Curr Opin Cell Biol. 1995;7(5):619–27.PubMed
42.
go back to reference Provost E, Rimm DL. Controversies at the cytoplasmic face of the cadherin-based adhesion complex. Curr Opin Cell Biol. 1999;11(5):567–72.PubMed Provost E, Rimm DL. Controversies at the cytoplasmic face of the cadherin-based adhesion complex. Curr Opin Cell Biol. 1999;11(5):567–72.PubMed
43.
go back to reference Nagafuchi A. Molecular architecture of adherens junctions. Curr Opin Cell Biol. 2001;13(5):600–3.PubMed Nagafuchi A. Molecular architecture of adherens junctions. Curr Opin Cell Biol. 2001;13(5):600–3.PubMed
44.
go back to reference Ozawa M, Ringwald M, Kemler R. Uvomorulin-catenin complex formation is regulated by a specific domain in the cytoplasmic region of the cell adhesion molecule. Proc Natl Acad Sci U S A. 1990;87(11):4246–50.PubMedCentralPubMed Ozawa M, Ringwald M, Kemler R. Uvomorulin-catenin complex formation is regulated by a specific domain in the cytoplasmic region of the cell adhesion molecule. Proc Natl Acad Sci U S A. 1990;87(11):4246–50.PubMedCentralPubMed
45.
go back to reference Hazan RB et al. Cadherin switch in tumor progression. Ann N Y Acad Sci. 2004;1014(1):155–63.PubMed Hazan RB et al. Cadherin switch in tumor progression. Ann N Y Acad Sci. 2004;1014(1):155–63.PubMed
47.
go back to reference Niessen CM, Leckband D. Tissue organization by cadherin adhesion molecules: dynamic molecular and cellular mechanisms of morphogenetic regulation. Physiol Rev. 2011;91(2):691–731.PubMedCentralPubMed Niessen CM, Leckband D. Tissue organization by cadherin adhesion molecules: dynamic molecular and cellular mechanisms of morphogenetic regulation. Physiol Rev. 2011;91(2):691–731.PubMedCentralPubMed
48.
go back to reference Gumbiner BM. Cell adhesion: the molecular basis of tissue architecture and morphogenesis. Cell. 1996;84(3):345–57.PubMed Gumbiner BM. Cell adhesion: the molecular basis of tissue architecture and morphogenesis. Cell. 1996;84(3):345–57.PubMed
49.
go back to reference Hajra KM, Fearon ER. Cadherin and catenin alterations in human cancer. Genes Chromosom Cancer. 2002;34(3):255–68.PubMed Hajra KM, Fearon ER. Cadherin and catenin alterations in human cancer. Genes Chromosom Cancer. 2002;34(3):255–68.PubMed
51.
go back to reference Zhao D et al. Derivation and characterization of hepatic progenitor cells from human embryonic stem cells. PLoS One. 2009;4(7):e6468.PubMedCentralPubMed Zhao D et al. Derivation and characterization of hepatic progenitor cells from human embryonic stem cells. PLoS One. 2009;4(7):e6468.PubMedCentralPubMed
52.
go back to reference Li G, Satyamoorthy K, Herlyn M. N-cadherin-mediated intercellular interactions promote survival and migration of melanoma cells. Cancer Res. 2001;61(9):3819–25.PubMed Li G, Satyamoorthy K, Herlyn M. N-cadherin-mediated intercellular interactions promote survival and migration of melanoma cells. Cancer Res. 2001;61(9):3819–25.PubMed
53.
go back to reference Ramis-Conde I et al. Multi-scale modelling of cancer cell intravasation: the role of cadherins in metastasis. Phys Biol. 2009;6(1):016008.PubMed Ramis-Conde I et al. Multi-scale modelling of cancer cell intravasation: the role of cadherins in metastasis. Phys Biol. 2009;6(1):016008.PubMed
54.
go back to reference Takeichi M. The cadherins: cell-cell adhesion molecules controlling animal morphogenesis. Development. 1988;102(4):639–55.PubMed Takeichi M. The cadherins: cell-cell adhesion molecules controlling animal morphogenesis. Development. 1988;102(4):639–55.PubMed
55.
go back to reference Hazan RB et al. Exogenous expression of N-cadherin in breast cancer cells induces cell migration, invasion, and metastasis. J Cell Biol. 2000;148(4):779–90.PubMedCentralPubMed Hazan RB et al. Exogenous expression of N-cadherin in breast cancer cells induces cell migration, invasion, and metastasis. J Cell Biol. 2000;148(4):779–90.PubMedCentralPubMed
56.
go back to reference De Wever O et al. Critical role of N-cadherin in myofibroblast invasion and migration in vitro stimulated by colon-cancer-cell-derived TGF-β or wounding. J Cell Sci. 2004;117(20):4691–703.PubMed De Wever O et al. Critical role of N-cadherin in myofibroblast invasion and migration in vitro stimulated by colon-cancer-cell-derived TGF-β or wounding. J Cell Sci. 2004;117(20):4691–703.PubMed
57.
go back to reference Islam S et al. Expression of N-cadherin by human squamous carcinoma cells induces a scattered fibroblastic phenotype with disrupted cell-cell adhesion. J Cell Biol. 1996;135(6):1643–54.PubMed Islam S et al. Expression of N-cadherin by human squamous carcinoma cells induces a scattered fibroblastic phenotype with disrupted cell-cell adhesion. J Cell Biol. 1996;135(6):1643–54.PubMed
58.
go back to reference Tomita K et al. Cadherin switching in human prostate cancer progression. Cancer Res. 2000;60(13):3650–4.PubMed Tomita K et al. Cadherin switching in human prostate cancer progression. Cancer Res. 2000;60(13):3650–4.PubMed
59.
go back to reference Hsu M et al. Cadherin repertoire determines partner-specific gap junctional communication during melanoma progression. J Cell Sci. 2000;113(Pt 9):1535–42.PubMed Hsu M et al. Cadherin repertoire determines partner-specific gap junctional communication during melanoma progression. J Cell Sci. 2000;113(Pt 9):1535–42.PubMed
60.
go back to reference Shintani Y et al. Collagen I-mediated up-regulation of N-cadherin requires cooperative signals from integrins and discoidin domain receptor 1. J Cell Biol. 2008;180(6):1277–89.PubMedCentralPubMed Shintani Y et al. Collagen I-mediated up-regulation of N-cadherin requires cooperative signals from integrins and discoidin domain receptor 1. J Cell Biol. 2008;180(6):1277–89.PubMedCentralPubMed
61.
go back to reference Augustine CK et al. Targeting N-cadherin enhances antitumor activity of cytotoxic therapies in melanoma treatment. Cancer Res. 2008;68(10):3777–84.PubMed Augustine CK et al. Targeting N-cadherin enhances antitumor activity of cytotoxic therapies in melanoma treatment. Cancer Res. 2008;68(10):3777–84.PubMed
62.
go back to reference Beasley GM et al. A phase 1 study of systemic ADH-1 in combination with melphalan via isolated limb infusion in patients with locally advanced in-transit malignant melanoma. Cancer. 2009;115(20):4766–74.PubMed Beasley GM et al. A phase 1 study of systemic ADH-1 in combination with melphalan via isolated limb infusion in patients with locally advanced in-transit malignant melanoma. Cancer. 2009;115(20):4766–74.PubMed
63.
go back to reference Tanaka H et al. Monoclonal antibody targeting of N-cadherin inhibits prostate cancer growth, metastasis and castration resistance. Nat Med. 2010;16(12):1414–20.PubMedCentralPubMed Tanaka H et al. Monoclonal antibody targeting of N-cadherin inhibits prostate cancer growth, metastasis and castration resistance. Nat Med. 2010;16(12):1414–20.PubMedCentralPubMed
65.
go back to reference Pećina-Šlaus N. Tumor suppressor gene E-cadherin and its role in normal and malignant cells. Cancer Cell Int. 2003;3(1):17.PubMedCentralPubMed Pećina-Šlaus N. Tumor suppressor gene E-cadherin and its role in normal and malignant cells. Cancer Cell Int. 2003;3(1):17.PubMedCentralPubMed
67.
go back to reference Pinho SS et al. Modulation of E-cadherin function and dysfunction by N-glycosylation. Cell Mol Life Sci. 2011;68(6):1011–20.PubMed Pinho SS et al. Modulation of E-cadherin function and dysfunction by N-glycosylation. Cell Mol Life Sci. 2011;68(6):1011–20.PubMed
68.
go back to reference Van Roy F, Berx G. The cell-cell adhesion molecule E-cadherin. Cell Mol Life Sci. 2008;65(23):3756–88.PubMed Van Roy F, Berx G. The cell-cell adhesion molecule E-cadherin. Cell Mol Life Sci. 2008;65(23):3756–88.PubMed
69.
go back to reference Jiang W. E‐cadherin and its associated protein catenins, cancer invasion and metastasis. Br J Surg. 1996;83(4):437–46.PubMed Jiang W. E‐cadherin and its associated protein catenins, cancer invasion and metastasis. Br J Surg. 1996;83(4):437–46.PubMed
70.
go back to reference Larue L et al. A role for cadherins in tissue formation. Development. 1996;122(10):3185–94.PubMed Larue L et al. A role for cadherins in tissue formation. Development. 1996;122(10):3185–94.PubMed
71.
go back to reference Luo J, Lubaroff DM, Hendrix MJ. Suppression of prostate cancer invasive potential and matrix metalloproteinase activity by E-cadherin transfection. Cancer Res. 1999;59(15):3552–6.PubMed Luo J, Lubaroff DM, Hendrix MJ. Suppression of prostate cancer invasive potential and matrix metalloproteinase activity by E-cadherin transfection. Cancer Res. 1999;59(15):3552–6.PubMed
72.
go back to reference Hsu M-Y et al. E-cadherin expression in melanoma cells restores keratinocyte-mediated growth control and down-regulates expression of invasion-related adhesion receptors. Am J Pathol. 2000;156(5):1515–25.PubMedCentralPubMed Hsu M-Y et al. E-cadherin expression in melanoma cells restores keratinocyte-mediated growth control and down-regulates expression of invasion-related adhesion receptors. Am J Pathol. 2000;156(5):1515–25.PubMedCentralPubMed
73.
go back to reference Bogenrieder T, Herlyn M. Axis of evil: molecular mechanisms of cancer metastasis. Oncogene. 2003;22(42):6524–36.PubMed Bogenrieder T, Herlyn M. Axis of evil: molecular mechanisms of cancer metastasis. Oncogene. 2003;22(42):6524–36.PubMed
74.
go back to reference Vleminckx K et al. Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell. 1991;66(1):107–19.PubMed Vleminckx K et al. Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell. 1991;66(1):107–19.PubMed
75.
go back to reference Riethmacher D, Brinkmann V, Birchmeier C. A targeted mutation in the mouse E-cadherin gene results in defective preimplantation development. Proc Natl Acad Sci U S A. 1995;92(3):855–9.PubMedCentralPubMed Riethmacher D, Brinkmann V, Birchmeier C. A targeted mutation in the mouse E-cadherin gene results in defective preimplantation development. Proc Natl Acad Sci U S A. 1995;92(3):855–9.PubMedCentralPubMed
76.
go back to reference Hermiston ML, Wong MH, Gordon JI. Forced expression of E-cadherin in the mouse intestinal epithelium slows cell migration and provides evidence for nonautonomous regulation of cell fate in a self-renewing system. Genes Dev. 1996;10(8):985–96.PubMed Hermiston ML, Wong MH, Gordon JI. Forced expression of E-cadherin in the mouse intestinal epithelium slows cell migration and provides evidence for nonautonomous regulation of cell fate in a self-renewing system. Genes Dev. 1996;10(8):985–96.PubMed
77.
go back to reference Schmalhofer O, Brabletz S, Brabletz T. E-cadherin, β-catenin, and ZEB1 in malignant progression of cancer. Cancer Metastasis Rev. 2009;28(1–2):151–66.PubMed Schmalhofer O, Brabletz S, Brabletz T. E-cadherin, β-catenin, and ZEB1 in malignant progression of cancer. Cancer Metastasis Rev. 2009;28(1–2):151–66.PubMed
78.
go back to reference Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis 1. Endocr Rev. 2000;21(2):115–37.PubMed Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis 1. Endocr Rev. 2000;21(2):115–37.PubMed
79.
go back to reference Bennett J, Moffatt S, Horton M. Cell adhesion molecules in human osteoblasts: structure and function. 2001. Bennett J, Moffatt S, Horton M. Cell adhesion molecules in human osteoblasts: structure and function. 2001.
80.
go back to reference Ferrari SL et al. A role for N‐cadherin in the development of the differentiated osteoblastic phenotype. J Bone Miner Res. 2000;15(2):198–208.PubMed Ferrari SL et al. A role for N‐cadherin in the development of the differentiated osteoblastic phenotype. J Bone Miner Res. 2000;15(2):198–208.PubMed
81.
go back to reference Hippo Y et al. Differential gene expression profiles of scirrhous gastric cancer cells with high metastatic potential to peritoneum or lymph nodes. Cancer Res. 2001;61(3):889–95.PubMed Hippo Y et al. Differential gene expression profiles of scirrhous gastric cancer cells with high metastatic potential to peritoneum or lymph nodes. Cancer Res. 2001;61(3):889–95.PubMed
82.
go back to reference Rathinam R, Alahari SK. Important role of integrins in the cancer biology. Cancer Metastasis Rev. 2010;29(1):223–37.PubMed Rathinam R, Alahari SK. Important role of integrins in the cancer biology. Cancer Metastasis Rev. 2010;29(1):223–37.PubMed
83.
go back to reference Gahmberg CG et al. Regulation of integrin activity and signalling. Biochim Biophys Acta (BBA)-Gen Subj. 2009;1790(6):431–44. Gahmberg CG et al. Regulation of integrin activity and signalling. Biochim Biophys Acta (BBA)-Gen Subj. 2009;1790(6):431–44.
84.
go back to reference Plow EF et al. Ligand binding to integrins. J Biol Chem. 2000;275(29):21785–8.PubMed Plow EF et al. Ligand binding to integrins. J Biol Chem. 2000;275(29):21785–8.PubMed
85.
go back to reference Takagi J et al. Global conformational rearrangements in integrin extracellular domains in outside-in and inside-out signaling. Cell. 2002;110(5):599–611.PubMed Takagi J et al. Global conformational rearrangements in integrin extracellular domains in outside-in and inside-out signaling. Cell. 2002;110(5):599–611.PubMed
86.
87.
go back to reference Sastry SK, Burridge K. Focal adhesions: a nexus for intracellular signaling and cytoskeletal dynamics. Exp Cell Res. 2000;261(1):25–36.PubMed Sastry SK, Burridge K. Focal adhesions: a nexus for intracellular signaling and cytoskeletal dynamics. Exp Cell Res. 2000;261(1):25–36.PubMed
88.
go back to reference Berman A, Kozlova N, Morozevich G. Integrins: structure and signaling. Biochemistry (Moscow). 2003;68(12):1284–99. Berman A, Kozlova N, Morozevich G. Integrins: structure and signaling. Biochemistry (Moscow). 2003;68(12):1284–99.
90.
go back to reference Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell. 2002;110(6):673–87.PubMed Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell. 2002;110(6):673–87.PubMed
91.
92.
go back to reference White DE, Muller WJ. Multifaceted roles of integrins in breast cancer metastasis. J Mammary Gland Biol Neoplasia. 2007;12(2–3):135–42.PubMed White DE, Muller WJ. Multifaceted roles of integrins in breast cancer metastasis. J Mammary Gland Biol Neoplasia. 2007;12(2–3):135–42.PubMed
93.
go back to reference Janik ME, Lityńska A, Vereecken P. Cell migration—the role of integrin glycosylation. Biochim Biophys Acta (BBA)-Gen Subj. 2010;1800(6):545–55. Janik ME, Lityńska A, Vereecken P. Cell migration—the role of integrin glycosylation. Biochim Biophys Acta (BBA)-Gen Subj. 2010;1800(6):545–55.
94.
go back to reference Chung J, Kim TH. Integrin‐dependent translational control: implication in cancer progression. Microsc Res Tech. 2008;71(5):380–6.PubMed Chung J, Kim TH. Integrin‐dependent translational control: implication in cancer progression. Microsc Res Tech. 2008;71(5):380–6.PubMed
95.
go back to reference Schlaepfer DD, Jones K, Hunter T. Multiple Grb2-mediated integrin-stimulated signaling pathways to ERK2/mitogen-activated protein kinase: summation of both c-Src- and focal adhesion kinase-initiated tyrosine phosphorylation events. Mol Cell Biol. 1998;18(5):2571–85.PubMedCentralPubMed Schlaepfer DD, Jones K, Hunter T. Multiple Grb2-mediated integrin-stimulated signaling pathways to ERK2/mitogen-activated protein kinase: summation of both c-Src- and focal adhesion kinase-initiated tyrosine phosphorylation events. Mol Cell Biol. 1998;18(5):2571–85.PubMedCentralPubMed
96.
go back to reference Liapis H, Flath A, Kitazawa S. Integrin [alpha] v [beta] 3 expression by bone-residing breast cancer metastases. Diagn Mol Pathol. 1996;5(2):127–35.PubMed Liapis H, Flath A, Kitazawa S. Integrin [alpha] v [beta] 3 expression by bone-residing breast cancer metastases. Diagn Mol Pathol. 1996;5(2):127–35.PubMed
97.
go back to reference Pawelek JM, Chakraborty AK. Fusion of tumour cells with bone marrow-derived cells: a unifying explanation for metastasis. Nat Rev Cancer. 2008;8(5):377–86.PubMed Pawelek JM, Chakraborty AK. Fusion of tumour cells with bone marrow-derived cells: a unifying explanation for metastasis. Nat Rev Cancer. 2008;8(5):377–86.PubMed
98.
go back to reference Aumailley M et al. Altered synthesis of laminin 1 and absence of basement membrane component deposition in (beta) 1 integrin-deficient embryoid bodies. J Cell Sci. 2000;113(2):259–68.PubMed Aumailley M et al. Altered synthesis of laminin 1 and absence of basement membrane component deposition in (beta) 1 integrin-deficient embryoid bodies. J Cell Sci. 2000;113(2):259–68.PubMed
99.
go back to reference Brooks PC et al. Localization of matrix metalloproteinase MMP-2 to the surface of invasive cells by interaction with integrin αvβ3. Cell. 1996;85(5):683–93.PubMed Brooks PC et al. Localization of matrix metalloproteinase MMP-2 to the surface of invasive cells by interaction with integrin αvβ3. Cell. 1996;85(5):683–93.PubMed
100.
go back to reference Deryugina EI et al. Matrix metalloproteinase-2 activation modulates glioma cell migration. J Cell Sci. 1997;110(19):2473–82.PubMed Deryugina EI et al. Matrix metalloproteinase-2 activation modulates glioma cell migration. J Cell Sci. 1997;110(19):2473–82.PubMed
101.
go back to reference He Y et al. Interaction between cancer cells and stromal fibroblasts is required for activation of the uPAR-uPA-MMP-2 cascade in pancreatic cancer metastasis. Clin Cancer Res. 2007;13(11):3115–24.PubMed He Y et al. Interaction between cancer cells and stromal fibroblasts is required for activation of the uPAR-uPA-MMP-2 cascade in pancreatic cancer metastasis. Clin Cancer Res. 2007;13(11):3115–24.PubMed
102.
go back to reference Desgrosellier JS et al. An integrin αvβ3–c-Src oncogenic unit promotes anchorage-independence and tumor progression. Nat Med. 2009;15(10):1163–9.PubMedCentralPubMed Desgrosellier JS et al. An integrin αvβ3–c-Src oncogenic unit promotes anchorage-independence and tumor progression. Nat Med. 2009;15(10):1163–9.PubMedCentralPubMed
103.
go back to reference Wang Y et al. RGD-modified polymeric micelles as potential carriers for targeted delivery to integrin-overexpressing tumor vasculature and tumor cells. J Drug Targeting. 2009;17(6):459–67. Wang Y et al. RGD-modified polymeric micelles as potential carriers for targeted delivery to integrin-overexpressing tumor vasculature and tumor cells. J Drug Targeting. 2009;17(6):459–67.
104.
go back to reference Meyer T, Marshall J, Hart I. Expression of alphav integrins and vitronectin receptor identity in breast cancer cells. Br J Cancer. 1998;77(4):530.PubMedCentralPubMed Meyer T, Marshall J, Hart I. Expression of alphav integrins and vitronectin receptor identity in breast cancer cells. Br J Cancer. 1998;77(4):530.PubMedCentralPubMed
105.
go back to reference Rolli M et al. Activated integrin alphavbeta3 cooperates with metalloproteinase MMP-9 in regulating migration of metastatic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100(16):9482–7.PubMedCentralPubMed Rolli M et al. Activated integrin alphavbeta3 cooperates with metalloproteinase MMP-9 in regulating migration of metastatic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100(16):9482–7.PubMedCentralPubMed
106.
go back to reference Nieswandt B et al. Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Res. 1999;59(6):1295–300.PubMed Nieswandt B et al. Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Res. 1999;59(6):1295–300.PubMed
107.
go back to reference Tang BL, Ng EL. Rabs and cancer cell motility. Cell Motil Cytoskeleton. 2009;66(7):365–70.PubMed Tang BL, Ng EL. Rabs and cancer cell motility. Cell Motil Cytoskeleton. 2009;66(7):365–70.PubMed
108.
go back to reference Giancotti FG, Ruoslahti E. Integrin signaling. Science. 1999;285(5430):1028–33.PubMed Giancotti FG, Ruoslahti E. Integrin signaling. Science. 1999;285(5430):1028–33.PubMed
109.
go back to reference Morozevich GE et al. Implication of alpha5beta1 integrin in invasion of drug-resistant MCF-7/ADR breast carcinoma cells: a role for MMP-2 collagenase. Biochemistry (Mosc). 2008;73(7):791–6. Morozevich GE et al. Implication of alpha5beta1 integrin in invasion of drug-resistant MCF-7/ADR breast carcinoma cells: a role for MMP-2 collagenase. Biochemistry (Mosc). 2008;73(7):791–6.
110.
go back to reference Humphries MJ, Olden K, Yamada KM. A synthetic peptide from fibronectin inhibits experimental metastasis of murine melanoma cells. Science. 1986;233(4762):467–70.PubMed Humphries MJ, Olden K, Yamada KM. A synthetic peptide from fibronectin inhibits experimental metastasis of murine melanoma cells. Science. 1986;233(4762):467–70.PubMed
111.
go back to reference Hehlgans S, Haase M, Cordes N. Signalling via integrins: implications for cell survival and anticancer strategies. Biochim Biophys Acta. 2007;1775(1):163–80.PubMed Hehlgans S, Haase M, Cordes N. Signalling via integrins: implications for cell survival and anticancer strategies. Biochim Biophys Acta. 2007;1775(1):163–80.PubMed
112.
go back to reference Cox D, Brennan M, Moran N. Integrins as therapeutic targets: lessons and opportunities. Nat Rev Drug Discovery. 2010;9(10):804–20.PubMed Cox D, Brennan M, Moran N. Integrins as therapeutic targets: lessons and opportunities. Nat Rev Drug Discovery. 2010;9(10):804–20.PubMed
113.
go back to reference Desgrosellier JS, Cheresh DA. Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer. 2010;10(1):9–22.PubMedCentralPubMed Desgrosellier JS, Cheresh DA. Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer. 2010;10(1):9–22.PubMedCentralPubMed
114.
go back to reference Mullamitha SA et al. Phase I evaluation of a fully human anti-alphav integrin monoclonal antibody (CNTO 95) in patients with advanced solid tumors. Clin Cancer Res. 2007;13(7):2128–35.PubMed Mullamitha SA et al. Phase I evaluation of a fully human anti-alphav integrin monoclonal antibody (CNTO 95) in patients with advanced solid tumors. Clin Cancer Res. 2007;13(7):2128–35.PubMed
115.
go back to reference Nabors LB et al. Phase I and correlative biology study of cilengitide in patients with recurrent malignant glioma. J Clin Oncol. 2007;25(13):1651–7.PubMed Nabors LB et al. Phase I and correlative biology study of cilengitide in patients with recurrent malignant glioma. J Clin Oncol. 2007;25(13):1651–7.PubMed
116.
go back to reference Reardon DA et al. Randomized phase II study of cilengitide, an integrin-targeting arginine-glycine-aspartic acid peptide, in recurrent glioblastoma multiforme. J Clin Oncol. 2008;26(34):5610–7.PubMed Reardon DA et al. Randomized phase II study of cilengitide, an integrin-targeting arginine-glycine-aspartic acid peptide, in recurrent glioblastoma multiforme. J Clin Oncol. 2008;26(34):5610–7.PubMed
117.
go back to reference MacDonald TJ et al. Phase I clinical trial of cilengitide in children with refractory brain tumors: Pediatric Brain Tumor Consortium Study PBTC-012. J Clin Oncol. 2008;26(6):919–24.PubMed MacDonald TJ et al. Phase I clinical trial of cilengitide in children with refractory brain tumors: Pediatric Brain Tumor Consortium Study PBTC-012. J Clin Oncol. 2008;26(6):919–24.PubMed
118.
go back to reference Ricart AD et al. Volociximab, a chimeric monoclonal antibody that specifically binds alpha5beta1 integrin: a phase I, pharmacokinetic, and biological correlative study. Clin Cancer Res. 2008;14(23):7924–9.PubMedCentralPubMed Ricart AD et al. Volociximab, a chimeric monoclonal antibody that specifically binds alpha5beta1 integrin: a phase I, pharmacokinetic, and biological correlative study. Clin Cancer Res. 2008;14(23):7924–9.PubMedCentralPubMed
119.
go back to reference Danen EH et al. Requirement for the synergy site for cell adhesion to fibronectin depends on the activation state of integrin alpha 5 beta 1. J Biol Chem. 1995;270(37):21612–8.PubMed Danen EH et al. Requirement for the synergy site for cell adhesion to fibronectin depends on the activation state of integrin alpha 5 beta 1. J Biol Chem. 1995;270(37):21612–8.PubMed
120.
go back to reference Khalili P et al. A non-RGD-based integrin binding peptide (ATN-161) blocks breast cancer growth and metastasis in vivo. Mol Cancer Ther. 2006;5(9):2271–80.PubMed Khalili P et al. A non-RGD-based integrin binding peptide (ATN-161) blocks breast cancer growth and metastasis in vivo. Mol Cancer Ther. 2006;5(9):2271–80.PubMed
121.
go back to reference Cianfrocca ME et al. Phase 1 trial of the antiangiogenic peptide ATN-161 (Ac-PHSCN-NH(2)), a beta integrin antagonist, in patients with solid tumours. Br J Cancer. 2006;94(11):1621–6.PubMedCentralPubMed Cianfrocca ME et al. Phase 1 trial of the antiangiogenic peptide ATN-161 (Ac-PHSCN-NH(2)), a beta integrin antagonist, in patients with solid tumours. Br J Cancer. 2006;94(11):1621–6.PubMedCentralPubMed
122.
go back to reference Mulgrew K et al. Direct targeting of alphavbeta3 integrin on tumor cells with a monoclonal antibody. Abegrin Mol Cancer Ther. 2006;5(12):3122–9.PubMed Mulgrew K et al. Direct targeting of alphavbeta3 integrin on tumor cells with a monoclonal antibody. Abegrin Mol Cancer Ther. 2006;5(12):3122–9.PubMed
123.
go back to reference Gutheil JC et al. Targeted antiangiogenic therapy for cancer using Vitaxin: a humanized monoclonal antibody to the integrin alphavbeta3. Clin Cancer Res. 2000;6(8):3056–61.PubMed Gutheil JC et al. Targeted antiangiogenic therapy for cancer using Vitaxin: a humanized monoclonal antibody to the integrin alphavbeta3. Clin Cancer Res. 2000;6(8):3056–61.PubMed
124.
go back to reference McNeel DG et al. Phase I trial of a monoclonal antibody specific for alphavbeta3 integrin (MEDI-522) in patients with advanced malignancies, including an assessment of effect on tumor perfusion. Clin Cancer Res. 2005;11(21):7851–60.PubMed McNeel DG et al. Phase I trial of a monoclonal antibody specific for alphavbeta3 integrin (MEDI-522) in patients with advanced malignancies, including an assessment of effect on tumor perfusion. Clin Cancer Res. 2005;11(21):7851–60.PubMed
125.
go back to reference Hersey P et al. Phase I/II study of immunotherapy with T-cell peptide epitopes in patients with stage IV melanoma. Cancer Immunol Immunother. 2005;54(3):208–18.PubMed Hersey P et al. Phase I/II study of immunotherapy with T-cell peptide epitopes in patients with stage IV melanoma. Cancer Immunol Immunother. 2005;54(3):208–18.PubMed
126.
go back to reference Trikha M et al. CNTO 95, a fully human monoclonal antibody that inhibits alphav integrins, has antitumor and antiangiogenic activity in vivo. Int J Cancer. 2004;110(3):326–35.PubMed Trikha M et al. CNTO 95, a fully human monoclonal antibody that inhibits alphav integrins, has antitumor and antiangiogenic activity in vivo. Int J Cancer. 2004;110(3):326–35.PubMed
127.
go back to reference Chen Q et al. CNTO 95, a fully human anti alphav integrin antibody, inhibits cell signaling, migration, invasion, and spontaneous metastasis of human breast cancer cells. Clin Exp Metastasis. 2008;25(2):139–48.PubMed Chen Q et al. CNTO 95, a fully human anti alphav integrin antibody, inhibits cell signaling, migration, invasion, and spontaneous metastasis of human breast cancer cells. Clin Exp Metastasis. 2008;25(2):139–48.PubMed
128.
go back to reference Martin PL et al. Reviews preclinical safety and immune-modulating effects of therapeutic monoclonal antibodies to interleukin-6 and tumor necrosis factor-alpha in cynomolgus macaques. J Immunotoxicol. 2005;1(3):131–9.PubMed Martin PL et al. Reviews preclinical safety and immune-modulating effects of therapeutic monoclonal antibodies to interleukin-6 and tumor necrosis factor-alpha in cynomolgus macaques. J Immunotoxicol. 2005;1(3):131–9.PubMed
129.
go back to reference Ley K. The role of selectins in inflammation and disease. Trends Mol Med. 2003;9(6):263–8.PubMed Ley K. The role of selectins in inflammation and disease. Trends Mol Med. 2003;9(6):263–8.PubMed
130.
go back to reference McEver RP. Selectins: lectins that initiate cell adhesion under flow. Curr Opin Cell Biol. 2002;14(5):581–6.PubMed McEver RP. Selectins: lectins that initiate cell adhesion under flow. Curr Opin Cell Biol. 2002;14(5):581–6.PubMed
131.
go back to reference Läubli H. and Borsig.L., Selectins promote tumor metastasis. in Seminars in cancer biology. 2010. Elsevier. Läubli H. and Borsig.L., Selectins promote tumor metastasis. in Seminars in cancer biology. 2010. Elsevier.
132.
go back to reference Kansas GS. Selectins and their ligands: current concepts and controversies. Blood. 1996;88(9):3259–87.PubMed Kansas GS. Selectins and their ligands: current concepts and controversies. Blood. 1996;88(9):3259–87.PubMed
133.
go back to reference Rosen SD. Ligands for L-selectin: homing, inflammation, and beyond. Annu Rev Immunol. 2004;22:129–56.PubMed Rosen SD. Ligands for L-selectin: homing, inflammation, and beyond. Annu Rev Immunol. 2004;22:129–56.PubMed
134.
go back to reference Lowe JB. Glycan-dependent leukocyte adhesion and recruitment in inflammation. Curr Opin Cell Biol. 2003;15(5):531–8.PubMed Lowe JB. Glycan-dependent leukocyte adhesion and recruitment in inflammation. Curr Opin Cell Biol. 2003;15(5):531–8.PubMed
135.
go back to reference Bendas G, Borsig L. Cancer cell adhesion and metastasis: selectins, integrins, and the inhibitory potential of heparins. Int J Cell Biol. 2012;2012:1–10. Bendas G, Borsig L. Cancer cell adhesion and metastasis: selectins, integrins, and the inhibitory potential of heparins. Int J Cell Biol. 2012;2012:1–10.
136.
go back to reference Sperandio M. Selectins and glycosyltransferases in leukocyte rolling in vivo. Febs J. 2006;273(19):4377–89.PubMed Sperandio M. Selectins and glycosyltransferases in leukocyte rolling in vivo. Febs J. 2006;273(19):4377–89.PubMed
137.
go back to reference Köhler S et al. E-/P-selectins and colon carcinoma metastasis: first in vivo evidence for their crucial role in a clinically relevant model of spontaneous metastasis formation in the lung. Br J Cancer. 2009;102(3):602–9.PubMedCentralPubMed Köhler S et al. E-/P-selectins and colon carcinoma metastasis: first in vivo evidence for their crucial role in a clinically relevant model of spontaneous metastasis formation in the lung. Br J Cancer. 2009;102(3):602–9.PubMedCentralPubMed
138.
go back to reference Ley K et al. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol. 2007;7(9):678–89.PubMed Ley K et al. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol. 2007;7(9):678–89.PubMed
139.
go back to reference Sallusto F et al. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401(6754):708–12.PubMed Sallusto F et al. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401(6754):708–12.PubMed
140.
go back to reference Sipkins DA et al. In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment. Nature. 2005;435(7044):969–73.PubMedCentralPubMed Sipkins DA et al. In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment. Nature. 2005;435(7044):969–73.PubMedCentralPubMed
141.
go back to reference Mannori G et al. Differential colon cancer cell adhesion to E-, P-, and L-selectin: role of mucin-type glycoproteins. Cancer Res. 1995;55(19):4425–31.PubMed Mannori G et al. Differential colon cancer cell adhesion to E-, P-, and L-selectin: role of mucin-type glycoproteins. Cancer Res. 1995;55(19):4425–31.PubMed
142.
go back to reference Witz IP. The selectin–selectin ligand axis in tumor progression. Cancer Metastasis Rev. 2008;27(1):19–30.PubMed Witz IP. The selectin–selectin ligand axis in tumor progression. Cancer Metastasis Rev. 2008;27(1):19–30.PubMed
143.
go back to reference Läubli H, Spanaus K-S, Borsig L. Selectin-mediated activation of endothelial cells induces expression of CCL5 and promotes metastasis through recruitment of monocytes. Blood. 2009;114(20):4583–91.PubMed Läubli H, Spanaus K-S, Borsig L. Selectin-mediated activation of endothelial cells induces expression of CCL5 and promotes metastasis through recruitment of monocytes. Blood. 2009;114(20):4583–91.PubMed
144.
go back to reference Barthel SR et al. Targeting selectins and selectin ligands in inflammation and cancer. Expert Opin Ther Targets. 2007;11(11):1473–91.PubMedCentralPubMed Barthel SR et al. Targeting selectins and selectin ligands in inflammation and cancer. Expert Opin Ther Targets. 2007;11(11):1473–91.PubMedCentralPubMed
145.
go back to reference Preusser M et al. Brain metastases: pathobiology and emerging targeted therapies. Acta Neuropathologica. 2012;123(2):205–22.PubMed Preusser M et al. Brain metastases: pathobiology and emerging targeted therapies. Acta Neuropathologica. 2012;123(2):205–22.PubMed
146.
go back to reference Crockett-Torabi E. Selectins and mechanisms of signal transduction. J Leukocyte Biol. 1998;63(1):1–14.PubMed Crockett-Torabi E. Selectins and mechanisms of signal transduction. J Leukocyte Biol. 1998;63(1):1–14.PubMed
147.
go back to reference Burdick MM et al. HCELL is the major E- and L-selectin ligand expressed on LS174T colon carcinoma cells. J Biol Chem. 2006;281(20):13899–905.PubMed Burdick MM et al. HCELL is the major E- and L-selectin ligand expressed on LS174T colon carcinoma cells. J Biol Chem. 2006;281(20):13899–905.PubMed
148.
go back to reference Gout S et al. Death receptor-3, a new E-Selectin counter-receptor that confers migration and survival advantages to colon carcinoma cells by triggering p38 and ERK MAPK activation. Cancer Res. 2006;66(18):9117–24.PubMed Gout S et al. Death receptor-3, a new E-Selectin counter-receptor that confers migration and survival advantages to colon carcinoma cells by triggering p38 and ERK MAPK activation. Cancer Res. 2006;66(18):9117–24.PubMed
149.
go back to reference Aychek T et al. E‐selectin regulates gene expression in metastatic colorectal carcinoma cells and enhances HMGB1 release. Int J Cancer. 2008;123(8):1741–50.PubMed Aychek T et al. E‐selectin regulates gene expression in metastatic colorectal carcinoma cells and enhances HMGB1 release. Int J Cancer. 2008;123(8):1741–50.PubMed
150.
go back to reference Ludwig RJ et al. Endothelial P-selectin as a target of heparin action in experimental melanoma lung metastasis. Cancer Res. 2004;64(8):2743–50.PubMed Ludwig RJ et al. Endothelial P-selectin as a target of heparin action in experimental melanoma lung metastasis. Cancer Res. 2004;64(8):2743–50.PubMed
151.
go back to reference Borsig L et al. Sulfated hexasaccharides attenuate metastasis by inhibition of P-selectin and heparanase. Neoplasia. 2011;13(5):445–52.PubMedCentralPubMed Borsig L et al. Sulfated hexasaccharides attenuate metastasis by inhibition of P-selectin and heparanase. Neoplasia. 2011;13(5):445–52.PubMedCentralPubMed
152.
go back to reference Soroka, V., Kasper C., and Poulsen F.M. Structural biology of NCAM, in structure and function of the neural cell adhesion molecule NCAM. 2010, Springer. p. 3-22. Soroka, V., Kasper C., and Poulsen F.M. Structural biology of NCAM, in structure and function of the neural cell adhesion molecule NCAM. 2010, Springer. p. 3-22.
153.
go back to reference Wai Wong C, Dye DE, Coombe DR. The role of immunoglobulin superfamily cell dhesion molecules in cancer metastasis. Int J Cell Biol. 2012;2012:340296.PubMedCentralPubMed Wai Wong C, Dye DE, Coombe DR. The role of immunoglobulin superfamily cell dhesion molecules in cancer metastasis. Int J Cell Biol. 2012;2012:340296.PubMedCentralPubMed
154.
go back to reference Barclay A.N. Membrane proteins with immunoglobulin-like domains—a master superfamily of interaction molecules. in Seminars in immunology. 2003. Elsevier. Barclay A.N. Membrane proteins with immunoglobulin-like domains—a master superfamily of interaction molecules. in Seminars in immunology. 2003. Elsevier.
155.
go back to reference Maddaluno L et al. The adhesion molecule L1 regulates transendothelial migration and trafficking of dendritic cells. J Exp Med. 2009;206(3):623–35.PubMedCentralPubMed Maddaluno L et al. The adhesion molecule L1 regulates transendothelial migration and trafficking of dendritic cells. J Exp Med. 2009;206(3):623–35.PubMedCentralPubMed
156.
go back to reference Garrido-Urbani S et al. Vascular and epithelial junctions: a barrier for leucocyte migration. Biochem Soc Trans. 2008;36(2):203–12.PubMed Garrido-Urbani S et al. Vascular and epithelial junctions: a barrier for leucocyte migration. Biochem Soc Trans. 2008;36(2):203–12.PubMed
157.
go back to reference Francavilla C., Maddaluno L., and Cavallaro U. The functional role of cell adhesion molecules in tumor angiogenesis. in Seminars in cancer biology. 2009. Elsevier. Francavilla C., Maddaluno L., and Cavallaro U. The functional role of cell adhesion molecules in tumor angiogenesis. in Seminars in cancer biology. 2009. Elsevier.
158.
go back to reference Johnson JP et al. Melanoma progression-associated glycoprotein MUC18/MCAM mediates homotypic cell adhesion through interaction with a heterophilic ligand. Int J Cancer. 1997;73(5):769–74.PubMed Johnson JP et al. Melanoma progression-associated glycoprotein MUC18/MCAM mediates homotypic cell adhesion through interaction with a heterophilic ligand. Int J Cancer. 1997;73(5):769–74.PubMed
159.
go back to reference Wu G-J et al. Enforced expression of METCAM/MUC18 increases tumorigenesis of human prostate cancer LNCaP cells in nude mice. J Urol. 2011;185(4):1504–12.PubMed Wu G-J et al. Enforced expression of METCAM/MUC18 increases tumorigenesis of human prostate cancer LNCaP cells in nude mice. J Urol. 2011;185(4):1504–12.PubMed
160.
go back to reference Zeng G-F, Cai S-X, Wu G-J. Up-regulation of METCAM/MUC18 promotes motility, invasion, and tumorigenesis of human breast cancer cells. BMC Cancer. 2011;11(1):113.PubMedCentralPubMed Zeng G-F, Cai S-X, Wu G-J. Up-regulation of METCAM/MUC18 promotes motility, invasion, and tumorigenesis of human breast cancer cells. BMC Cancer. 2011;11(1):113.PubMedCentralPubMed
161.
go back to reference Roland CL et al. ICAM-1 expression determines malignant potential of cancer. Surgery. 2007;141(6):705–7.PubMed Roland CL et al. ICAM-1 expression determines malignant potential of cancer. Surgery. 2007;141(6):705–7.PubMed
162.
go back to reference Siesser PF, Maness PF. L1 cell adhesion molecules as regulators of tumor cell invasiveness. Cell Adh Migr. 2009;3(3):275–7.PubMedCentralPubMed Siesser PF, Maness PF. L1 cell adhesion molecules as regulators of tumor cell invasiveness. Cell Adh Migr. 2009;3(3):275–7.PubMedCentralPubMed
163.
go back to reference Jezierska A, Matysiak W, Motyl T. ALCAM/CD166 protects breast cancer cells against apoptosis and autophagy. Med Sci Monit Basic Res. 2006;12(8):BR263–73. Jezierska A, Matysiak W, Motyl T. ALCAM/CD166 protects breast cancer cells against apoptosis and autophagy. Med Sci Monit Basic Res. 2006;12(8):BR263–73.
164.
go back to reference Deng C et al. Angiogenic effect of intercellular adhesion molecule-1. J Huazhong Univ Sci Technol. 2007;27:9–12. Deng C et al. Angiogenic effect of intercellular adhesion molecule-1. J Huazhong Univ Sci Technol. 2007;27:9–12.
165.
go back to reference Kevil CG et al. Intercellular adhesion molecule-1 (ICAM-1) regulates endothelial cell motility through a nitric oxide-dependent pathway. J Biol Chem. 2004;279(18):19230–8.PubMed Kevil CG et al. Intercellular adhesion molecule-1 (ICAM-1) regulates endothelial cell motility through a nitric oxide-dependent pathway. J Biol Chem. 2004;279(18):19230–8.PubMed
166.
go back to reference Gratzinger D, Barreuther M, Madri JA. Platelet–endothelial cell adhesion molecule-1 modulates endothelial migration through its immunoreceptor tyrosine-based inhibitory motif. Biochem Biophys Res Commun. 2003;301(1):243–9.PubMed Gratzinger D, Barreuther M, Madri JA. Platelet–endothelial cell adhesion molecule-1 modulates endothelial migration through its immunoreceptor tyrosine-based inhibitory motif. Biochem Biophys Res Commun. 2003;301(1):243–9.PubMed
167.
go back to reference Park S et al. PECAM-1 regulates proangiogenic properties of endothelial cells through modulation of cell-cell and cell-matrix interactions. Am J Physiol-Cell Physiol. 2010;299(6):C1468–84.PubMedCentralPubMed Park S et al. PECAM-1 regulates proangiogenic properties of endothelial cells through modulation of cell-cell and cell-matrix interactions. Am J Physiol-Cell Physiol. 2010;299(6):C1468–84.PubMedCentralPubMed
168.
go back to reference Gavert N et al. L1-CAM in cancerous tissues. Expert Opin Biol Ther. 2008;8(11):1749–57.PubMed Gavert N et al. L1-CAM in cancerous tissues. Expert Opin Biol Ther. 2008;8(11):1749–57.PubMed
169.
go back to reference Friedl P, Gilmour D. Collective cell migration in morphogenesis, regeneration and cancer. Nat Rev Mol Cell Biol. 2009;10(7):445–57.PubMed Friedl P, Gilmour D. Collective cell migration in morphogenesis, regeneration and cancer. Nat Rev Mol Cell Biol. 2009;10(7):445–57.PubMed
170.
go back to reference Orian-Rousseau V. CD44, a therapeutic target for metastasising tumours. Eur J Cancer. 2010;46(7):1271–7.PubMed Orian-Rousseau V. CD44, a therapeutic target for metastasising tumours. Eur J Cancer. 2010;46(7):1271–7.PubMed
171.
go back to reference Fox SB et al. Normal human tissues, in addition to some tumors, express multiple different CD44 isoforms. Cancer Res. 1994;54(16):4539–46.PubMed Fox SB et al. Normal human tissues, in addition to some tumors, express multiple different CD44 isoforms. Cancer Res. 1994;54(16):4539–46.PubMed
172.
go back to reference Richter U et al. The interaction between CD44 on tumour cells and hyaluronan under physiologic flow conditions: implications for metastasis formation. Histochem Cell Biol. 2012;137(5):687–95.PubMed Richter U et al. The interaction between CD44 on tumour cells and hyaluronan under physiologic flow conditions: implications for metastasis formation. Histochem Cell Biol. 2012;137(5):687–95.PubMed
173.
go back to reference UNDERHILL C. CD44: the hyaluronan receptor. J Cell Sci. 1992;103(2):293–8.PubMed UNDERHILL C. CD44: the hyaluronan receptor. J Cell Sci. 1992;103(2):293–8.PubMed
174.
go back to reference Lesley J, Hyman R, Kincade PW. CD44 and its interaction with extracellular matrix. Adv Immunol. 1993;54:271–335.PubMed Lesley J, Hyman R, Kincade PW. CD44 and its interaction with extracellular matrix. Adv Immunol. 1993;54:271–335.PubMed
175.
go back to reference Ponta H, Sherman L, Herrlich PA. CD44: from adhesion molecules to signalling regulators. Nat Rev Mol Cell Biol. 2003;4(1):33–45.PubMed Ponta H, Sherman L, Herrlich PA. CD44: from adhesion molecules to signalling regulators. Nat Rev Mol Cell Biol. 2003;4(1):33–45.PubMed
176.
go back to reference Ishii S et al. CD44 participates in the adhesion of human colorectal carcinoma cells to laminin and type IV collagen. Surg Oncol. 1993;2(4):255–64.PubMed Ishii S et al. CD44 participates in the adhesion of human colorectal carcinoma cells to laminin and type IV collagen. Surg Oncol. 1993;2(4):255–64.PubMed
177.
go back to reference Jalkanen S, Jalkanen M. Lymphocyte CD44 binds the COOH-terminal heparin-binding domain of fibronectin. J Cell Biol. 1992;116(3):817–25.PubMed Jalkanen S, Jalkanen M. Lymphocyte CD44 binds the COOH-terminal heparin-binding domain of fibronectin. J Cell Biol. 1992;116(3):817–25.PubMed
178.
go back to reference Marhaba R, Zöller M. CD44 in cancer progression: adhesion, migration and growth regulation. J Mol Histol. 2004;35(3):211–31.PubMed Marhaba R, Zöller M. CD44 in cancer progression: adhesion, migration and growth regulation. J Mol Histol. 2004;35(3):211–31.PubMed
179.
go back to reference Legg JW et al. A novel PKC-regulated mechanism controls CD44–ezrin association and directional cell motility. Nat Cell Biol. 2002;4(6):399–407.PubMed Legg JW et al. A novel PKC-regulated mechanism controls CD44–ezrin association and directional cell motility. Nat Cell Biol. 2002;4(6):399–407.PubMed
180.
go back to reference Lokeshwar V, Bourguignon L. Post-translational protein modification and expression of ankyrin-binding site (s) in GP85 (Pgp-1/CD44) and its biosynthetic precursors during T-lymphoma membrane biosynthesis. J Biol Chem. 1991;266(27):17983–9.PubMed Lokeshwar V, Bourguignon L. Post-translational protein modification and expression of ankyrin-binding site (s) in GP85 (Pgp-1/CD44) and its biosynthetic precursors during T-lymphoma membrane biosynthesis. J Biol Chem. 1991;266(27):17983–9.PubMed
181.
go back to reference Iczkowski KA. Cell adhesion molecule CD44: its functional roles in prostate cancer. Am J Transl Res. 2011;3(1):1.PubMedCentral Iczkowski KA. Cell adhesion molecule CD44: its functional roles in prostate cancer. Am J Transl Res. 2011;3(1):1.PubMedCentral
182.
go back to reference Shimizu Y et al. Dual role of the CD44 molecule in T cell adhesion and activation. J Immunol. 1989;143(8):2457–63.PubMed Shimizu Y et al. Dual role of the CD44 molecule in T cell adhesion and activation. J Immunol. 1989;143(8):2457–63.PubMed
183.
go back to reference Trochon V et al. Evidence of involvement of CD44 in endothelial cell proliferation, migration and angiogenesis in vitro. Int J Cancer. 1996;66(5):664–8.PubMed Trochon V et al. Evidence of involvement of CD44 in endothelial cell proliferation, migration and angiogenesis in vitro. Int J Cancer. 1996;66(5):664–8.PubMed
184.
go back to reference Webb D et al. LFA-3, CD44, and CD45: physiologic triggers of human monocyte TNF and IL-1 release. Science. 1990;249(4974):1295–7.PubMed Webb D et al. LFA-3, CD44, and CD45: physiologic triggers of human monocyte TNF and IL-1 release. Science. 1990;249(4974):1295–7.PubMed
185.
go back to reference Aruffo A et al. CD44 is the principal cell surface receptor for hyaluronate. Cell. 1990;61(7):1303–13.PubMed Aruffo A et al. CD44 is the principal cell surface receptor for hyaluronate. Cell. 1990;61(7):1303–13.PubMed
186.
go back to reference Naor D, Sionov RV, Ish-Shalom D. CD44: structure, function and association with the malignant process. Adv Cancer Res. 1997;71:241–319.PubMed Naor D, Sionov RV, Ish-Shalom D. CD44: structure, function and association with the malignant process. Adv Cancer Res. 1997;71:241–319.PubMed
187.
go back to reference Naor D et al. CD44 in cancer. Crit Rev Clin Lab Sci. 2002;39(6):527–79.PubMed Naor D et al. CD44 in cancer. Crit Rev Clin Lab Sci. 2002;39(6):527–79.PubMed
188.
go back to reference Wielenga VJ et al. CD44 glycoproteins in colorectal cancer: expression, function, and prognostic value. Adv Cancer Res. 1999;77:169–87. Wielenga VJ et al. CD44 glycoproteins in colorectal cancer: expression, function, and prognostic value. Adv Cancer Res. 1999;77:169–87.
189.
go back to reference Hsieh H et al. Molecular studies into the role of CD44 variants in metastasis in gastric cancer. Mol Pathol. 1999;52(1):25.PubMedCentralPubMed Hsieh H et al. Molecular studies into the role of CD44 variants in metastasis in gastric cancer. Mol Pathol. 1999;52(1):25.PubMedCentralPubMed
190.
go back to reference Seiter S et al. Prevention of tumor metastasis formation by anti-variant CD44. J Exp Med. 1993;177(2):443–55.PubMed Seiter S et al. Prevention of tumor metastasis formation by anti-variant CD44. J Exp Med. 1993;177(2):443–55.PubMed
191.
go back to reference Ayhan A, Tok EC, Bildirici I. Overexpression of CD44 variant 6 in human endometrial cancer and its prognostic significance. Gynecol Oncol. 2001;80(3):355–8.PubMed Ayhan A, Tok EC, Bildirici I. Overexpression of CD44 variant 6 in human endometrial cancer and its prognostic significance. Gynecol Oncol. 2001;80(3):355–8.PubMed
192.
go back to reference Tijink BM et al. A phase I dose escalation study with anti-CD44v6 bivatuzumab mertansine in patients with incurable squamous cell carcinoma of the head and neck or esophagus. Clin Cancer Res. 2006;12(20 Pt 1):6064–72.PubMed Tijink BM et al. A phase I dose escalation study with anti-CD44v6 bivatuzumab mertansine in patients with incurable squamous cell carcinoma of the head and neck or esophagus. Clin Cancer Res. 2006;12(20 Pt 1):6064–72.PubMed
193.
go back to reference Orian-Rousseau V, Ponta H. Adhesion proteins meet receptors: a common theme? Adv Cancer Res. 2008;101:63–92.PubMed Orian-Rousseau V, Ponta H. Adhesion proteins meet receptors: a common theme? Adv Cancer Res. 2008;101:63–92.PubMed
194.
go back to reference Liu C et al. The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med. 2011;17(2):211–5.PubMedCentralPubMed Liu C et al. The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med. 2011;17(2):211–5.PubMedCentralPubMed
195.
go back to reference Sahai E. Illuminating the metastatic process. Nat Rev Cancer. 2007;7(10):737–49.PubMed Sahai E. Illuminating the metastatic process. Nat Rev Cancer. 2007;7(10):737–49.PubMed
196.
go back to reference Friedl P, Alexander S. Cancer invasion and the microenvironment: plasticity and reciprocity. Cell. 2011;147(5):992–1009.PubMed Friedl P, Alexander S. Cancer invasion and the microenvironment: plasticity and reciprocity. Cell. 2011;147(5):992–1009.PubMed
197.
198.
go back to reference Friedl P. Prespecification and plasticity: shifting mechanisms of cell migration. Curr Opin Cell Biol. 2004;16(1):14–23.PubMed Friedl P. Prespecification and plasticity: shifting mechanisms of cell migration. Curr Opin Cell Biol. 2004;16(1):14–23.PubMed
199.
go back to reference Yilmaz M, Christofori G, Lehembre F. Distinct mechanisms of tumor invasion and metastasis. Trends Mol Med. 2007;13(12):535–41.PubMed Yilmaz M, Christofori G, Lehembre F. Distinct mechanisms of tumor invasion and metastasis. Trends Mol Med. 2007;13(12):535–41.PubMed
200.
go back to reference Gray RS, Cheung KJ, Ewald AJ. Cellular mechanisms regulating epithelial morphogenesis and cancer invasion. Curr Opin Cell Biol. 2010;22(5):640–50.PubMedCentralPubMed Gray RS, Cheung KJ, Ewald AJ. Cellular mechanisms regulating epithelial morphogenesis and cancer invasion. Curr Opin Cell Biol. 2010;22(5):640–50.PubMedCentralPubMed
201.
go back to reference Gaggioli C et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat Cell Biol. 2007;9(12):1392–400.PubMed Gaggioli C et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat Cell Biol. 2007;9(12):1392–400.PubMed
202.
go back to reference Condeelis J, Segall JE. Intravital imaging of cell movement in tumours. Nat Rev Cancer. 2003;3(12):921–30.PubMed Condeelis J, Segall JE. Intravital imaging of cell movement in tumours. Nat Rev Cancer. 2003;3(12):921–30.PubMed
203.
go back to reference Mandeville J, Lawson MA, Maxfield FR. Dynamic imaging of neutrophil migration in three dimensions: mechanical interactions between cells and matrix. J Leukocyte Biol. 1997;61(2):188–200.PubMed Mandeville J, Lawson MA, Maxfield FR. Dynamic imaging of neutrophil migration in three dimensions: mechanical interactions between cells and matrix. J Leukocyte Biol. 1997;61(2):188–200.PubMed
204.
go back to reference Sahai E, Marshall CJ. Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nat Cell Biol. 2003;5(8):711–9.PubMed Sahai E, Marshall CJ. Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nat Cell Biol. 2003;5(8):711–9.PubMed
205.
go back to reference Rösel D et al. Up-regulation of Rho/ROCK signaling in sarcoma cells drives invasion and increased generation of protrusive forces. Mol Cancer Res. 2008;6(9):1410–20.PubMed Rösel D et al. Up-regulation of Rho/ROCK signaling in sarcoma cells drives invasion and increased generation of protrusive forces. Mol Cancer Res. 2008;6(9):1410–20.PubMed
206.
go back to reference Micuda S et al. ROCK inhibitors as emerging therapeutic candidates for sarcomas. Curr Cancer Drug Targets. 2010;10(2):127–34.PubMed Micuda S et al. ROCK inhibitors as emerging therapeutic candidates for sarcomas. Curr Cancer Drug Targets. 2010;10(2):127–34.PubMed
207.
go back to reference Friedl P, Wolf K. Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer. 2003;3(5):362–74.PubMed Friedl P, Wolf K. Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer. 2003;3(5):362–74.PubMed
208.
go back to reference Wyckoff JB et al. ROCK- and myosin-dependent matrix deformation enables protease-independent tumor-cell invasion in vivo. Curr Biol. 2006;16(15):1515–23.PubMed Wyckoff JB et al. ROCK- and myosin-dependent matrix deformation enables protease-independent tumor-cell invasion in vivo. Curr Biol. 2006;16(15):1515–23.PubMed
209.
go back to reference Sabeh F, Shimizu-Hirota R, Weiss SJ. Protease-dependent versus -independent cancer cell invasion programs: three-dimensional amoeboid movement revisited. J Cell Biol. 2009;185(1):11–9.PubMedCentralPubMed Sabeh F, Shimizu-Hirota R, Weiss SJ. Protease-dependent versus -independent cancer cell invasion programs: three-dimensional amoeboid movement revisited. J Cell Biol. 2009;185(1):11–9.PubMedCentralPubMed
210.
go back to reference Turk B. Targeting proteases: successes, failures and future prospects. Nat Rev Drug Discovery. 2006;5(9):785–99.PubMed Turk B. Targeting proteases: successes, failures and future prospects. Nat Rev Drug Discovery. 2006;5(9):785–99.PubMed
211.
212.
go back to reference Boy R.G., et al. Enzymes/transporters, in Molecular imaging II. 2008, Springer. p. 131-143. Boy R.G., et al. Enzymes/transporters, in Molecular imaging II. 2008, Springer. p. 131-143.
214.
go back to reference Woodward JK et al. The roles of proteolytic enzymes in the development of tumour-induced bone disease in breast and prostate cancer. Bone. 2007;41(6):912–27.PubMed Woodward JK et al. The roles of proteolytic enzymes in the development of tumour-induced bone disease in breast and prostate cancer. Bone. 2007;41(6):912–27.PubMed
215.
go back to reference Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002;2(3):161–74.PubMed Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002;2(3):161–74.PubMed
216.
go back to reference Blobel CP. ADAMs: key components in EGFR signalling and development. Nat Rev Mol Cell Biol. 2005;6(1):32–43.PubMed Blobel CP. ADAMs: key components in EGFR signalling and development. Nat Rev Mol Cell Biol. 2005;6(1):32–43.PubMed
217.
go back to reference López-Otín C, Overall CM. Protease degradomics: a new challenge for proteomics. Nat Rev Mol Cell Biol. 2002;3(7):509–19.PubMed López-Otín C, Overall CM. Protease degradomics: a new challenge for proteomics. Nat Rev Mol Cell Biol. 2002;3(7):509–19.PubMed
218.
go back to reference López-Otín C, Matrisian LM. Emerging roles of proteases in tumour suppression. Nat Rev Cancer. 2007;7(10):800–8.PubMed López-Otín C, Matrisian LM. Emerging roles of proteases in tumour suppression. Nat Rev Cancer. 2007;7(10):800–8.PubMed
219.
go back to reference Sympson C.J., Bissell M.J. ,and Werb Z. Mammary gland tumor formation in transgenic mice overexpressing stromelysin-1. in Seminars in cancer biology. 1995. NIH Public Access. Sympson C.J., Bissell M.J. ,and Werb Z. Mammary gland tumor formation in transgenic mice overexpressing stromelysin-1. in Seminars in cancer biology. 1995. NIH Public Access.
220.
go back to reference Koblinski JE, Ahram M, Sloane BF. Unraveling the role of proteases in cancer. Clin Chim Acta. 2000;291(2):113–35.PubMed Koblinski JE, Ahram M, Sloane BF. Unraveling the role of proteases in cancer. Clin Chim Acta. 2000;291(2):113–35.PubMed
221.
go back to reference Shim K-N et al. Clinical significance of tissue levels of matrix metalloproteinases and tissue inhibitors of metalloproteinases in gastric cancer. J Gastroenterol. 2007;42(2):120–8.PubMed Shim K-N et al. Clinical significance of tissue levels of matrix metalloproteinases and tissue inhibitors of metalloproteinases in gastric cancer. J Gastroenterol. 2007;42(2):120–8.PubMed
222.
go back to reference Roy R, Yang J, Moses MA. Matrix metalloproteinases as novel biomarker s and potential therapeutic targets in human cancer. J Clin Oncol. 2009;27(31):5287–97.PubMedCentralPubMed Roy R, Yang J, Moses MA. Matrix metalloproteinases as novel biomarker s and potential therapeutic targets in human cancer. J Clin Oncol. 2009;27(31):5287–97.PubMedCentralPubMed
223.
go back to reference Chu D et al. Matrix metalloproteinase‐9 is associated with disease‐free survival and overall survival in patients with gastric cancer. Int J Cancer. 2011;129(4):887–95.PubMed Chu D et al. Matrix metalloproteinase‐9 is associated with disease‐free survival and overall survival in patients with gastric cancer. Int J Cancer. 2011;129(4):887–95.PubMed
224.
go back to reference Gerstein E et al. Comparative enzyme immunoassay of matrix metalloproteinases-2,-7,-9 and their tissue inhibitor-2 in tumors and plasma of patients with gastric cancer. Bull Exp Biol Med. 2009;148(6):899–902.PubMed Gerstein E et al. Comparative enzyme immunoassay of matrix metalloproteinases-2,-7,-9 and their tissue inhibitor-2 in tumors and plasma of patients with gastric cancer. Bull Exp Biol Med. 2009;148(6):899–902.PubMed
225.
go back to reference Fanjul-Fernández M et al. Matrix metalloproteinases: evolution, gene regulation and functional analysis in mouse models. Biochimica et Biophysica Acta (BBA)-Molecular. Cell Res. 2010;1803(1):3–19. Fanjul-Fernández M et al. Matrix metalloproteinases: evolution, gene regulation and functional analysis in mouse models. Biochimica et Biophysica Acta (BBA)-Molecular. Cell Res. 2010;1803(1):3–19.
226.
go back to reference Lynch CC. Matrix metalloproteinases as master regulators of the vicious cycle of bone metastasis. Bone. 2011;48(1):44–53.PubMed Lynch CC. Matrix metalloproteinases as master regulators of the vicious cycle of bone metastasis. Bone. 2011;48(1):44–53.PubMed
227.
go back to reference Brew K, Nagase H. The tissue inhibitors of metalloproteinases (TIMPs): an ancient family with structural and functional diversity. Biochimica et Biophysica Acta (BBA)-Molecular. Cell Res. 2010;1803(1):55–71. Brew K, Nagase H. The tissue inhibitors of metalloproteinases (TIMPs): an ancient family with structural and functional diversity. Biochimica et Biophysica Acta (BBA)-Molecular. Cell Res. 2010;1803(1):55–71.
228.
go back to reference Lambert E et al. TIMPs as multifacial proteins. Crit Rev Oncol Hematol. 2004;49(3):187–98.PubMed Lambert E et al. TIMPs as multifacial proteins. Crit Rev Oncol Hematol. 2004;49(3):187–98.PubMed
229.
go back to reference Remacle A et al. Furin regulates the intracellular activation and the uptake rate of cell surface-associated MT1-MMP. Oncogene. 2006;25(41):5648–55.PubMed Remacle A et al. Furin regulates the intracellular activation and the uptake rate of cell surface-associated MT1-MMP. Oncogene. 2006;25(41):5648–55.PubMed
230.
go back to reference Hua H et al. Matrix metalloproteinases in tumorigenesis: an evolving paradigm. Cell Mol Life Sci. 2011;68(23):3853–68.PubMed Hua H et al. Matrix metalloproteinases in tumorigenesis: an evolving paradigm. Cell Mol Life Sci. 2011;68(23):3853–68.PubMed
231.
go back to reference Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res. 2006;69(3):562–73.PubMed Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res. 2006;69(3):562–73.PubMed
232.
go back to reference Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell. 2010;141(1):52–67.PubMedCentralPubMed Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell. 2010;141(1):52–67.PubMedCentralPubMed
233.
go back to reference Hojilla CV, Wood GA, Khokha R. Metalloproteinases as common effectors of inflammation and extracellular matrix breakdown in breast cancer. Breast Cancer Res. 2008;10(2):205.PubMedCentralPubMed Hojilla CV, Wood GA, Khokha R. Metalloproteinases as common effectors of inflammation and extracellular matrix breakdown in breast cancer. Breast Cancer Res. 2008;10(2):205.PubMedCentralPubMed
234.
go back to reference Kähäri V-M, Saarialho-Kere U. Trends in molecular medicine: matrix metalloproteinases and their inhibitors in tumour growth and invasion. Ann Med. 1999;31(1):34–45.PubMed Kähäri V-M, Saarialho-Kere U. Trends in molecular medicine: matrix metalloproteinases and their inhibitors in tumour growth and invasion. Ann Med. 1999;31(1):34–45.PubMed
235.
go back to reference Ala-aho R, Kähäri V-M. Collagenases in cancer. Biochimie. 2005;87(3):273–86.PubMed Ala-aho R, Kähäri V-M. Collagenases in cancer. Biochimie. 2005;87(3):273–86.PubMed
236.
go back to reference Husmann K et al. Matrix metalloproteinase 1 promotes tumor formation and lung metastasis in an intratibial injection osteosarcoma mouse model. Biochim Biophys Acta (BBA)-Mol Basis Dis. 2013;1832(2):p. 347–354. Husmann K et al. Matrix metalloproteinase 1 promotes tumor formation and lung metastasis in an intratibial injection osteosarcoma mouse model. Biochim Biophys Acta (BBA)-Mol Basis Dis. 2013;1832(2):p. 347–354.
237.
go back to reference Brinckerhoff CE, Rutter JL, Benbow U. Interstitial collagenases as markers of tumor progression. Clin Cancer Res. 2000;6(12):4823–30.PubMed Brinckerhoff CE, Rutter JL, Benbow U. Interstitial collagenases as markers of tumor progression. Clin Cancer Res. 2000;6(12):4823–30.PubMed
238.
go back to reference Trivedi V et al. Platelet matrix metalloprotease-1 mediates thrombogenesis by activating PAR1 at a cryptic ligand site. Cell. 2009;137(2):332–43.PubMedCentralPubMed Trivedi V et al. Platelet matrix metalloprotease-1 mediates thrombogenesis by activating PAR1 at a cryptic ligand site. Cell. 2009;137(2):332–43.PubMedCentralPubMed
239.
go back to reference Vincenti MP et al. Regulating expression of the gene for matrix metalloproteinase-1 (collagenase): mechanisms that control enzyme activity, transcription, and mRNA stability. Crit Rev™ Eukaryot Gene Expr. 1996;6(4):391–411. Vincenti MP et al. Regulating expression of the gene for matrix metalloproteinase-1 (collagenase): mechanisms that control enzyme activity, transcription, and mRNA stability. Crit Rev™ Eukaryot Gene Expr. 1996;6(4):391–411.
240.
go back to reference Huntington JT et al. Overexpression of collagenase 1 (MMP-1) is mediated by the ERK pathway in invasive melanoma cells: role of BRAF mutation and fibroblast growth factor signaling. J Biol Chem. 2004;279(32):33168–76.PubMed Huntington JT et al. Overexpression of collagenase 1 (MMP-1) is mediated by the ERK pathway in invasive melanoma cells: role of BRAF mutation and fibroblast growth factor signaling. J Biol Chem. 2004;279(32):33168–76.PubMed
241.
go back to reference Nikkola J et al. High expression levels of collagenase‐1 and stromelysin‐1 correlate with shorter disease‐free survival in human metastatic melanoma. Int J Cancer. 2002;97(4):432–8.PubMed Nikkola J et al. High expression levels of collagenase‐1 and stromelysin‐1 correlate with shorter disease‐free survival in human metastatic melanoma. Int J Cancer. 2002;97(4):432–8.PubMed
242.
go back to reference Iida J, McCarthy JB. Expression of collagenase-1 (MMP-1) promotes melanoma growth through the generation of active transforming growth factor-β. Melanoma Res. 2007;17(4):205–13.PubMed Iida J, McCarthy JB. Expression of collagenase-1 (MMP-1) promotes melanoma growth through the generation of active transforming growth factor-β. Melanoma Res. 2007;17(4):205–13.PubMed
243.
go back to reference Hotary KB et al. Membrane type I matrix metalloproteinase usurps tumor growth control imposed by the three-dimensional extracellular matrix. Cell. 2003;114(1):33–45.PubMed Hotary KB et al. Membrane type I matrix metalloproteinase usurps tumor growth control imposed by the three-dimensional extracellular matrix. Cell. 2003;114(1):33–45.PubMed
244.
go back to reference Goerge T et al. Tumor-derived matrix metalloproteinase-1 targets endothelial proteinase-activated receptor 1 promoting endothelial cell activation. Cancer Res. 2006;66(15):7766–74.PubMed Goerge T et al. Tumor-derived matrix metalloproteinase-1 targets endothelial proteinase-activated receptor 1 promoting endothelial cell activation. Cancer Res. 2006;66(15):7766–74.PubMed
245.
go back to reference Blackburn JS et al. A matrix metalloproteinase-1/protease activated receptor-1 signaling axis promotes melanoma invasion and metastasis. Oncogene. 2009;28(48):4237–48.PubMedCentralPubMed Blackburn JS et al. A matrix metalloproteinase-1/protease activated receptor-1 signaling axis promotes melanoma invasion and metastasis. Oncogene. 2009;28(48):4237–48.PubMedCentralPubMed
246.
247.
go back to reference Kamel H et al. Immunoexpression of matrix metalloproteinase-2 (MMP-2) in malignant ovarian epithelial tumours. J Obstet Gynaecol Can. 2010;32(6):580–6.PubMed Kamel H et al. Immunoexpression of matrix metalloproteinase-2 (MMP-2) in malignant ovarian epithelial tumours. J Obstet Gynaecol Can. 2010;32(6):580–6.PubMed
248.
go back to reference Määttä M et al. Differential expression of matrix metalloproteinase (MMP)-2, MMP-9, and membrane type 1-MMP in hepatocellular and pancreatic adenocarcinoma: implications for tumor progression and clinical prognosis. Clin Cancer Res. 2000;6(7):2726–34.PubMed Määttä M et al. Differential expression of matrix metalloproteinase (MMP)-2, MMP-9, and membrane type 1-MMP in hepatocellular and pancreatic adenocarcinoma: implications for tumor progression and clinical prognosis. Clin Cancer Res. 2000;6(7):2726–34.PubMed
249.
go back to reference Galis ZS et al. Cytokine-stimulated human vascular smooth muscle cells synthesize a complement of enzymes required for extracellular matrix digestion. Circ Res. 1994;75(1):181–9.PubMed Galis ZS et al. Cytokine-stimulated human vascular smooth muscle cells synthesize a complement of enzymes required for extracellular matrix digestion. Circ Res. 1994;75(1):181–9.PubMed
250.
go back to reference Łukaszewicz-Zając M, Mroczko B, Szmitkowski M. Gastric cancer—the role of matrix metalloproteinases in tumor progression. Clin Chim Acta. 2011;412(19):1725–30.PubMed Łukaszewicz-Zając M, Mroczko B, Szmitkowski M. Gastric cancer—the role of matrix metalloproteinases in tumor progression. Clin Chim Acta. 2011;412(19):1725–30.PubMed
251.
go back to reference Ben-Yosef Y et al. Hypoxia of endothelial cells leads to MMP-2-dependent survival and death. Am J Physiol-Cell Physiol. 2005;289(5):C1321–31.PubMed Ben-Yosef Y et al. Hypoxia of endothelial cells leads to MMP-2-dependent survival and death. Am J Physiol-Cell Physiol. 2005;289(5):C1321–31.PubMed
252.
go back to reference McQuibban GA et al. Inflammation dampened by gelatinase A cleavage of monocyte chemoattractant protein-3. Science. 2000;289(5482):1202–6.PubMed McQuibban GA et al. Inflammation dampened by gelatinase A cleavage of monocyte chemoattractant protein-3. Science. 2000;289(5482):1202–6.PubMed
253.
go back to reference Duong TD, Erickson CA. MMP‐2 plays an essential role in producing epithelial‐mesenchymal transformations in the avian embryo. Dev Dyn. 2004;229(1):42–53.PubMed Duong TD, Erickson CA. MMP‐2 plays an essential role in producing epithelial‐mesenchymal transformations in the avian embryo. Dev Dyn. 2004;229(1):42–53.PubMed
254.
go back to reference Chetty C et al. MMP-2 siRNA induced Fas/CD95-mediated extrinsic II apoptotic pathway in the A549 lung adenocarcinoma cell line. Oncogene. 2007;26(55):7675–83.PubMedCentralPubMed Chetty C et al. MMP-2 siRNA induced Fas/CD95-mediated extrinsic II apoptotic pathway in the A549 lung adenocarcinoma cell line. Oncogene. 2007;26(55):7675–83.PubMedCentralPubMed
255.
go back to reference Yang Z, Strickland DK, Bornstein P. Extracellular matrix metalloproteinase 2 levels are regulated by the low density lipoprotein-related scavenger receptor and thrombospondin 2. J Biol Chem. 2001;276(11):8403–8.PubMed Yang Z, Strickland DK, Bornstein P. Extracellular matrix metalloproteinase 2 levels are regulated by the low density lipoprotein-related scavenger receptor and thrombospondin 2. J Biol Chem. 2001;276(11):8403–8.PubMed
256.
go back to reference López-Otín C, Palavalli LH, Samuels Y. Protective roles of matrix metalloproteinases. Cell Cycle. 2009;8(22):3657–62.PubMedCentralPubMed López-Otín C, Palavalli LH, Samuels Y. Protective roles of matrix metalloproteinases. Cell Cycle. 2009;8(22):3657–62.PubMedCentralPubMed
257.
go back to reference Van Lint P, Libert C. Matrix metalloproteinase-8: cleavage can be decisive. Cytokine Growth Factor Rev. 2006;17(4):217–23.PubMed Van Lint P, Libert C. Matrix metalloproteinase-8: cleavage can be decisive. Cytokine Growth Factor Rev. 2006;17(4):217–23.PubMed
258.
go back to reference Väyrynen JP et al. Serum MMP‐8 levels increase in colorectal cancer and correlate with disease course and inflammatory properties of primary tumors. Int J Cancer. 2012;131(4):E463–74.PubMed Väyrynen JP et al. Serum MMP‐8 levels increase in colorectal cancer and correlate with disease course and inflammatory properties of primary tumors. Int J Cancer. 2012;131(4):E463–74.PubMed
259.
go back to reference Thirkettle S et al. Matrix metalloproteinase 8 (collagenase 2) induces the expression of interleukins 6 and 8 in breast cancer cells. J Biol Chem. 2013;288(23):16282–94.PubMedCentralPubMed Thirkettle S et al. Matrix metalloproteinase 8 (collagenase 2) induces the expression of interleukins 6 and 8 in breast cancer cells. J Biol Chem. 2013;288(23):16282–94.PubMedCentralPubMed
260.
go back to reference Decock J et al. Association of matrix metalloproteinase-8 gene variation with breast cancer prognosis. Cancer Res. 2007;67(21):10214–21.PubMed Decock J et al. Association of matrix metalloproteinase-8 gene variation with breast cancer prognosis. Cancer Res. 2007;67(21):10214–21.PubMed
261.
go back to reference Stuelten CH et al. Breast cancer cells induce stromal fibroblasts to express MMP-9 via secretion of TNF-α and TGF-β. J Cell Sci. 2005;118(10):2143–53.PubMed Stuelten CH et al. Breast cancer cells induce stromal fibroblasts to express MMP-9 via secretion of TNF-α and TGF-β. J Cell Sci. 2005;118(10):2143–53.PubMed
262.
go back to reference Hallett MA et al. Anti-matrix metalloproteinase-9 DNAzyme decreases tumor growth in the MMTV-PyMT mouse model of breast cancer. Breast Cancer Res. 2013;15(1):R12.PubMedCentralPubMed Hallett MA et al. Anti-matrix metalloproteinase-9 DNAzyme decreases tumor growth in the MMTV-PyMT mouse model of breast cancer. Breast Cancer Res. 2013;15(1):R12.PubMedCentralPubMed
263.
go back to reference Morini M et al. The α3β1 integrin is associated with mammary carcinoma cell metastasis, invasion, and gelatinase B (MMP‐9) activity. Int J Cancer. 2000;87(3):336–42.PubMed Morini M et al. The α3β1 integrin is associated with mammary carcinoma cell metastasis, invasion, and gelatinase B (MMP‐9) activity. Int J Cancer. 2000;87(3):336–42.PubMed
264.
go back to reference Watanabe H et al. Matrix metalloproteinase-9 (92 kDa gelatinase/type IV collagenase) from U937 monoblastoid cells: correlation with cellular invasion. J Cell Sci. 1993;104(4):991–9.PubMed Watanabe H et al. Matrix metalloproteinase-9 (92 kDa gelatinase/type IV collagenase) from U937 monoblastoid cells: correlation with cellular invasion. J Cell Sci. 1993;104(4):991–9.PubMed
265.
go back to reference Coussens LM et al. Inflammatory mast cells up-regulate angiogenesis during squamous epithelial carcinogenesis. Genes Dev. 1999;13(11):1382–97.PubMedCentralPubMed Coussens LM et al. Inflammatory mast cells up-regulate angiogenesis during squamous epithelial carcinogenesis. Genes Dev. 1999;13(11):1382–97.PubMedCentralPubMed
266.
go back to reference Hojilla C, Mohammed F, Khokha R. Matrix metalloproteinases and their tissue inhibitors direct cell fate during cancer development. Br J Cancer. 2003;89(10):1817–21.PubMedCentralPubMed Hojilla C, Mohammed F, Khokha R. Matrix metalloproteinases and their tissue inhibitors direct cell fate during cancer development. Br J Cancer. 2003;89(10):1817–21.PubMedCentralPubMed
267.
go back to reference Bhoopathi P et al. Blockade of tumor growth due to matrix metalloproteinase-9 inhibition is mediated by sequential activation of β1-integrin, ERK, and NF-κB. J Biol Chem. 2008;283(3):1545–52.PubMedCentralPubMed Bhoopathi P et al. Blockade of tumor growth due to matrix metalloproteinase-9 inhibition is mediated by sequential activation of β1-integrin, ERK, and NF-κB. J Biol Chem. 2008;283(3):1545–52.PubMedCentralPubMed
268.
go back to reference Jordà M et al. Upregulation of MMP-9 in MDCK epithelial cell line in response to expression of the Snail transcription factor. J Cell Sci. 2005;118(15):3371–85.PubMed Jordà M et al. Upregulation of MMP-9 in MDCK epithelial cell line in response to expression of the Snail transcription factor. J Cell Sci. 2005;118(15):3371–85.PubMed
269.
go back to reference Xu D et al. Matrix metalloproteinase-9 regulates tumor cell invasion through cleavage of protease nexin-1. Cancer Res. 2010;70(17):6988–98.PubMedCentralPubMed Xu D et al. Matrix metalloproteinase-9 regulates tumor cell invasion through cleavage of protease nexin-1. Cancer Res. 2010;70(17):6988–98.PubMedCentralPubMed
270.
go back to reference Tester AM et al. LPS responsiveness and neutrophil chemotaxis in vivo require PMN MMP-8 activity. PLoS One. 2007;2(3):e312.PubMedCentralPubMed Tester AM et al. LPS responsiveness and neutrophil chemotaxis in vivo require PMN MMP-8 activity. PLoS One. 2007;2(3):e312.PubMedCentralPubMed
271.
go back to reference Kim M-J et al. TNF-α induces expression of urokinase-type plasminogen activator and β-catenin activation through generation of ROS in human breast epithelial cells. Biochem Pharmacol. 2010;80(12):2092–100.PubMed Kim M-J et al. TNF-α induces expression of urokinase-type plasminogen activator and β-catenin activation through generation of ROS in human breast epithelial cells. Biochem Pharmacol. 2010;80(12):2092–100.PubMed
272.
go back to reference Zhang S et al. Imbalance between expression of matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 in invasiveness and metastasis of human gastric carcinoma. World J Gastroenterol: WJG. 2003;9(5):899–904.PubMed Zhang S et al. Imbalance between expression of matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 in invasiveness and metastasis of human gastric carcinoma. World J Gastroenterol: WJG. 2003;9(5):899–904.PubMed
273.
go back to reference Wu ZS et al. Prognostic significance of MMP‐9 and TIMP‐1 serum and tissue expression in breast cancer. Int J Cancer. 2008;122(9):2050–6.PubMed Wu ZS et al. Prognostic significance of MMP‐9 and TIMP‐1 serum and tissue expression in breast cancer. Int J Cancer. 2008;122(9):2050–6.PubMed
274.
go back to reference Mroczko B et al. The diagnostic value of matrix metalloproteinase 9 (MMP-9) and tissue inhibitor of matrix metalloproteinases 1 (TIMP-1) determination in the sera of colorectal adenoma and cancer patients. Int J Color Dis. 2010;25(10):1177–84. Mroczko B et al. The diagnostic value of matrix metalloproteinase 9 (MMP-9) and tissue inhibitor of matrix metalloproteinases 1 (TIMP-1) determination in the sera of colorectal adenoma and cancer patients. Int J Color Dis. 2010;25(10):1177–84.
275.
go back to reference Zhang M et al. Expression of tissue levels of matrix metalloproteinases and tissue inhibitors of metalloproteinases in gastric adenocarcinoma. J Surg Oncol. 2011;103(3):243–7.PubMed Zhang M et al. Expression of tissue levels of matrix metalloproteinases and tissue inhibitors of metalloproteinases in gastric adenocarcinoma. J Surg Oncol. 2011;103(3):243–7.PubMed
276.
go back to reference Freije JM et al. Molecular cloning and expression of collagenase-3, a novel human matrix metalloproteinase produced by breast carcinomas. J Biol Chem. 1994;269(24):16766–73.PubMed Freije JM et al. Molecular cloning and expression of collagenase-3, a novel human matrix metalloproteinase produced by breast carcinomas. J Biol Chem. 1994;269(24):16766–73.PubMed
277.
go back to reference Knäuper V et al. Cellular mechanisms for human procollagenase-3 (MMP-13) activation: evidence that MT1-MMP (MMP-14) and gelatinase A (MMP-2) are able to generate active enzyme. J Biol Chem. 1996;271(29):17124–31.PubMed Knäuper V et al. Cellular mechanisms for human procollagenase-3 (MMP-13) activation: evidence that MT1-MMP (MMP-14) and gelatinase A (MMP-2) are able to generate active enzyme. J Biol Chem. 1996;271(29):17124–31.PubMed
278.
go back to reference Cowell S et al. Induction of matrix metalloproteinase activation cascades based on membrane-type 1 matrix metalloproteinase: associated activation of gelatinase A, gelatinase B and collagenase 3. Biochem J. 1998;331:453–8.PubMedCentralPubMed Cowell S et al. Induction of matrix metalloproteinase activation cascades based on membrane-type 1 matrix metalloproteinase: associated activation of gelatinase A, gelatinase B and collagenase 3. Biochem J. 1998;331:453–8.PubMedCentralPubMed
279.
go back to reference Johansson N et al. Collagenase‐3 (MMP‐13) is expressed by hypertrophic chondrocytes, periosteal cells, and osteoblasts during human fetal bone development. Dev Dyn. 1997;208(3):387–97.PubMed Johansson N et al. Collagenase‐3 (MMP‐13) is expressed by hypertrophic chondrocytes, periosteal cells, and osteoblasts during human fetal bone development. Dev Dyn. 1997;208(3):387–97.PubMed
280.
go back to reference Ravanti L et al. Expression of human collagenase-3 (MMP-13) by fetal skin fibroblasts is induced by transforming growth factor β via p38 mitogen-activated protein kinase. FASEB J. 2001;15(6):1098–100.PubMed Ravanti L et al. Expression of human collagenase-3 (MMP-13) by fetal skin fibroblasts is induced by transforming growth factor β via p38 mitogen-activated protein kinase. FASEB J. 2001;15(6):1098–100.PubMed
281.
go back to reference Mitchell PG et al. Cloning, expression, and type II collagenolytic activity of matrix metalloproteinase-13 from human osteoarthritic cartilage. J Clin Invest. 1996;97(3):761.PubMedCentralPubMed Mitchell PG et al. Cloning, expression, and type II collagenolytic activity of matrix metalloproteinase-13 from human osteoarthritic cartilage. J Clin Invest. 1996;97(3):761.PubMedCentralPubMed
282.
go back to reference McQuibban GA et al. Matrix metalloproteinase activity inactivates the CXC chemokine stromal cell-derived factor-1. J Biol Chem. 2001;276(47):43503–8.PubMed McQuibban GA et al. Matrix metalloproteinase activity inactivates the CXC chemokine stromal cell-derived factor-1. J Biol Chem. 2001;276(47):43503–8.PubMed
283.
go back to reference Uitto V-J et al. Collagenase-3 (matrix metalloproteinase-13) expression is induced in oral mucosal epithelium during chronic inflammation. Am J Pathol. 1998;152(6):1489.PubMedCentralPubMed Uitto V-J et al. Collagenase-3 (matrix metalloproteinase-13) expression is induced in oral mucosal epithelium during chronic inflammation. Am J Pathol. 1998;152(6):1489.PubMedCentralPubMed
284.
go back to reference Zhang Y et al. Overexpression of tyrosine kinase B protein as a predictor for distant metastases and prognosis in gastric carcinoma. Oncology. 2008;75(1–2):17–26.PubMed Zhang Y et al. Overexpression of tyrosine kinase B protein as a predictor for distant metastases and prognosis in gastric carcinoma. Oncology. 2008;75(1–2):17–26.PubMed
285.
go back to reference Yamada T et al. Overexpression of MMP-13 gene in colorectal cancer with liver metastasis. Anticancer Res. 2010;30(7):2693–9.PubMed Yamada T et al. Overexpression of MMP-13 gene in colorectal cancer with liver metastasis. Anticancer Res. 2010;30(7):2693–9.PubMed
286.
go back to reference Mäkinen LK et al. Prognostic significance of matrix metalloproteinase‐2, ‐8, ‐9, and ‐13 in oral tongue cancer. J Oral Pathol Med. 2012;41(5):394–9.PubMed Mäkinen LK et al. Prognostic significance of matrix metalloproteinase‐2, ‐8, ‐9, and ‐13 in oral tongue cancer. J Oral Pathol Med. 2012;41(5):394–9.PubMed
287.
go back to reference Kominsky SL et al. MMP-13 is over-expressed in renal cell carcinoma bone metastasis and is induced by TGF-β1. Clin Exp Metastasis. 2008;25(8):865–70.PubMed Kominsky SL et al. MMP-13 is over-expressed in renal cell carcinoma bone metastasis and is induced by TGF-β1. Clin Exp Metastasis. 2008;25(8):865–70.PubMed
288.
go back to reference Hsu C-P, Shen G-H, Ko J-L. Matrix metalloproteinase-13 expression is associated with bone marrow microinvolvement and prognosis in non-small cell lung cancer. Lung Cancer. 2006;52(3):349–57.PubMed Hsu C-P, Shen G-H, Ko J-L. Matrix metalloproteinase-13 expression is associated with bone marrow microinvolvement and prognosis in non-small cell lung cancer. Lung Cancer. 2006;52(3):349–57.PubMed
289.
go back to reference Heikkilä P et al. Bisphosphonates inhibit stromelysin-1 (MMP-3), matrix metalloelastase (MMP-12), collagenase-3 (MMP-13) and enamelysin (MMP-20), but not urokinase-type plasminogen activator, and diminish invasion and migration of human malignant and endothelial cell lines. Anti-Cancer Drugs. 2002;13(3):245–54.PubMed Heikkilä P et al. Bisphosphonates inhibit stromelysin-1 (MMP-3), matrix metalloelastase (MMP-12), collagenase-3 (MMP-13) and enamelysin (MMP-20), but not urokinase-type plasminogen activator, and diminish invasion and migration of human malignant and endothelial cell lines. Anti-Cancer Drugs. 2002;13(3):245–54.PubMed
290.
go back to reference Luukkaa M et al. Association between high collagenase‐3 expression levels and poor prognosis in patients with head and neck cancer. Head Neck. 2006;28(3):225–34.PubMed Luukkaa M et al. Association between high collagenase‐3 expression levels and poor prognosis in patients with head and neck cancer. Head Neck. 2006;28(3):225–34.PubMed
291.
go back to reference Wang J et al. Expression of MMP-13 is associated with invasion and metastasis of papillary thyroid carcinoma. Eur Rev Med Pharmacol Sci. 2013;17(4):427–35.PubMed Wang J et al. Expression of MMP-13 is associated with invasion and metastasis of papillary thyroid carcinoma. Eur Rev Med Pharmacol Sci. 2013;17(4):427–35.PubMed
292.
go back to reference Koshikawa N et al. Role of cell surface metalloprotease MT1-MMP in epithelial cell migration over laminin-5. J Cell Biol. 2000;148(3):615–24.PubMedCentralPubMed Koshikawa N et al. Role of cell surface metalloprotease MT1-MMP in epithelial cell migration over laminin-5. J Cell Biol. 2000;148(3):615–24.PubMedCentralPubMed
293.
go back to reference Koshikawa N et al. Membrane-type matrix metalloproteinase-1 (MT1-MMP) is a processing enzyme for human laminin γ2 chain. J Biol Chem. 2005;280(1):88–93.PubMed Koshikawa N et al. Membrane-type matrix metalloproteinase-1 (MT1-MMP) is a processing enzyme for human laminin γ2 chain. J Biol Chem. 2005;280(1):88–93.PubMed
294.
go back to reference Sadowski T et al. Matrix metalloproteinase 19 processes the laminin 5 gamma 2 chain and induces epithelial cell migration. Cell Mol Life Sci CMLS. 2005;62(7–8):870–80.PubMed Sadowski T et al. Matrix metalloproteinase 19 processes the laminin 5 gamma 2 chain and induces epithelial cell migration. Cell Mol Life Sci CMLS. 2005;62(7–8):870–80.PubMed
295.
go back to reference Mañes S et al. The matrix metalloproteinase-9 regulates the insulin-like growth factor-triggered autocrine response in DU-145 carcinoma cells. J Biol Chem. 1999;274(11):6935–45.PubMed Mañes S et al. The matrix metalloproteinase-9 regulates the insulin-like growth factor-triggered autocrine response in DU-145 carcinoma cells. J Biol Chem. 1999;274(11):6935–45.PubMed
296.
go back to reference Rorive S et al. Matrix metalloproteinase‐9 interplays with the IGFBP2–IGFII complex to promote cell growth and motility in astrocytomas. Glia. 2008;56(15):1679–90.PubMed Rorive S et al. Matrix metalloproteinase‐9 interplays with the IGFBP2–IGFII complex to promote cell growth and motility in astrocytomas. Glia. 2008;56(15):1679–90.PubMed
297.
go back to reference Noë V et al. Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J Cell Sci. 2001;114(1):111–8.PubMed Noë V et al. Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J Cell Sci. 2001;114(1):111–8.PubMed
298.
go back to reference Maretzky T et al. ADAM10 mediates E-cadherin shedding and regulates epithelial cell-cell adhesion, migration, and β-catenin translocation. Proc Natl Acad Sci U S A. 2005;102(26):9182–7.PubMedCentralPubMed Maretzky T et al. ADAM10 mediates E-cadherin shedding and regulates epithelial cell-cell adhesion, migration, and β-catenin translocation. Proc Natl Acad Sci U S A. 2005;102(26):9182–7.PubMedCentralPubMed
299.
go back to reference Illman SA et al. Epilysin (MMP-28) induces TGF-β mediated epithelial to mesenchymal transition in lung carcinoma cells. J Cell Sci. 2006;119(18):3856–65.PubMed Illman SA et al. Epilysin (MMP-28) induces TGF-β mediated epithelial to mesenchymal transition in lung carcinoma cells. J Cell Sci. 2006;119(18):3856–65.PubMed
300.
go back to reference Koshikawa N et al. Membrane type 1-matrix metalloproteinase cleaves off the NH2-terminal portion of heparin-binding epidermal growth factor and converts it into a heparin-independent growth factor. Cancer Res. 2010;70(14):6093–103.PubMed Koshikawa N et al. Membrane type 1-matrix metalloproteinase cleaves off the NH2-terminal portion of heparin-binding epidermal growth factor and converts it into a heparin-independent growth factor. Cancer Res. 2010;70(14):6093–103.PubMed
301.
go back to reference Loechel F, Wewer UM. Activation of ADAM 12 protease by copper. FEBS Lett. 2001;506(1):65–8.PubMed Loechel F, Wewer UM. Activation of ADAM 12 protease by copper. FEBS Lett. 2001;506(1):65–8.PubMed
302.
go back to reference Birkedal-Hansen H. Proteolytic remodeling of extracellular matrix. Curr Opin Cell Biol. 1995;7(5):728–35.PubMed Birkedal-Hansen H. Proteolytic remodeling of extracellular matrix. Curr Opin Cell Biol. 1995;7(5):728–35.PubMed
303.
go back to reference Hawinkels LJ et al. Matrix metalloproteinase-14 (MT1-MMP)-mediated endoglin shedding inhibits tumor angiogenesis. Cancer Res. 2010;70(10):4141–50.PubMed Hawinkels LJ et al. Matrix metalloproteinase-14 (MT1-MMP)-mediated endoglin shedding inhibits tumor angiogenesis. Cancer Res. 2010;70(10):4141–50.PubMed
304.
go back to reference Pozzi A et al. Elevated matrix metalloprotease and angiostatin levels in integrin α1 knockout mice cause reduced tumor vascularization. Proc Natl Acad Sci U S A. 2000;97(5):2202–7.PubMedCentralPubMed Pozzi A et al. Elevated matrix metalloprotease and angiostatin levels in integrin α1 knockout mice cause reduced tumor vascularization. Proc Natl Acad Sci U S A. 2000;97(5):2202–7.PubMedCentralPubMed
305.
go back to reference Chang J-H et al. Functional characterization of neostatins, the MMP-derived, enzymatic cleavage products of type XVIII collagen. FEBS Lett. 2005;579(17):3601–6.PubMed Chang J-H et al. Functional characterization of neostatins, the MMP-derived, enzymatic cleavage products of type XVIII collagen. FEBS Lett. 2005;579(17):3601–6.PubMed
306.
go back to reference Wen W et al. The generation of endostatin is mediated by elastase. Cancer Res. 1999;59(24):6052–6.PubMed Wen W et al. The generation of endostatin is mediated by elastase. Cancer Res. 1999;59(24):6052–6.PubMed
308.
go back to reference Cho A, Reidy MA. Matrix metalloproteinase-9 is necessary for the regulation of smooth muscle cell replication and migration after arterial injury. Circ Res. 2002;91(9):845–51.PubMed Cho A, Reidy MA. Matrix metalloproteinase-9 is necessary for the regulation of smooth muscle cell replication and migration after arterial injury. Circ Res. 2002;91(9):845–51.PubMed
309.
go back to reference Limb GA et al. Matrix metalloproteinase-1 associates with intracellular organelles and confers resistance to lamin A/C degradation during apoptosis. Am J Pathol. 2005;166(5):1555–63.PubMedCentralPubMed Limb GA et al. Matrix metalloproteinase-1 associates with intracellular organelles and confers resistance to lamin A/C degradation during apoptosis. Am J Pathol. 2005;166(5):1555–63.PubMedCentralPubMed
310.
go back to reference Balbín M et al. Loss of collagenase-2 confers increased skin tumor susceptibility to male mice. Nat Genet. 2003;35(3):252–7.PubMed Balbín M et al. Loss of collagenase-2 confers increased skin tumor susceptibility to male mice. Nat Genet. 2003;35(3):252–7.PubMed
311.
go back to reference Gutiérrez-Fernández A et al. Matrix metalloproteinase-8 functions as a metastasis suppressor through modulation of tumor cell adhesion and invasion. Cancer Res. 2008;68(8):2755–63.PubMed Gutiérrez-Fernández A et al. Matrix metalloproteinase-8 functions as a metastasis suppressor through modulation of tumor cell adhesion and invasion. Cancer Res. 2008;68(8):2755–63.PubMed
312.
go back to reference Acuff HB et al. Analysis of host- and tumor-derived proteinases using a custom dual species microarray reveals a protective role for stromal matrix metalloproteinase-12 in non-small cell lung cancer. Cancer Res. 2006;66(16):7968–75.PubMed Acuff HB et al. Analysis of host- and tumor-derived proteinases using a custom dual species microarray reveals a protective role for stromal matrix metalloproteinase-12 in non-small cell lung cancer. Cancer Res. 2006;66(16):7968–75.PubMed
313.
go back to reference Houghton AM et al. Macrophage elastase (matrix metalloproteinase-12) suppresses growth of lung metastases. Cancer Res. 2006;66(12):6149–55.PubMed Houghton AM et al. Macrophage elastase (matrix metalloproteinase-12) suppresses growth of lung metastases. Cancer Res. 2006;66(12):6149–55.PubMed
314.
go back to reference Deryugina EI, Quigley JP. Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev. 2006;25(1):9–34.PubMed Deryugina EI, Quigley JP. Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev. 2006;25(1):9–34.PubMed
315.
go back to reference Ramnath N, Creaven PJ. Matrix metalloproteinase inhibitors. Curr Oncol Rep. 2004;6(2):96–102.PubMed Ramnath N, Creaven PJ. Matrix metalloproteinase inhibitors. Curr Oncol Rep. 2004;6(2):96–102.PubMed
316.
go back to reference Bourboulia D. and Stetler-Stevenson.W.G. Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs): positive and negative regulators in tumor cell adhesion. in Seminars in cancer biology. 2010. Elsevier. Bourboulia D. and Stetler-Stevenson.W.G. Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs): positive and negative regulators in tumor cell adhesion. in Seminars in cancer biology. 2010. Elsevier.
317.
go back to reference Khokha R. Suppression of the tumorigenic and metastatic abilities of murine B16-F10 melanoma cells in vivo by the overexpression of the tissue inhibitor of the metalloproteinases-1. J National Cancer Inst. 1994;86(4):299–304. Khokha R. Suppression of the tumorigenic and metastatic abilities of murine B16-F10 melanoma cells in vivo by the overexpression of the tissue inhibitor of the metalloproteinases-1. J National Cancer Inst. 1994;86(4):299–304.
318.
go back to reference Schrötzlmair F et al. Tissue inhibitor of metalloproteinases‐1‐induced scattered liver metastasis is mediated by host‐derived urokinase‐type plasminogen activator. J Cell Mol Med. 2010;14(12):2760–70.PubMedCentralPubMed Schrötzlmair F et al. Tissue inhibitor of metalloproteinases‐1‐induced scattered liver metastasis is mediated by host‐derived urokinase‐type plasminogen activator. J Cell Mol Med. 2010;14(12):2760–70.PubMedCentralPubMed
319.
go back to reference Bigelow RL et al. TIMP-1 overexpression promotes tumorigenesis of MDA-MB-231 breast cancer cells and alters expression of a subset of cancer promoting genes in vivo distinct from those observed in vitro. Breast Cancer Res Treat. 2009;117(1):31–44.PubMed Bigelow RL et al. TIMP-1 overexpression promotes tumorigenesis of MDA-MB-231 breast cancer cells and alters expression of a subset of cancer promoting genes in vivo distinct from those observed in vitro. Breast Cancer Res Treat. 2009;117(1):31–44.PubMed
320.
go back to reference Guedez L et al. In vitro suppression of programmed cell death of B cells by tissue inhibitor of metalloproteinases-1. J Clin Invest. 1998;102(11):2002.PubMedCentralPubMed Guedez L et al. In vitro suppression of programmed cell death of B cells by tissue inhibitor of metalloproteinases-1. J Clin Invest. 1998;102(11):2002.PubMedCentralPubMed
321.
go back to reference Hayakawa T et al. Growth-promoting activity of tissue inhibitor of metalloproteinases-1 (TIMP-1) for a wide range of cells. A possible new growth factor in serum. FEBS Lett. 1992;298(1):29–32.PubMed Hayakawa T et al. Growth-promoting activity of tissue inhibitor of metalloproteinases-1 (TIMP-1) for a wide range of cells. A possible new growth factor in serum. FEBS Lett. 1992;298(1):29–32.PubMed
322.
go back to reference Jezierska A, Motyl T. Matrix metalloproteinase-2 involvement in breast cancer progression: a mini-review. Med Sci Monit Basic Res. 2009;15(2):RA32–40. Jezierska A, Motyl T. Matrix metalloproteinase-2 involvement in breast cancer progression: a mini-review. Med Sci Monit Basic Res. 2009;15(2):RA32–40.
323.
go back to reference Caterina JJ et al. Inactivating mutation of the mouse tissue inhibitor of metalloproteinases-2 (Timp-2) gene alters proMMP-2 activation. J Biol Chem. 2000;275(34):26416–22.PubMed Caterina JJ et al. Inactivating mutation of the mouse tissue inhibitor of metalloproteinases-2 (Timp-2) gene alters proMMP-2 activation. J Biol Chem. 2000;275(34):26416–22.PubMed
324.
go back to reference Bigg HF et al. Specific, high affinity binding of tissue inhibitor of metalloproteinases-4 (TIMP-4) to the COOH-terminal hemopexin-like domain of human gelatinase A TIMP-4 binds progelatinase A and the COOH-terminal domain in a similar manner to TIMP-2. J Biol Chem. 1997;272(24):15496–500.PubMed Bigg HF et al. Specific, high affinity binding of tissue inhibitor of metalloproteinases-4 (TIMP-4) to the COOH-terminal hemopexin-like domain of human gelatinase A TIMP-4 binds progelatinase A and the COOH-terminal domain in a similar manner to TIMP-2. J Biol Chem. 1997;272(24):15496–500.PubMed
325.
go back to reference Liacini A et al. Induction of matrix metalloproteinase-13 gene expression by TNF-α is mediated by MAP kinases, AP-1, and NF-κB transcription factors in articular chondrocytes. Exp Cell Res. 2003;288(1):208–17.PubMed Liacini A et al. Induction of matrix metalloproteinase-13 gene expression by TNF-α is mediated by MAP kinases, AP-1, and NF-κB transcription factors in articular chondrocytes. Exp Cell Res. 2003;288(1):208–17.PubMed
326.
go back to reference Rozanov DV et al. The low density lipoprotein receptor-related protein LRP is regulated by membrane type-1 matrix metalloproteinase (MT1-MMP) proteolysis in malignant cells. J Biol Chem. 2004;279(6):4260–8.PubMed Rozanov DV et al. The low density lipoprotein receptor-related protein LRP is regulated by membrane type-1 matrix metalloproteinase (MT1-MMP) proteolysis in malignant cells. J Biol Chem. 2004;279(6):4260–8.PubMed
327.
go back to reference Eccles SA et al. Control of lymphatic and hematogenous metastasis of a rat mammary carcinoma by the matrix metalloproteinase inhibitor batimastat (BB-94). Cancer Res. 1996;56(12):2815–22.PubMed Eccles SA et al. Control of lymphatic and hematogenous metastasis of a rat mammary carcinoma by the matrix metalloproteinase inhibitor batimastat (BB-94). Cancer Res. 1996;56(12):2815–22.PubMed
328.
go back to reference Prontera C et al. Inhibition of gelatinase A (MMP‐2) by batimastat and captopril reduces tumor growth and lung metastases in mice bearing Lewis lung carcinoma. Int J Cancer. 1999;81(5):761–6.PubMed Prontera C et al. Inhibition of gelatinase A (MMP‐2) by batimastat and captopril reduces tumor growth and lung metastases in mice bearing Lewis lung carcinoma. Int J Cancer. 1999;81(5):761–6.PubMed
329.
go back to reference Hidalgo M, Eckhardt SG. Development of matrix metalloproteinase inhibitors in cancer therapy. J Natl Cancer Inst. 2001;93(3):178–93.PubMed Hidalgo M, Eckhardt SG. Development of matrix metalloproteinase inhibitors in cancer therapy. J Natl Cancer Inst. 2001;93(3):178–93.PubMed
330.
go back to reference Scatena R. Prinomastat, a hydroxamate-based matrix metalloproteinase inhibitor. A novel pharmacological approach for tissue remodelling-related diseases. Expert Opin Invest Drugs. 2000;9(9):2159–65. Scatena R. Prinomastat, a hydroxamate-based matrix metalloproteinase inhibitor. A novel pharmacological approach for tissue remodelling-related diseases. Expert Opin Invest Drugs. 2000;9(9):2159–65.
331.
go back to reference Liu J et al. Early combined treatment with carboplatin and the MMP inhibitor, prinomastat, prolongs survival and reduces systemic metastasis in an aggressive orthotopic lung cancer model. Lung Cancer. 2003;42(3):335–44.PubMed Liu J et al. Early combined treatment with carboplatin and the MMP inhibitor, prinomastat, prolongs survival and reduces systemic metastasis in an aggressive orthotopic lung cancer model. Lung Cancer. 2003;42(3):335–44.PubMed
332.
go back to reference Hoffman A et al. Carbamoylphosphonate matrix metalloproteinase inhibitors 6: cis-2-aminocyclohexylcarbamoylphosphonic acid, a novel orally active antimetastatic matrix metalloproteinase-2 selective inhibitor—synthesis and pharmacodynamic and pharmacokinetic analysis. J Med Chem. 2008;51(5):1406–14.PubMed Hoffman A et al. Carbamoylphosphonate matrix metalloproteinase inhibitors 6: cis-2-aminocyclohexylcarbamoylphosphonic acid, a novel orally active antimetastatic matrix metalloproteinase-2 selective inhibitor—synthesis and pharmacodynamic and pharmacokinetic analysis. J Med Chem. 2008;51(5):1406–14.PubMed
333.
go back to reference Maquoi E et al. Anti-invasive, antitumoral, and antiangiogenic efficacy of a pyrimidine-2,4,6-trione derivative, an orally active and selective matrix metalloproteinases inhibitor. Clin Cancer Res. 2004;10(12):4038–47.PubMed Maquoi E et al. Anti-invasive, antitumoral, and antiangiogenic efficacy of a pyrimidine-2,4,6-trione derivative, an orally active and selective matrix metalloproteinases inhibitor. Clin Cancer Res. 2004;10(12):4038–47.PubMed
334.
go back to reference Lubbe WJ et al. Tumor epithelial cell matrix metalloproteinase 9 is a target for antimetastatic therapy in colorectal cancer. Clin Cancer Res. 2006;12(6):1876–82.PubMed Lubbe WJ et al. Tumor epithelial cell matrix metalloproteinase 9 is a target for antimetastatic therapy in colorectal cancer. Clin Cancer Res. 2006;12(6):1876–82.PubMed
335.
go back to reference Tao P et al. Matrix metalloproteinase 2 inhibition: combined quantum mechanics and molecular mechanics studies of the inhibition mechanism of (4-phenoxyphenylsulfonyl) methylthiirane and its oxirane analogue. Biochemistry. 2009;48(41):9839–47.PubMedCentralPubMed Tao P et al. Matrix metalloproteinase 2 inhibition: combined quantum mechanics and molecular mechanics studies of the inhibition mechanism of (4-phenoxyphenylsulfonyl) methylthiirane and its oxirane analogue. Biochemistry. 2009;48(41):9839–47.PubMedCentralPubMed
336.
go back to reference Krüger A et al. Antimetastatic activity of a novel mechanism-based gelatinase inhibitor. Cancer Res. 2005;65(9):3523–6.PubMed Krüger A et al. Antimetastatic activity of a novel mechanism-based gelatinase inhibitor. Cancer Res. 2005;65(9):3523–6.PubMed
337.
go back to reference Bonfil RD et al. Inhibition of human prostate cancer growth, osteolysis and angiogenesis in a bone metastasis model by a novel mechanism‐based selective gelatinase inhibitor. Int J Cancer. 2006;118(11):2721–6.PubMed Bonfil RD et al. Inhibition of human prostate cancer growth, osteolysis and angiogenesis in a bone metastasis model by a novel mechanism‐based selective gelatinase inhibitor. Int J Cancer. 2006;118(11):2721–6.PubMed
338.
go back to reference Devy L et al. Selective inhibition of matrix metalloproteinase-14 blocks tumor growth, invasion, and angiogenesis. Cancer Res. 2009;69(4):1517–26.PubMed Devy L et al. Selective inhibition of matrix metalloproteinase-14 blocks tumor growth, invasion, and angiogenesis. Cancer Res. 2009;69(4):1517–26.PubMed
339.
go back to reference Rawlings ND, Barrett AJ. Families of serine peptidases. Methods Enzymol. 1993;244:19–61. Rawlings ND, Barrett AJ. Families of serine peptidases. Methods Enzymol. 1993;244:19–61.
340.
go back to reference Netzel-Arnett S et al. Membrane anchored serine proteases: a rapidly expanding group of cell surface proteolytic enzymes with potential roles in cancer. Cancer Metastasis Rev. 2003;22(2–3):237–58.PubMed Netzel-Arnett S et al. Membrane anchored serine proteases: a rapidly expanding group of cell surface proteolytic enzymes with potential roles in cancer. Cancer Metastasis Rev. 2003;22(2–3):237–58.PubMed
341.
go back to reference Duffy MJ et al. Urokinase‐plasminogen activator, a marker for aggressive breast carcinomas. Preliminary report. Cancer. 1988;62(3):531–3.PubMed Duffy MJ et al. Urokinase‐plasminogen activator, a marker for aggressive breast carcinomas. Preliminary report. Cancer. 1988;62(3):531–3.PubMed
342.
go back to reference Stepanova V, Tkachuk V. Urokinase as a multidomain protein and polyfunctional cell regulator. Biochemistry (Moscow). 2002;67(1):109–18. Stepanova V, Tkachuk V. Urokinase as a multidomain protein and polyfunctional cell regulator. Biochemistry (Moscow). 2002;67(1):109–18.
343.
go back to reference Thummarati P et al. High level of urokinase plasminogen activator contributes to cholangiocarcinoma invasion and metastasis. World J Gastroenterol: WJG. 2012;18(3):244.PubMedCentralPubMed Thummarati P et al. High level of urokinase plasminogen activator contributes to cholangiocarcinoma invasion and metastasis. World J Gastroenterol: WJG. 2012;18(3):244.PubMedCentralPubMed
344.
go back to reference Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100(1):57–70.PubMed Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100(1):57–70.PubMed
345.
go back to reference Ulisse S et al. The urokinase plasminogen activator system: a target for anti-cancer therapy. Curr Cancer Drug Targets. 2009;9(1):32–71.PubMed Ulisse S et al. The urokinase plasminogen activator system: a target for anti-cancer therapy. Curr Cancer Drug Targets. 2009;9(1):32–71.PubMed
346.
go back to reference Dass K et al. Evolving role of uPA/uPAR system in human cancers. Cancer Treat Rev. 2008;34(2):122–36.PubMed Dass K et al. Evolving role of uPA/uPAR system in human cancers. Cancer Treat Rev. 2008;34(2):122–36.PubMed
347.
go back to reference Ossowski L, Aguirre-Ghiso JA. Urokinase receptor and integrin partnership: coordination of signaling for cell adhesion, migration and growth. Curr Opin Cell Biol. 2000;12(5):613–20.PubMed Ossowski L, Aguirre-Ghiso JA. Urokinase receptor and integrin partnership: coordination of signaling for cell adhesion, migration and growth. Curr Opin Cell Biol. 2000;12(5):613–20.PubMed
348.
go back to reference Yu W, Kim J, Ossowski L. Reduction in surface urokinase receptor forces malignant cells into a protracted state of dormancy. J Cell Biol. 1997;137(3):767–77.PubMedCentralPubMed Yu W, Kim J, Ossowski L. Reduction in surface urokinase receptor forces malignant cells into a protracted state of dormancy. J Cell Biol. 1997;137(3):767–77.PubMedCentralPubMed
349.
go back to reference Choong PF, Nadesapillai AP. Urokinase plasminogen activator system: a multifunctional role in tumor progression and metastasis. Clin Orthop Relat Res. 2003;415:S46–58.PubMed Choong PF, Nadesapillai AP. Urokinase plasminogen activator system: a multifunctional role in tumor progression and metastasis. Clin Orthop Relat Res. 2003;415:S46–58.PubMed
350.
go back to reference Binder BR, Mihaly J, Prager GW. uPAR-uPA-PAI-1 interactions and signaling: a vascular biologist’s view. Thromb Haemost-Stuttgart-. 2007;97(3):336. Binder BR, Mihaly J, Prager GW. uPAR-uPA-PAI-1 interactions and signaling: a vascular biologist’s view. Thromb Haemost-Stuttgart-. 2007;97(3):336.
351.
go back to reference Pepper MS. Role of the matrix metalloproteinase and plasminogen activator–plasmin systems in angiogenesis. Arterioscler, Thromb, Vasc Biol. 2001;21(7):1104–17. Pepper MS. Role of the matrix metalloproteinase and plasminogen activator–plasmin systems in angiogenesis. Arterioscler, Thromb, Vasc Biol. 2001;21(7):1104–17.
352.
go back to reference Gondi CS et al. Downregulation of uPAR and uPA activates caspase mediated apoptosis, inhibits the PI3k/AKT pathway. Int J Oncol. 2007;31(1):19.PubMedCentralPubMed Gondi CS et al. Downregulation of uPAR and uPA activates caspase mediated apoptosis, inhibits the PI3k/AKT pathway. Int J Oncol. 2007;31(1):19.PubMedCentralPubMed
353.
go back to reference Prager GW et al. Urokinase mediates endothelial cell survival via induction of the X-linked inhibitor of apoptosis protein. Blood. 2009;113(6):1383–90.PubMed Prager GW et al. Urokinase mediates endothelial cell survival via induction of the X-linked inhibitor of apoptosis protein. Blood. 2009;113(6):1383–90.PubMed
354.
go back to reference Shetty S et al. Urokinase induces activation of STAT3 in lung epithelial cells. Am J Physiol-Lung Cell Mol Physiol. 2006;291(4):L772–80.PubMed Shetty S et al. Urokinase induces activation of STAT3 in lung epithelial cells. Am J Physiol-Lung Cell Mol Physiol. 2006;291(4):L772–80.PubMed
355.
go back to reference Malinowsky K et al. uPA and PAI-1-related signaling pathways differ between primary breast cancers and lymph node metastases. Transl Oncol. 2012;5(2):98–IN3.PubMedCentralPubMed Malinowsky K et al. uPA and PAI-1-related signaling pathways differ between primary breast cancers and lymph node metastases. Transl Oncol. 2012;5(2):98–IN3.PubMedCentralPubMed
356.
go back to reference Ghamande SA et al. A phase 2, randomized, double-blind, placebo-controlled trial of clinical activity and safety of subcutaneous A6 in women with asymptomatic CA125 progression after first-line chemotherapy of epithelial ovarian cancer. Gynecol Oncol. 2008;111(1):89–94.PubMed Ghamande SA et al. A phase 2, randomized, double-blind, placebo-controlled trial of clinical activity and safety of subcutaneous A6 in women with asymptomatic CA125 progression after first-line chemotherapy of epithelial ovarian cancer. Gynecol Oncol. 2008;111(1):89–94.PubMed
357.
go back to reference Ellis V, Dano K. Specific inhibition of the activity of the urokinase receptor-mediated cell-surface plasminogen activation system by suramin. Biochem J. 1993;296(Pt 2):505–10.PubMedCentralPubMed Ellis V, Dano K. Specific inhibition of the activity of the urokinase receptor-mediated cell-surface plasminogen activation system by suramin. Biochem J. 1993;296(Pt 2):505–10.PubMedCentralPubMed
358.
go back to reference Schmitt M et al. Cancer therapy trials employing level-of-evidence-1 disease forecast cancer biomarkers uPA and its inhibitor PAI-1. Expert Rev Mol Diagn. 2011;11(6):617–34.PubMed Schmitt M et al. Cancer therapy trials employing level-of-evidence-1 disease forecast cancer biomarkers uPA and its inhibitor PAI-1. Expert Rev Mol Diagn. 2011;11(6):617–34.PubMed
359.
go back to reference Benes P, Vetvicka V, Fusek M. Cathepsin D—many functions of one aspartic protease. Crit Rev Oncol Hematol. 2008;68(1):12–28.PubMedCentralPubMed Benes P, Vetvicka V, Fusek M. Cathepsin D—many functions of one aspartic protease. Crit Rev Oncol Hematol. 2008;68(1):12–28.PubMedCentralPubMed
360.
go back to reference Liaudet-Coopman E et al. Cathepsin D: newly discovered functions of a long-standing aspartic protease in cancer and apoptosis. Cancer Lett. 2006;237(2):167–79.PubMed Liaudet-Coopman E et al. Cathepsin D: newly discovered functions of a long-standing aspartic protease in cancer and apoptosis. Cancer Lett. 2006;237(2):167–79.PubMed
361.
go back to reference Masson O et al. Pathophysiological functions of cathepsin D: targeting its catalytic activity versus its protein binding activity? Biochimie. 2010;92(11):1635–43.PubMed Masson O et al. Pathophysiological functions of cathepsin D: targeting its catalytic activity versus its protein binding activity? Biochimie. 2010;92(11):1635–43.PubMed
362.
go back to reference Fuseka M, Vetvicka V. Dual role of cathepsin D: ligand and protease. Biomed Papers. 2005;149(1):43–50. Fuseka M, Vetvicka V. Dual role of cathepsin D: ligand and protease. Biomed Papers. 2005;149(1):43–50.
363.
go back to reference Baechle D et al. Cathepsin D is present in human eccrine sweat and involved in the postsecretory processing of the antimicrobial peptide DCD-1 L. J Biol Chem. 2006;281(9):5406–15.PubMed Baechle D et al. Cathepsin D is present in human eccrine sweat and involved in the postsecretory processing of the antimicrobial peptide DCD-1 L. J Biol Chem. 2006;281(9):5406–15.PubMed
364.
go back to reference Nirdé P et al. Heat shock cognate 70 protein secretion as a new growth arrest signal for cancer cells. Oncogene. 2009;29(1):117–27.PubMedCentralPubMed Nirdé P et al. Heat shock cognate 70 protein secretion as a new growth arrest signal for cancer cells. Oncogene. 2009;29(1):117–27.PubMedCentralPubMed
365.
go back to reference Erdmann S et al. Inflammatory cytokines increase extracellular procathepsin D in permanent and primary endothelial cell cultures. Eur J Cell Biol. 2008;87(5):311–23.PubMed Erdmann S et al. Inflammatory cytokines increase extracellular procathepsin D in permanent and primary endothelial cell cultures. Eur J Cell Biol. 2008;87(5):311–23.PubMed
366.
go back to reference Bromme D, Lecaille F. Cathepsin K inhibitors for osteoporosis and potential off-target effects. Expert Opin Investig Drugs. 2009;18(5):585–600.PubMedCentralPubMed Bromme D, Lecaille F. Cathepsin K inhibitors for osteoporosis and potential off-target effects. Expert Opin Investig Drugs. 2009;18(5):585–600.PubMedCentralPubMed
367.
368.
go back to reference Sudhan DR, Siemann DW. Cathepsin L inhibition by the small molecule KGP94 suppresses tumor microenvironment enhanced metastasis associated cell functions of prostate and breast cancer cells. Clin Exp Metastasis. 2013;30(7):891–902.PubMed Sudhan DR, Siemann DW. Cathepsin L inhibition by the small molecule KGP94 suppresses tumor microenvironment enhanced metastasis associated cell functions of prostate and breast cancer cells. Clin Exp Metastasis. 2013;30(7):891–902.PubMed
369.
go back to reference Ward C et al. Antibody targeting of cathepsin S inhibits angiogenesis and synergistically enhances anti-VEGF. PLoS One. 2010;5(9):e12543.PubMedCentralPubMed Ward C et al. Antibody targeting of cathepsin S inhibits angiogenesis and synergistically enhances anti-VEGF. PLoS One. 2010;5(9):e12543.PubMedCentralPubMed
370.
go back to reference Burden RE et al. Inhibition of cathepsin S by Fsn0503 enhances the efficacy of chemotherapy in colorectal carcinomas. Biochimie. 2012;94(2):487–93.PubMed Burden RE et al. Inhibition of cathepsin S by Fsn0503 enhances the efficacy of chemotherapy in colorectal carcinomas. Biochimie. 2012;94(2):487–93.PubMed
371.
go back to reference Dabrosin C, Johansson AC, Ollinger K. Decreased secretion of cathepsin D in breast cancer in vivo by tamoxifen: mediated by the mannose-6-phosphate/IGF-II receptor? Breast Cancer Res Treat. 2004;85(3):229–38.PubMed Dabrosin C, Johansson AC, Ollinger K. Decreased secretion of cathepsin D in breast cancer in vivo by tamoxifen: mediated by the mannose-6-phosphate/IGF-II receptor? Breast Cancer Res Treat. 2004;85(3):229–38.PubMed
372.
go back to reference Vasiljeva O et al. Emerging roles of cysteine cathepsins in disease and their potential as drug targets. Curr Pharm Des. 2007;13(4):387–403.PubMed Vasiljeva O et al. Emerging roles of cysteine cathepsins in disease and their potential as drug targets. Curr Pharm Des. 2007;13(4):387–403.PubMed
373.
go back to reference Elie BT et al. Identification and pre-clinical testing of a reversible cathepsin protease inhibitor reveals anti-tumor efficacy in a pancreatic cancer model. Biochimie. 2010;92(11):1618–24.PubMed Elie BT et al. Identification and pre-clinical testing of a reversible cathepsin protease inhibitor reveals anti-tumor efficacy in a pancreatic cancer model. Biochimie. 2010;92(11):1618–24.PubMed
374.
375.
go back to reference Palermo C, Joyce JA. Cysteine cathepsin proteases as pharmacological targets in cancer. Trends Pharmacol Sci. 2008;29(1):22–8.PubMed Palermo C, Joyce JA. Cysteine cathepsin proteases as pharmacological targets in cancer. Trends Pharmacol Sci. 2008;29(1):22–8.PubMed
376.
go back to reference Mohamed MM, Sloane BF. Cysteine cathepsins: multifunctional enzymes in cancer. Nat Rev Cancer. 2006;6(10):764–75.PubMed Mohamed MM, Sloane BF. Cysteine cathepsins: multifunctional enzymes in cancer. Nat Rev Cancer. 2006;6(10):764–75.PubMed
377.
go back to reference Reiser J, Adair B, Reinheckel T. Specialized roles for cysteine cathepsins in health and disease. J Clin Investig. 2010;120(10):3421–31.PubMedCentralPubMed Reiser J, Adair B, Reinheckel T. Specialized roles for cysteine cathepsins in health and disease. J Clin Investig. 2010;120(10):3421–31.PubMedCentralPubMed
378.
go back to reference Kosa’b J. Lysosomal cathepsins: structure, role in antigen processing and presentation, and cancer. Adv Enzyme Regul. 2002;42:285. Kosa’b J. Lysosomal cathepsins: structure, role in antigen processing and presentation, and cancer. Adv Enzyme Regul. 2002;42:285.
379.
go back to reference Watts C. The endosome–lysosome pathway and information generation in the immune system. Biochimica et Biophysica Acta (BBA)-Proteins and. Proteomics. 2012;1824(1):14–21. Watts C. The endosome–lysosome pathway and information generation in the immune system. Biochimica et Biophysica Acta (BBA)-Proteins and. Proteomics. 2012;1824(1):14–21.
380.
go back to reference Rothberg JM et al. Live-cell imaging of tumor proteolysis: impact of cellular and non-cellular microenvironment. Biochimica et Biophysica Acta (BBA)-Proteins and. Proteomics. 2012;1824(1):123–32. Rothberg JM et al. Live-cell imaging of tumor proteolysis: impact of cellular and non-cellular microenvironment. Biochimica et Biophysica Acta (BBA)-Proteins and. Proteomics. 2012;1824(1):123–32.
381.
go back to reference Dauth S et al. Cathepsin K deficiency in mice induces structural and metabolic changes in the central nervous system that are associated with learning and memory deficits. BMC Neurosci. 2011;12(1):74.PubMedCentralPubMed Dauth S et al. Cathepsin K deficiency in mice induces structural and metabolic changes in the central nervous system that are associated with learning and memory deficits. BMC Neurosci. 2011;12(1):74.PubMedCentralPubMed
382.
go back to reference Jedeszko C, Sloane BF. Cysteine cathepsins in human cancer. Biol Chem. 2004;385(11):1017–27.PubMed Jedeszko C, Sloane BF. Cysteine cathepsins in human cancer. Biol Chem. 2004;385(11):1017–27.PubMed
383.
go back to reference Berdowska I. Cysteine proteases as disease markers. Clin Chim Acta. 2004;342(1):41–69.PubMed Berdowska I. Cysteine proteases as disease markers. Clin Chim Acta. 2004;342(1):41–69.PubMed
384.
go back to reference Arvatz G et al. The heparanase system and tumor metastasis: is heparanase the seed and soil? Cancer Metastasis Rev. 2011;30(2):253–68.PubMed Arvatz G et al. The heparanase system and tumor metastasis: is heparanase the seed and soil? Cancer Metastasis Rev. 2011;30(2):253–68.PubMed
385.
go back to reference Green KA, Lund LR. ECM degrading proteases and tissue remodelling in the mammary gland. Bioessays. 2005;27(9):894–903.PubMed Green KA, Lund LR. ECM degrading proteases and tissue remodelling in the mammary gland. Bioessays. 2005;27(9):894–903.PubMed
387.
go back to reference Reinheckel T et al. Differential impact of cysteine cathepsins on genetic mouse models of de novo carcinogenesis: cathepsin B as emerging therapeutic target. Front Pharmacol. 2012;3:133.PubMedCentralPubMed Reinheckel T et al. Differential impact of cysteine cathepsins on genetic mouse models of de novo carcinogenesis: cathepsin B as emerging therapeutic target. Front Pharmacol. 2012;3:133.PubMedCentralPubMed
388.
go back to reference Mitchell BS. The proteasome—an emerging therapeutic target in cancer. N Engl J Med. 2003;348(26):2597–8.PubMed Mitchell BS. The proteasome—an emerging therapeutic target in cancer. N Engl J Med. 2003;348(26):2597–8.PubMed
389.
go back to reference Kane RC et al. Velcade®: US FDA approval for the treatment of multiple myeloma progressing on prior therapy. Oncologist. 2003;8(6):508–13.PubMed Kane RC et al. Velcade®: US FDA approval for the treatment of multiple myeloma progressing on prior therapy. Oncologist. 2003;8(6):508–13.PubMed
390.
go back to reference Zavrski I et al. Proteasome: an emerging target for cancer therapy. Anti-Cancer Drugs. 2005;16(5):475–81.PubMed Zavrski I et al. Proteasome: an emerging target for cancer therapy. Anti-Cancer Drugs. 2005;16(5):475–81.PubMed
391.
go back to reference Kane RC et al. Bortezomib for the treatment of mantle cell lymphoma. Clin Cancer Res. 2007;13(18):5291–4.PubMed Kane RC et al. Bortezomib for the treatment of mantle cell lymphoma. Clin Cancer Res. 2007;13(18):5291–4.PubMed
392.
go back to reference Chen D et al. Bortezomib as the first proteasome inhibitor anticancer drug: current status and future perspectives. Curr Cancer Drug Targets. 2011;11(3):239.PubMedCentralPubMed Chen D et al. Bortezomib as the first proteasome inhibitor anticancer drug: current status and future perspectives. Curr Cancer Drug Targets. 2011;11(3):239.PubMedCentralPubMed
393.
go back to reference Berenson JR et al. Phase I/II trial assessing bortezomib and melphalan combination therapy for the treatment of patients with relapsed or refractory multiple myeloma. J Clin Oncol. 2006;24(6):937–44.PubMed Berenson JR et al. Phase I/II trial assessing bortezomib and melphalan combination therapy for the treatment of patients with relapsed or refractory multiple myeloma. J Clin Oncol. 2006;24(6):937–44.PubMed
394.
go back to reference Kane RC et al. United States Food and Drug Administration approval summary: bortezomib for the treatment of progressive multiple myeloma after one prior therapy. Clin Cancer Res. 2006;12(10):2955–60.PubMed Kane RC et al. United States Food and Drug Administration approval summary: bortezomib for the treatment of progressive multiple myeloma after one prior therapy. Clin Cancer Res. 2006;12(10):2955–60.PubMed
395.
go back to reference Liotta LA, Kohn EC. The microenvironment of the tumour–host interface. Nature. 2001;411(6835):375–9.PubMed Liotta LA, Kohn EC. The microenvironment of the tumour–host interface. Nature. 2001;411(6835):375–9.PubMed
396.
go back to reference Hughes-Alford SK, Lauffenburger DA. Quantitative analysis of gradient sensing: towards building predictive models of chemotaxis in cancer. Curr Opin Cell Biol. 2012;24(2):284–91.PubMedCentralPubMed Hughes-Alford SK, Lauffenburger DA. Quantitative analysis of gradient sensing: towards building predictive models of chemotaxis in cancer. Curr Opin Cell Biol. 2012;24(2):284–91.PubMedCentralPubMed
397.
go back to reference Onuffer JJ, Horuk R. Chemokines, chemokine receptors and small-molecule antagonists: recent developments. Trends Pharmacol Sci. 2002;23(10):459–67.PubMed Onuffer JJ, Horuk R. Chemokines, chemokine receptors and small-molecule antagonists: recent developments. Trends Pharmacol Sci. 2002;23(10):459–67.PubMed
398.
go back to reference Ebert LM, Schaerli P, Moser B. Chemokine-mediated control of T cell traffic in lymphoid and peripheral tissues. Mol Immunol. 2005;42(7):799–809.PubMed Ebert LM, Schaerli P, Moser B. Chemokine-mediated control of T cell traffic in lymphoid and peripheral tissues. Mol Immunol. 2005;42(7):799–809.PubMed
399.
go back to reference Kedrin D et al. Cell motility and cytoskeletal regulation in invasion and metastasis. J Mammary Gland Biol Neoplasia. 2007;12(2–3):143–52.PubMed Kedrin D et al. Cell motility and cytoskeletal regulation in invasion and metastasis. J Mammary Gland Biol Neoplasia. 2007;12(2–3):143–52.PubMed
400.
401.
go back to reference Wells A et al. Targeting tumor cell motility as a strategy against invasion and metastasis. Trends Pharmacol Sci. 2013;34(5):283–9.PubMedCentralPubMed Wells A et al. Targeting tumor cell motility as a strategy against invasion and metastasis. Trends Pharmacol Sci. 2013;34(5):283–9.PubMedCentralPubMed
402.
go back to reference do Carmo A et al. CXCL12/CXCR4 promotes motility and proliferation of glioma cells. Cancer Biol Ther. 2010;9(1):p. 56–65. do Carmo A et al. CXCL12/CXCR4 promotes motility and proliferation of glioma cells. Cancer Biol Ther. 2010;9(1):p. 56–65.
403.
go back to reference Dai X et al. The CXCL12/CXCR4 autocrine loop increases the metastatic potential of non-small cell lung cancer in vitro. Oncol Lett. 2013;5(1):277–82.PubMedCentralPubMed Dai X et al. The CXCL12/CXCR4 autocrine loop increases the metastatic potential of non-small cell lung cancer in vitro. Oncol Lett. 2013;5(1):277–82.PubMedCentralPubMed
404.
go back to reference Zlotnik A. Chemokines and cancer. Int J Cancer. 2006;119(9):2026–9.PubMed Zlotnik A. Chemokines and cancer. Int J Cancer. 2006;119(9):2026–9.PubMed
405.
go back to reference Dewan M et al. Stromal cell-derived factor-1 and CXCR4 receptor interaction in tumor growth and metastasis of breast cancer. Biomed Pharmacother. 2006;60(6):273–6.PubMed Dewan M et al. Stromal cell-derived factor-1 and CXCR4 receptor interaction in tumor growth and metastasis of breast cancer. Biomed Pharmacother. 2006;60(6):273–6.PubMed
406.
go back to reference Kato M et al. Expression pattern of CXC chemokine receptor-4 is correlated with lymph node metastasis in human invasive ductal carcinoma. Breast Cancer Res. 2003;5(5):R144–50.PubMedCentralPubMed Kato M et al. Expression pattern of CXC chemokine receptor-4 is correlated with lymph node metastasis in human invasive ductal carcinoma. Breast Cancer Res. 2003;5(5):R144–50.PubMedCentralPubMed
407.
go back to reference Riese DJ, Stern DF. Specificity within the EGF family/ErbB receptor family signaling network. Bioessays. 1998;20(1):41–8.PubMed Riese DJ, Stern DF. Specificity within the EGF family/ErbB receptor family signaling network. Bioessays. 1998;20(1):41–8.PubMed
408.
go back to reference Xue C et al. Epidermal growth factor receptor overexpression results in increased tumor cell motility in vivo coordinately with enhanced intravasation and metastasis. Cancer Res. 2006;66(1):192–7.PubMed Xue C et al. Epidermal growth factor receptor overexpression results in increased tumor cell motility in vivo coordinately with enhanced intravasation and metastasis. Cancer Res. 2006;66(1):192–7.PubMed
409.
go back to reference Ma PC et al. c-Met: structure, functions and potential for therapeutic inhibition. Cancer Metastasis Rev. 2003;22(4):309–25.PubMed Ma PC et al. c-Met: structure, functions and potential for therapeutic inhibition. Cancer Metastasis Rev. 2003;22(4):309–25.PubMed
410.
go back to reference Micke P. Tumour-stroma interaction: cancer-associated fibroblasts as novel targets in anti-cancer therapy? Lung Cancer. 2004;45:S163–75.PubMed Micke P. Tumour-stroma interaction: cancer-associated fibroblasts as novel targets in anti-cancer therapy? Lung Cancer. 2004;45:S163–75.PubMed
411.
go back to reference Qian L-W et al. Co-cultivation of pancreatic cancer cells with orthotopic tumor-derived fibroblasts: fibroblasts stimulate tumor cell invasion via HGF secretion whereas cancer cells exert a minor regulative effect on fibroblasts HGF production. Cancer Lett. 2003;190(1):105–12.PubMed Qian L-W et al. Co-cultivation of pancreatic cancer cells with orthotopic tumor-derived fibroblasts: fibroblasts stimulate tumor cell invasion via HGF secretion whereas cancer cells exert a minor regulative effect on fibroblasts HGF production. Cancer Lett. 2003;190(1):105–12.PubMed
412.
go back to reference Luker KE, Luker GD. Functions of CXCL12 and CXCR4 in breast cancer. Cancer Lett. 2006;238(1):30–41.PubMed Luker KE, Luker GD. Functions of CXCL12 and CXCR4 in breast cancer. Cancer Lett. 2006;238(1):30–41.PubMed
413.
go back to reference Burger JA, Kipps TJ. CXCR4: a key receptor in the crosstalk between tumor cells and their microenvironment. Blood. 2006;107(5):1761–7.PubMed Burger JA, Kipps TJ. CXCR4: a key receptor in the crosstalk between tumor cells and their microenvironment. Blood. 2006;107(5):1761–7.PubMed
414.
go back to reference Takebe N et al. Review of cancer-associated fibroblasts and therapies that interfere with their activity. Tumor Microenvironment Ther. 2013;1:19–36. Takebe N et al. Review of cancer-associated fibroblasts and therapies that interfere with their activity. Tumor Microenvironment Ther. 2013;1:19–36.
415.
go back to reference Yin HL, Janmey PA. Phosphoinositide regulation of the actin cytoskeleton. Annu Rev Physiol. 2003;65(1):761–89.PubMed Yin HL, Janmey PA. Phosphoinositide regulation of the actin cytoskeleton. Annu Rev Physiol. 2003;65(1):761–89.PubMed
416.
go back to reference Hurley JH. Membrane binding domains. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of. Lipids. 2006;1761(8):805–11. Hurley JH. Membrane binding domains. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of. Lipids. 2006;1761(8):805–11.
417.
go back to reference Barber MA, Welch HC. PI3K and RAC signalling in leukocyte and cancer cell migration. Bull Cancer. 2006;93(5):10044–52. Barber MA, Welch HC. PI3K and RAC signalling in leukocyte and cancer cell migration. Bull Cancer. 2006;93(5):10044–52.
418.
go back to reference Hall A. Rho GTPases and the control of cell behaviour. Biochem Soc Trans. 2005;33:891–5.PubMed Hall A. Rho GTPases and the control of cell behaviour. Biochem Soc Trans. 2005;33:891–5.PubMed
419.
go back to reference Bravo-Cordero JJ et al. A novel spatiotemporal RhoC activation pathway locally regulates cofilin activity at invadopodia. Curr Biol. 2011;21(8):635–44.PubMedCentralPubMed Bravo-Cordero JJ et al. A novel spatiotemporal RhoC activation pathway locally regulates cofilin activity at invadopodia. Curr Biol. 2011;21(8):635–44.PubMedCentralPubMed
420.
go back to reference Pollard TD, Borisy GG. Cellular motility driven by assembly and disassembly of actin filaments. Cell. 2003;112(4):453–65.PubMed Pollard TD, Borisy GG. Cellular motility driven by assembly and disassembly of actin filaments. Cell. 2003;112(4):453–65.PubMed
421.
go back to reference Mouneimne G et al. Phospholipase C and cofilin are required for carcinoma cell directionality in response to EGF stimulation. J Cell Biol. 2004;166(5):697–708.PubMedCentralPubMed Mouneimne G et al. Phospholipase C and cofilin are required for carcinoma cell directionality in response to EGF stimulation. J Cell Biol. 2004;166(5):697–708.PubMedCentralPubMed
422.
go back to reference Wilkinson S, Paterson HF, Marshall CJ. Cdc42–MRCK and Rho–ROCK signalling cooperate in myosin phosphorylation and cell invasion. Nat Cell Biol. 2005;7(3):255–61.PubMed Wilkinson S, Paterson HF, Marshall CJ. Cdc42–MRCK and Rho–ROCK signalling cooperate in myosin phosphorylation and cell invasion. Nat Cell Biol. 2005;7(3):255–61.PubMed
423.
go back to reference Kopp F et al. Salinomycin treatment reduces metastatic tumor burden by hampering cancer cell migration. Mol Cancer. 2014;13:16.PubMedCentralPubMed Kopp F et al. Salinomycin treatment reduces metastatic tumor burden by hampering cancer cell migration. Mol Cancer. 2014;13:16.PubMedCentralPubMed
424.
go back to reference Kumar P et al. Tetrathiomolybdate inhibits head and neck cancer metastasis by decreasing tumor cell motility, invasiveness and by promoting tumor cell anoikis. Mol Cancer. 2010;9:206.PubMedCentralPubMed Kumar P et al. Tetrathiomolybdate inhibits head and neck cancer metastasis by decreasing tumor cell motility, invasiveness and by promoting tumor cell anoikis. Mol Cancer. 2010;9:206.PubMedCentralPubMed
425.
go back to reference Palmer TD et al. Integrin-free tetraspanin CD151 can inhibit tumor cell motility upon clustering and is a clinical indicator of prostate cancer progression. Cancer Res. 2014;74(1):173–87.PubMedCentralPubMed Palmer TD et al. Integrin-free tetraspanin CD151 can inhibit tumor cell motility upon clustering and is a clinical indicator of prostate cancer progression. Cancer Res. 2014;74(1):173–87.PubMedCentralPubMed
426.
go back to reference Wang S et al. Study on invadopodia formation for lung carcinoma invasion with a microfluidic 3D culture device. PLoS One. 2013;8(2):e56448.PubMedCentralPubMed Wang S et al. Study on invadopodia formation for lung carcinoma invasion with a microfluidic 3D culture device. PLoS One. 2013;8(2):e56448.PubMedCentralPubMed
427.
go back to reference Wolf K et al. Compensation mechanism in tumor cell migration mesenchymal–amoeboid transition after blocking of pericellular proteolysis. J Cell Biol. 2003;160(2):267–77.PubMedCentralPubMed Wolf K et al. Compensation mechanism in tumor cell migration mesenchymal–amoeboid transition after blocking of pericellular proteolysis. J Cell Biol. 2003;160(2):267–77.PubMedCentralPubMed
428.
go back to reference Thiery JP et al. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139(5):871–90.PubMed Thiery JP et al. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139(5):871–90.PubMed
429.
go back to reference Bravo-Cordero JJ, Hodgson L, Condeelis J. Directed cell invasion and migration during metastasis. Curr Opin Cell Biol. 2012;24(2):277–83.PubMedCentralPubMed Bravo-Cordero JJ, Hodgson L, Condeelis J. Directed cell invasion and migration during metastasis. Curr Opin Cell Biol. 2012;24(2):277–83.PubMedCentralPubMed
430.
go back to reference Wang W et al. Coordinated regulation of pathways for enhanced cell motility and chemotaxis is conserved in rat and mouse mammary tumors. Cancer Res. 2007;67(8):3505–11.PubMed Wang W et al. Coordinated regulation of pathways for enhanced cell motility and chemotaxis is conserved in rat and mouse mammary tumors. Cancer Res. 2007;67(8):3505–11.PubMed
431.
go back to reference Olson MF, Sahai E. The actin cytoskeleton in cancer cell motility. Clin Exp Metastasis. 2009;26(4):273–87.PubMed Olson MF, Sahai E. The actin cytoskeleton in cancer cell motility. Clin Exp Metastasis. 2009;26(4):273–87.PubMed
432.
go back to reference Vignjevic, D. and Montagnac G. Reorganisation of the dendritic actin network during cancer cell migration and invasion. in Seminars in cancer biology. 2008. Elsevier. Vignjevic, D. and Montagnac G. Reorganisation of the dendritic actin network during cancer cell migration and invasion. in Seminars in cancer biology. 2008. Elsevier.
433.
go back to reference Buccione R, Caldieri G, Ayala I. Invadopodia: specialized tumor cell structures for the focal degradation of the extracellular matrix. Cancer Metastasis Rev. 2009;28(1–2):137–49.PubMed Buccione R, Caldieri G, Ayala I. Invadopodia: specialized tumor cell structures for the focal degradation of the extracellular matrix. Cancer Metastasis Rev. 2009;28(1–2):137–49.PubMed
434.
go back to reference Yilmaz M, Christofori G. Mechanisms of motility in metastasizing cells. Mol Cancer Res. 2010;8(5):629–42.PubMed Yilmaz M, Christofori G. Mechanisms of motility in metastasizing cells. Mol Cancer Res. 2010;8(5):629–42.PubMed
435.
go back to reference Condeelis J.S. et al. Lamellipodia in invasion. in Seminars in cancer biology. 2001. Elsevier. Condeelis J.S. et al. Lamellipodia in invasion. in Seminars in cancer biology. 2001. Elsevier.
436.
go back to reference Wang W et al. Single cell behavior in metastatic primary mammary tumors correlated with gene expression patterns revealed by molecular profiling. Cancer Res. 2002;62(21):6278–88.PubMed Wang W et al. Single cell behavior in metastatic primary mammary tumors correlated with gene expression patterns revealed by molecular profiling. Cancer Res. 2002;62(21):6278–88.PubMed
437.
go back to reference Vignjevic D et al. Fascin, a novel target of beta-catenin-TCF signaling, is expressed at the invasive front of human colon cancer. Cancer Res. 2007;67(14):6844–53.PubMed Vignjevic D et al. Fascin, a novel target of beta-catenin-TCF signaling, is expressed at the invasive front of human colon cancer. Cancer Res. 2007;67(14):6844–53.PubMed
438.
go back to reference Mongiu AK et al. Kinetic-structural analysis of neuronal growth cone veil motility. J Cell Sci. 2007;120(6):1113–25.PubMed Mongiu AK et al. Kinetic-structural analysis of neuronal growth cone veil motility. J Cell Sci. 2007;120(6):1113–25.PubMed
439.
go back to reference Yilmaz M, Christofori G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009;28(1–2):15–33.PubMed Yilmaz M, Christofori G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009;28(1–2):15–33.PubMed
440.
go back to reference Khoury H et al. Distinct tyrosine autophosphorylation sites mediate induction of epithelial mesenchymal like transition by an activated ErbB-2/Neu receptor. Oncogene. 2001;20(7):788–99.PubMed Khoury H et al. Distinct tyrosine autophosphorylation sites mediate induction of epithelial mesenchymal like transition by an activated ErbB-2/Neu receptor. Oncogene. 2001;20(7):788–99.PubMed
441.
442.
go back to reference Caldieri G et al. Invadopodia biogenesis is regulated by caveolin‐mediated modulation of membrane cholesterol levels. J Cell Mol Med. 2009;13(8b):1728–40.PubMed Caldieri G et al. Invadopodia biogenesis is regulated by caveolin‐mediated modulation of membrane cholesterol levels. J Cell Mol Med. 2009;13(8b):1728–40.PubMed
443.
go back to reference Kelly T et al. Proteolysis of extracellular matrix by invadopodia facilitates human breast cancer cell invasion and is mediated by matrix metalloproteinases. Clin Exp Metastasis. 1998;16(6):501–12.PubMed Kelly T et al. Proteolysis of extracellular matrix by invadopodia facilitates human breast cancer cell invasion and is mediated by matrix metalloproteinases. Clin Exp Metastasis. 1998;16(6):501–12.PubMed
444.
go back to reference Tague SE, Muralidharan V, D’Souza-Schorey C. ADP-ribosylation factor 6 regulates tumor cell invasion through the activation of the MEK/ERK signaling pathway. Proc Natl Acad Sci U S A. 2004;101(26):9671–6.PubMedCentralPubMed Tague SE, Muralidharan V, D’Souza-Schorey C. ADP-ribosylation factor 6 regulates tumor cell invasion through the activation of the MEK/ERK signaling pathway. Proc Natl Acad Sci U S A. 2004;101(26):9671–6.PubMedCentralPubMed
445.
go back to reference Oxmann D et al. Endoglin expression in metastatic breast cancer cells enhances their invasive phenotype. Oncogene. 2008;27(25):3567–75.PubMed Oxmann D et al. Endoglin expression in metastatic breast cancer cells enhances their invasive phenotype. Oncogene. 2008;27(25):3567–75.PubMed
446.
go back to reference Clark ES et al. Cortactin is an essential regulator of matrix metalloproteinase secretion and extracellular matrix degradation in invadopodia. Cancer Res. 2007;67(9):4227–35.PubMed Clark ES et al. Cortactin is an essential regulator of matrix metalloproteinase secretion and extracellular matrix degradation in invadopodia. Cancer Res. 2007;67(9):4227–35.PubMed
447.
go back to reference Artym VV et al. Dynamic interactions of cortactin and membrane type 1 matrix metalloproteinase at invadopodia: defining the stages of invadopodia formation and function. Cancer Res. 2006;66(6):3034–43.PubMed Artym VV et al. Dynamic interactions of cortactin and membrane type 1 matrix metalloproteinase at invadopodia: defining the stages of invadopodia formation and function. Cancer Res. 2006;66(6):3034–43.PubMed
448.
go back to reference Steeg PS. Tumor metastasis: mechanistic insights and clinical challenges. Nat Med. 2006;12(8):895–904.PubMed Steeg PS. Tumor metastasis: mechanistic insights and clinical challenges. Nat Med. 2006;12(8):895–904.PubMed
449.
go back to reference Zervantonakis IK et al. Three-dimensional microfluidic model for tumor cell intravasation and endothelial barrier function. Proc Natl Acad Sci U S A. 2012;109(34):13515–20.PubMedCentralPubMed Zervantonakis IK et al. Three-dimensional microfluidic model for tumor cell intravasation and endothelial barrier function. Proc Natl Acad Sci U S A. 2012;109(34):13515–20.PubMedCentralPubMed
450.
go back to reference Quigley JP, Armstrong PB. Tumor cell intravasation Alu-cidated: the chick embryo opens the window. Cell. 1998;94(3):281–4.PubMed Quigley JP, Armstrong PB. Tumor cell intravasation Alu-cidated: the chick embryo opens the window. Cell. 1998;94(3):281–4.PubMed
451.
go back to reference Wirtz D. Particle-tracking microrheology of living cells: principles and applications. Annu Rev Biophys. 2009;38:301–26.PubMed Wirtz D. Particle-tracking microrheology of living cells: principles and applications. Annu Rev Biophys. 2009;38:301–26.PubMed
452.
go back to reference Yeung T et al. Effects of substrate stiffness on cell morphology, cytoskeletal structure, and adhesion. Cell Mot Cytoskeleton. 2005;60(1):24–34. Yeung T et al. Effects of substrate stiffness on cell morphology, cytoskeletal structure, and adhesion. Cell Mot Cytoskeleton. 2005;60(1):24–34.
453.
go back to reference Baker EL, Bonnecaze RT, Zaman MH. Extracellular matrix stiffness and architecture govern intracellular rheology in cancer. Biophys J. 2009;97(4):1013–21.PubMedCentralPubMed Baker EL, Bonnecaze RT, Zaman MH. Extracellular matrix stiffness and architecture govern intracellular rheology in cancer. Biophys J. 2009;97(4):1013–21.PubMedCentralPubMed
454.
go back to reference Baker EL et al. Cancer cell stiffness: integrated roles of three-dimensional matrix stiffness and transforming potential. Biophys J. 2010;99(7):2048–57.PubMedCentralPubMed Baker EL et al. Cancer cell stiffness: integrated roles of three-dimensional matrix stiffness and transforming potential. Biophys J. 2010;99(7):2048–57.PubMedCentralPubMed
455.
go back to reference Lee JS et al. Ballistic intracellular nanorheology reveals ROCK-hard cytoplasmic stiffening response to fluid flow. J Cell Sci. 2006;119(9):1760–8.PubMed Lee JS et al. Ballistic intracellular nanorheology reveals ROCK-hard cytoplasmic stiffening response to fluid flow. J Cell Sci. 2006;119(9):1760–8.PubMed
456.
go back to reference Cross SE et al. Nanomechanical analysis of cells from cancer patients. Nat Nanotechnol. 2007;2(12):780–3.PubMed Cross SE et al. Nanomechanical analysis of cells from cancer patients. Nat Nanotechnol. 2007;2(12):780–3.PubMed
457.
go back to reference Swartz MA, Fleury ME. Interstitial flow and its effects in soft tissues. Annu Rev Biomed Eng. 2007;9:229–56.PubMed Swartz MA, Fleury ME. Interstitial flow and its effects in soft tissues. Annu Rev Biomed Eng. 2007;9:229–56.PubMed
458.
go back to reference Mycielska ME, Djamgoz MB. Cellular mechanisms of direct-current electric field effects: galvanotaxis and metastatic disease. J Cell Sci. 2004;117(9):1631–9.PubMed Mycielska ME, Djamgoz MB. Cellular mechanisms of direct-current electric field effects: galvanotaxis and metastatic disease. J Cell Sci. 2004;117(9):1631–9.PubMed
459.
go back to reference Wirtz D, Konstantopoulos K, Searson PC. The physics of cancer: the role of physical interactions and mechanical forces in metastasis. Nat Rev Cancer. 2011;11(7):512–22.PubMedCentralPubMed Wirtz D, Konstantopoulos K, Searson PC. The physics of cancer: the role of physical interactions and mechanical forces in metastasis. Nat Rev Cancer. 2011;11(7):512–22.PubMedCentralPubMed
460.
go back to reference Roussos ET et al. Mena invasive (MenaINV) promotes multicellular streaming motility and transendothelial migration in a mouse model of breast cancer. J Cell Sci. 2011;124(13):2120–31.PubMedCentralPubMed Roussos ET et al. Mena invasive (MenaINV) promotes multicellular streaming motility and transendothelial migration in a mouse model of breast cancer. J Cell Sci. 2011;124(13):2120–31.PubMedCentralPubMed
461.
go back to reference Calvo F, Sahai E. Cell communication networks in cancer invasion. Curr Opin Cell Biol. 2011;23(5):621–9.PubMed Calvo F, Sahai E. Cell communication networks in cancer invasion. Curr Opin Cell Biol. 2011;23(5):621–9.PubMed
462.
go back to reference Giampieri S et al. Localized and reversible TGFβ signalling switches breast cancer cells from cohesive to single cell motility. Nat Cell Biol. 2009;11(11):1287–96.PubMedCentralPubMed Giampieri S et al. Localized and reversible TGFβ signalling switches breast cancer cells from cohesive to single cell motility. Nat Cell Biol. 2009;11(11):1287–96.PubMedCentralPubMed
463.
go back to reference Zijlstra A et al. The inhibition of tumor cell intravasation and subsequent metastasis via regulation of in vivo tumor cell motility by the tetraspanin CD151. Cancer Cell. 2008;13(3):221–34.PubMedCentralPubMed Zijlstra A et al. The inhibition of tumor cell intravasation and subsequent metastasis via regulation of in vivo tumor cell motility by the tetraspanin CD151. Cancer Cell. 2008;13(3):221–34.PubMedCentralPubMed
464.
go back to reference Wyckoff JB et al. Direct visualization of macrophage-assisted tumor cell intravasation in mammary tumors. Cancer Res. 2007;67(6):2649–56.PubMed Wyckoff JB et al. Direct visualization of macrophage-assisted tumor cell intravasation in mammary tumors. Cancer Res. 2007;67(6):2649–56.PubMed
465.
go back to reference Bekes EM et al. Tumor-recruited neutrophils and neutrophil TIMP-free MMP-9 regulate coordinately the levels of tumor angiogenesis and efficiency of malignant cell intravasation. Am J Pathol. 2011;179(3):1455–70.PubMedCentralPubMed Bekes EM et al. Tumor-recruited neutrophils and neutrophil TIMP-free MMP-9 regulate coordinately the levels of tumor angiogenesis and efficiency of malignant cell intravasation. Am J Pathol. 2011;179(3):1455–70.PubMedCentralPubMed
466.
go back to reference Weis S et al. Endothelial barrier disruption by VEGF-mediated Src activity potentiates tumor cell extravasation and metastasis. J Cell Biol. 2004;167(2):223–9.PubMedCentralPubMed Weis S et al. Endothelial barrier disruption by VEGF-mediated Src activity potentiates tumor cell extravasation and metastasis. J Cell Biol. 2004;167(2):223–9.PubMedCentralPubMed
467.
go back to reference Bockhorn M, Jain RK, Munn LL. Active versus passive mechanisms in metastasis: do cancer cells crawl into vessels, or are they pushed? Lancet Oncol. 2007;8(5):444–8.PubMedCentralPubMed Bockhorn M, Jain RK, Munn LL. Active versus passive mechanisms in metastasis: do cancer cells crawl into vessels, or are they pushed? Lancet Oncol. 2007;8(5):444–8.PubMedCentralPubMed
468.
go back to reference Wong SY, Hynes RO. Lymphatic or hematogenous dissemination: how does a metastatic tumor cell decide? Cell Cycle (Georgetown, Tex). 2006;5(8):812. Wong SY, Hynes RO. Lymphatic or hematogenous dissemination: how does a metastatic tumor cell decide? Cell Cycle (Georgetown, Tex). 2006;5(8):812.
469.
go back to reference Bockhorn M et al. Differential gene expression in metastasizing cells shed from kidney tumors. Cancer Res. 2004;64(7):2469–73.PubMed Bockhorn M et al. Differential gene expression in metastasizing cells shed from kidney tumors. Cancer Res. 2004;64(7):2469–73.PubMed
470.
go back to reference Blood CH, Zetter BR. Tumor interactions with the vasculature: angiogenesis and tumor metastasis. Biochimica et Biophysica Acta (BBA)-Reviews on. Cancer. 1990;1032(1):89–118. Blood CH, Zetter BR. Tumor interactions with the vasculature: angiogenesis and tumor metastasis. Biochimica et Biophysica Acta (BBA)-Reviews on. Cancer. 1990;1032(1):89–118.
471.
go back to reference Nash G et al. Platelets and cancer. Lancet Oncol. 2002;3(7):425–30.PubMed Nash G et al. Platelets and cancer. Lancet Oncol. 2002;3(7):425–30.PubMed
473.
go back to reference Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5(12):953–64.PubMed Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5(12):953–64.PubMed
474.
go back to reference Qian B-Z, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell. 2010;141(1):39–51.PubMed Qian B-Z, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell. 2010;141(1):39–51.PubMed
475.
go back to reference Nathan C. Metchnikoff’s legacy in 2008. Nat Immunol. 2008;9(7):695–8.PubMed Nathan C. Metchnikoff’s legacy in 2008. Nat Immunol. 2008;9(7):695–8.PubMed
476.
go back to reference Hao N-B et al. Macrophages in tumor microenvironments and the progression of tumors. Clin Dev Immunol. 2012;2012:11. Hao N-B et al. Macrophages in tumor microenvironments and the progression of tumors. Clin Dev Immunol. 2012;2012:11.
477.
478.
go back to reference Condeelis J, Pollard JW. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell. 2006;124(2):263–6.PubMed Condeelis J, Pollard JW. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell. 2006;124(2):263–6.PubMed
480.
go back to reference Goswami S et al. Macrophages promote the invasion of breast carcinoma cells via a colony-stimulating factor-1/epidermal growth factor paracrine loop. Cancer Res. 2005;65(12):5278–83.PubMed Goswami S et al. Macrophages promote the invasion of breast carcinoma cells via a colony-stimulating factor-1/epidermal growth factor paracrine loop. Cancer Res. 2005;65(12):5278–83.PubMed
481.
go back to reference Wyckoff J et al. A paracrine loop between tumor cells and macrophages is required for tumor cell migration in mammary tumors. Cancer Res. 2004;64(19):7022–9.PubMed Wyckoff J et al. A paracrine loop between tumor cells and macrophages is required for tumor cell migration in mammary tumors. Cancer Res. 2004;64(19):7022–9.PubMed
482.
go back to reference Laufs S, Schumacher J, Allgayer H. Urokinase-receptor (u-PAR): an essential player in multiple games of cancer: a review on its role in tumor progression, invasion, metastasis, proliferation/dormancy, clinical outcome and minimal residual disease. Cell Cycle (Georgetown, Tex). 2006;5(16):1760–71. Laufs S, Schumacher J, Allgayer H. Urokinase-receptor (u-PAR): an essential player in multiple games of cancer: a review on its role in tumor progression, invasion, metastasis, proliferation/dormancy, clinical outcome and minimal residual disease. Cell Cycle (Georgetown, Tex). 2006;5(16):1760–71.
483.
go back to reference Kitamura T et al. SMAD4-deficient intestinal tumors recruit CCR1+ myeloid cells that promote invasion. Nat Genet. 2007;39(4):467–75.PubMed Kitamura T et al. SMAD4-deficient intestinal tumors recruit CCR1+ myeloid cells that promote invasion. Nat Genet. 2007;39(4):467–75.PubMed
484.
go back to reference van Kempen LC, de Visser KE, Coussens LM. Inflammation, proteases and cancer. Eur J Cancer. 2006;42(6):728–34.PubMed van Kempen LC, de Visser KE, Coussens LM. Inflammation, proteases and cancer. Eur J Cancer. 2006;42(6):728–34.PubMed
485.
go back to reference Cheng K, Xie G, Raufman J-P. Matrix metalloproteinase-7-catalyzed release of HB-EGF mediates deoxycholyltaurine-induced proliferation of a human colon cancer cell line. Biochem Pharmacol. 2007;73(7):1001–12.PubMedCentralPubMed Cheng K, Xie G, Raufman J-P. Matrix metalloproteinase-7-catalyzed release of HB-EGF mediates deoxycholyltaurine-induced proliferation of a human colon cancer cell line. Biochem Pharmacol. 2007;73(7):1001–12.PubMedCentralPubMed
486.
go back to reference Lin EY, Pollard JW. Tumor-associated macrophages press the angiogenic switch in breast cancer. Cancer Res. 2007;67(11):5064–6.PubMed Lin EY, Pollard JW. Tumor-associated macrophages press the angiogenic switch in breast cancer. Cancer Res. 2007;67(11):5064–6.PubMed
487.
go back to reference Tsutsui S et al. Macrophage infiltration and its prognostic implications in breast cancer: the relationship with VEGF expression and microvessel density. Oncol Rep. 2005;14(2):425–31.PubMed Tsutsui S et al. Macrophage infiltration and its prognostic implications in breast cancer: the relationship with VEGF expression and microvessel density. Oncol Rep. 2005;14(2):425–31.PubMed
488.
go back to reference Bolat F et al. Microvessel density, VEGF expression, and tumor-associated macrophages in breast tumors: correlations with prognostic parameters. Vascular. 2006;14:15. Bolat F et al. Microvessel density, VEGF expression, and tumor-associated macrophages in breast tumors: correlations with prognostic parameters. Vascular. 2006;14:15.
489.
go back to reference Oosterling SJ et al. Macrophages direct tumour histology and clinical outcome in a colon cancer model. J Pathol. 2005;207(2):147–55.PubMed Oosterling SJ et al. Macrophages direct tumour histology and clinical outcome in a colon cancer model. J Pathol. 2005;207(2):147–55.PubMed
490.
go back to reference DeNardo DG, Johansson M, Coussens LM. Immune cells as mediators of solid tumor metastasis. Cancer Metastasis Rev. 2008;27(1):11–8.PubMed DeNardo DG, Johansson M, Coussens LM. Immune cells as mediators of solid tumor metastasis. Cancer Metastasis Rev. 2008;27(1):11–8.PubMed
491.
go back to reference Wang W et al. Tumor cells caught in the act of invading: their strategy for enhanced cell motility. Trends Cell Biol. 2005;15(3):138–45.PubMed Wang W et al. Tumor cells caught in the act of invading: their strategy for enhanced cell motility. Trends Cell Biol. 2005;15(3):138–45.PubMed
492.
go back to reference Condeelis J, Singer RH, Segall JE. The great escape: when cancer cells hijack the genes for chemotaxis and motility. Annu Rev Cell Dev Biol. 2005;21:695–718.PubMed Condeelis J, Singer RH, Segall JE. The great escape: when cancer cells hijack the genes for chemotaxis and motility. Annu Rev Cell Dev Biol. 2005;21:695–718.PubMed
493.
go back to reference Yamaguchi H, Pixley F, Condeelis J. Invadopodia and podosomes in tumor invasion. Eur J Cell Biol. 2006;85(3):213–8.PubMed Yamaguchi H, Pixley F, Condeelis J. Invadopodia and podosomes in tumor invasion. Eur J Cell Biol. 2006;85(3):213–8.PubMed
494.
go back to reference Leek RD, Harris AL. Tumor-associated macrophages in breast cancer. J Mammary Gland Biol Neoplasia. 2002;7(2):177–89.PubMed Leek RD, Harris AL. Tumor-associated macrophages in breast cancer. J Mammary Gland Biol Neoplasia. 2002;7(2):177–89.PubMed
495.
go back to reference Ries, C.H., et al., Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell, 2014 Ries, C.H., et al., Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell, 2014
496.
go back to reference Ries CH et al. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell. 2014;25(6):846–59.PubMed Ries CH et al. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell. 2014;25(6):846–59.PubMed
497.
go back to reference Pyonteck SM et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med. 2013;19(10):1264–72.PubMed Pyonteck SM et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med. 2013;19(10):1264–72.PubMed
498.
go back to reference Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol. 2006;6(3):173–82.PubMed Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol. 2006;6(3):173–82.PubMed
499.
go back to reference Phillipson M, Kubes P. The neutrophil in vascular inflammation. Nat Med. 2011;17(11):1381–90.PubMed Phillipson M, Kubes P. The neutrophil in vascular inflammation. Nat Med. 2011;17(11):1381–90.PubMed
500.
go back to reference Huh SJ et al. Transiently entrapped circulating tumor cells interact with neutrophils to facilitate lung metastasis development. Cancer Res. 2010;70(14):6071–82.PubMedCentralPubMed Huh SJ et al. Transiently entrapped circulating tumor cells interact with neutrophils to facilitate lung metastasis development. Cancer Res. 2010;70(14):6071–82.PubMedCentralPubMed
501.
go back to reference Cools-Lartigue J et al. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Investig. 2013;123(8):3446–58.PubMedCentral Cools-Lartigue J et al. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Investig. 2013;123(8):3446–58.PubMedCentral
502.
go back to reference Teramukai S et al. Pretreatment neutrophil count as an independent prognostic factor in advanced non-small-cell lung cancer: an analysis of Japan Multinational Trial Organisation LC00-03. Eur J Cancer. 2009;45(11):1950–8.PubMed Teramukai S et al. Pretreatment neutrophil count as an independent prognostic factor in advanced non-small-cell lung cancer: an analysis of Japan Multinational Trial Organisation LC00-03. Eur J Cancer. 2009;45(11):1950–8.PubMed
503.
go back to reference McDonald B et al. Systemic inflammation increases cancer cell adhesion to hepatic sinusoids by neutrophil mediated mechanisms. Int J Cancer. 2009;125(6):1298–305.PubMed McDonald B et al. Systemic inflammation increases cancer cell adhesion to hepatic sinusoids by neutrophil mediated mechanisms. Int J Cancer. 2009;125(6):1298–305.PubMed
504.
go back to reference Spicer JD et al. Neutrophils promote liver metastasis via Mac-1-mediated interactions with circulating tumor cells. Cancer Res. 2012;72(16):3919–27.PubMed Spicer JD et al. Neutrophils promote liver metastasis via Mac-1-mediated interactions with circulating tumor cells. Cancer Res. 2012;72(16):3919–27.PubMed
505.
go back to reference Auguste P et al. The host inflammatory response promotes liver metastasis by increasing tumor cell arrest and extravasation. Am J Pathol. 2007;170(5):1781–92.PubMedCentralPubMed Auguste P et al. The host inflammatory response promotes liver metastasis by increasing tumor cell arrest and extravasation. Am J Pathol. 2007;170(5):1781–92.PubMedCentralPubMed
506.
go back to reference Liang S et al. Effects of the tumor-leukocyte microenvironment on melanoma–neutrophil adhesion to the endothelium in a shear flow. Cell Mol Biol. 2008;1(2–3):189–200. Liang S et al. Effects of the tumor-leukocyte microenvironment on melanoma–neutrophil adhesion to the endothelium in a shear flow. Cell Mol Biol. 2008;1(2–3):189–200.
507.
go back to reference Nozawa H, Chiu C, Hanahan D. Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis. Proc Natl Acad Sci U S A. 2006;103(33):12493–8.PubMedCentralPubMed Nozawa H, Chiu C, Hanahan D. Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis. Proc Natl Acad Sci U S A. 2006;103(33):12493–8.PubMedCentralPubMed
508.
go back to reference Lynn KD, Roland CL, Brekken RA. VEGF and pleiotrophin modulate the immune profile of breast cancer. Cancers. 2010;2(2):970–88.PubMedCentralPubMed Lynn KD, Roland CL, Brekken RA. VEGF and pleiotrophin modulate the immune profile of breast cancer. Cancers. 2010;2(2):970–88.PubMedCentralPubMed
509.
go back to reference Lee AM et al. Modeling and simulation of procoagulant circulating tumor cells in flow. Front Oncol. 2012;2:184. Lee AM et al. Modeling and simulation of procoagulant circulating tumor cells in flow. Front Oncol. 2012;2:184.
510.
go back to reference Mantovani A et al. Cancer-related inflammation. Nature. 2008;454(7203):436–44.PubMed Mantovani A et al. Cancer-related inflammation. Nature. 2008;454(7203):436–44.PubMed
511.
go back to reference Sethi G, Sung B, Aggarwal BB. TNF: a master switch for inflammation to cancer. Front Biosci. 2008;13(2):5094–107.PubMed Sethi G, Sung B, Aggarwal BB. TNF: a master switch for inflammation to cancer. Front Biosci. 2008;13(2):5094–107.PubMed
513.
go back to reference Kumar D et al. Synthesis and evaluation of anticancer benzoxazoles and benzimidazoles related to UK-1. Bioorg Med Chem. 2002;10(12):3997–4004.PubMed Kumar D et al. Synthesis and evaluation of anticancer benzoxazoles and benzimidazoles related to UK-1. Bioorg Med Chem. 2002;10(12):3997–4004.PubMed
514.
go back to reference Combe B. Thalidomide: new indications? Joint Bone Spine. 2001;68(6):582–7.PubMed Combe B. Thalidomide: new indications? Joint Bone Spine. 2001;68(6):582–7.PubMed
515.
516.
go back to reference Keifer JA et al. Inhibition of NF-kappa B activity by thalidomide through suppression of IkappaB kinase activity. J Biol Chem. 2001;276(25):22382–7.PubMed Keifer JA et al. Inhibition of NF-kappa B activity by thalidomide through suppression of IkappaB kinase activity. J Biol Chem. 2001;276(25):22382–7.PubMed
517.
go back to reference Laber DA et al. A phase I study of thalidomide, capecitabine and temozolomide in advanced cancer. Cancer Biol Ther. 2007;6(6):840–5.PubMed Laber DA et al. A phase I study of thalidomide, capecitabine and temozolomide in advanced cancer. Cancer Biol Ther. 2007;6(6):840–5.PubMed
518.
go back to reference Zidi I et al. TNF-alpha and its inhibitors in cancer. Med Oncol. 2010;27(2):185–98.PubMed Zidi I et al. TNF-alpha and its inhibitors in cancer. Med Oncol. 2010;27(2):185–98.PubMed
519.
go back to reference Zavadil J, Böttinger EP. TGF-β and epithelial-to-mesenchymal transitions. Oncogene. 2005;24(37):5764–74.PubMed Zavadil J, Böttinger EP. TGF-β and epithelial-to-mesenchymal transitions. Oncogene. 2005;24(37):5764–74.PubMed
520.
go back to reference Giampieri S, Pinner S, Sahai E. Intravital imaging illuminates transforming growth factor β signaling switches during metastasis. Cancer Res. 2010;70(9):3435–9.PubMedCentralPubMed Giampieri S, Pinner S, Sahai E. Intravital imaging illuminates transforming growth factor β signaling switches during metastasis. Cancer Res. 2010;70(9):3435–9.PubMedCentralPubMed
521.
go back to reference Yang L et al. Abrogation of TGFβ signaling in mammary carcinomas recruits Gr-1+ CD11b + myeloid cells that promote metastasis. Cancer Cell. 2008;13(1):23–35.PubMedCentralPubMed Yang L et al. Abrogation of TGFβ signaling in mammary carcinomas recruits Gr-1+ CD11b + myeloid cells that promote metastasis. Cancer Cell. 2008;13(1):23–35.PubMedCentralPubMed
522.
go back to reference Tseng D, Vasquez-Medrano DA, Brown JM. Targeting SDF-1/CXCR4 to inhibit tumour vasculature for treatment of glioblastomas. Br J Cancer. 2011;104(12):1805–9.PubMedCentralPubMed Tseng D, Vasquez-Medrano DA, Brown JM. Targeting SDF-1/CXCR4 to inhibit tumour vasculature for treatment of glioblastomas. Br J Cancer. 2011;104(12):1805–9.PubMedCentralPubMed
523.
go back to reference Redjal N et al. CXCR4 inhibition synergizes with cytotoxic chemotherapy in gliomas. Clin Cancer Res. 2006;12(22):6765–71.PubMed Redjal N et al. CXCR4 inhibition synergizes with cytotoxic chemotherapy in gliomas. Clin Cancer Res. 2006;12(22):6765–71.PubMed
525.
go back to reference Zeng Z et al. SDF-1 inhibition using Spiegelmer® Nox-A12 as a novel strategy for targeting AML cells within their BM microenvironment. Blood. 2013;122:2454–2454. Zeng Z et al. SDF-1 inhibition using Spiegelmer® Nox-A12 as a novel strategy for targeting AML cells within their BM microenvironment. Blood. 2013;122:2454–2454.
526.
527.
528.
go back to reference Frisch SM, Ruoslahti E. Integrins and anoikis. Curr Opin Cell Biol. 1997;9(5):701–6.PubMed Frisch SM, Ruoslahti E. Integrins and anoikis. Curr Opin Cell Biol. 1997;9(5):701–6.PubMed
529.
go back to reference Guo W, Giancotti FG. Integrin signalling during tumour progression. Nat Rev Mol Cell Biol. 2004;5(10):816–26.PubMed Guo W, Giancotti FG. Integrin signalling during tumour progression. Nat Rev Mol Cell Biol. 2004;5(10):816–26.PubMed
530.
go back to reference Guadamillas MC, Cerezo A, del Pozo MA. Overcoming anoikis—pathways to anchorage-independent growth in cancer. J Cell Sci. 2011;124(19):3189–97.PubMed Guadamillas MC, Cerezo A, del Pozo MA. Overcoming anoikis—pathways to anchorage-independent growth in cancer. J Cell Sci. 2011;124(19):3189–97.PubMed
531.
go back to reference Smets FN et al. Loss of cell anchorage triggers apoptosis (anoikis) in primary mouse hepatocytes. Mol Genet Metab. 2002;75(4):344–52.PubMed Smets FN et al. Loss of cell anchorage triggers apoptosis (anoikis) in primary mouse hepatocytes. Mol Genet Metab. 2002;75(4):344–52.PubMed
532.
go back to reference Tanaka K et al. Neurotrophic receptor, tropomyosin‐related kinase B as an independent prognostic marker in gastric cancer patients. J Surg Oncol. 2009;99(5):307–10.PubMed Tanaka K et al. Neurotrophic receptor, tropomyosin‐related kinase B as an independent prognostic marker in gastric cancer patients. J Surg Oncol. 2009;99(5):307–10.PubMed
533.
go back to reference Douma S et al. Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature. 2004;430(7003):1034–9.PubMed Douma S et al. Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature. 2004;430(7003):1034–9.PubMed
534.
go back to reference Kenific CM, Thorburn A, Debnath J. Autophagy and metastasis: another double-edged sword. Curr Opin Cell Biol. 2010;22(2):241–5.PubMedCentralPubMed Kenific CM, Thorburn A, Debnath J. Autophagy and metastasis: another double-edged sword. Curr Opin Cell Biol. 2010;22(2):241–5.PubMedCentralPubMed
535.
go back to reference Meng S et al. Circulating tumor cells in patients with breast cancer dormancy. Clin Cancer Res. 2004;10(24):8152–62.PubMed Meng S et al. Circulating tumor cells in patients with breast cancer dormancy. Clin Cancer Res. 2004;10(24):8152–62.PubMed
536.
go back to reference Kochetkova M, Kumar S, McColl SR. Chemokine receptors CXCR4 and CCR7 promote metastasis by preventing anoikis in cancer cells. Cell Death Differ. 2009;16(5):664–73.PubMed Kochetkova M, Kumar S, McColl SR. Chemokine receptors CXCR4 and CCR7 promote metastasis by preventing anoikis in cancer cells. Cell Death Differ. 2009;16(5):664–73.PubMed
537.
go back to reference Schempp CM et al. V-ATPase inhibition regulates anoikis resistance and metastasis of cancer cells. Mol Cancer Ther. 2014;13(4):926–37.PubMed Schempp CM et al. V-ATPase inhibition regulates anoikis resistance and metastasis of cancer cells. Mol Cancer Ther. 2014;13(4):926–37.PubMed
538.
go back to reference Klubo-Gwiezdzinska J et al. Metformin inhibits growth and decreases resistance to anoikis in medullary thyroid cancer cells. Endocr Relat Cancer. 2012;19(3):447–56.PubMed Klubo-Gwiezdzinska J et al. Metformin inhibits growth and decreases resistance to anoikis in medullary thyroid cancer cells. Endocr Relat Cancer. 2012;19(3):447–56.PubMed
539.
go back to reference Fidler IJ et al. The seed and soil hypothesis: vascularisation and brain metastases. Lancet Oncol. 2002;3(1):53–7.PubMed Fidler IJ et al. The seed and soil hypothesis: vascularisation and brain metastases. Lancet Oncol. 2002;3(1):53–7.PubMed
540.
go back to reference Korb T et al. Integrity of actin fibers and microtubules influences metastatic tumor cell adhesion. Exp Cell Res. 2004;299(1):236–47.PubMed Korb T et al. Integrity of actin fibers and microtubules influences metastatic tumor cell adhesion. Exp Cell Res. 2004;299(1):236–47.PubMed
541.
go back to reference Avvisato CL et al. Mechanical force modulates global gene expression and β-catenin signaling in colon cancer cells. J Cell Sci. 2007;120(15):2672–82.PubMed Avvisato CL et al. Mechanical force modulates global gene expression and β-catenin signaling in colon cancer cells. J Cell Sci. 2007;120(15):2672–82.PubMed
542.
go back to reference Chiu J-J, Chien S. Effects of disturbed flow on vascular endothelium: pathophysiological basis and clinical perspectives. Physiol Rev. 2011;91(1):327–87.PubMed Chiu J-J, Chien S. Effects of disturbed flow on vascular endothelium: pathophysiological basis and clinical perspectives. Physiol Rev. 2011;91(1):327–87.PubMed
543.
go back to reference Barnes JM, Nauseef JT, Henry MD. Resistance to fluid shear stress is a conserved biophysical property of malignant cells. PLoS One. 2012;7(12):e50973.PubMedCentralPubMed Barnes JM, Nauseef JT, Henry MD. Resistance to fluid shear stress is a conserved biophysical property of malignant cells. PLoS One. 2012;7(12):e50973.PubMedCentralPubMed
544.
go back to reference Swartz MA, Lund AW. Lymphatic and interstitial flow in the tumour microenvironment: linking mechanobiology with immunity. Nat Rev Cancer. 2012;12(3):210–9.PubMed Swartz MA, Lund AW. Lymphatic and interstitial flow in the tumour microenvironment: linking mechanobiology with immunity. Nat Rev Cancer. 2012;12(3):210–9.PubMed
545.
go back to reference Pedersen JA, Boschetti F, Swartz MA. Effects of extracellular fiber architecture on cell membrane shear stress in a 3D fibrous matrix. J Biomech. 2007;40(7):1484–92.PubMed Pedersen JA, Boschetti F, Swartz MA. Effects of extracellular fiber architecture on cell membrane shear stress in a 3D fibrous matrix. J Biomech. 2007;40(7):1484–92.PubMed
546.
go back to reference Burdick MM et al. Colon carcinoma cell glycolipids, integrins, and other glycoproteins mediate adhesion to HUVECs under flow. Am J Physiol-Cell Physiol. 2003;284(4):C977–87.PubMed Burdick MM et al. Colon carcinoma cell glycolipids, integrins, and other glycoproteins mediate adhesion to HUVECs under flow. Am J Physiol-Cell Physiol. 2003;284(4):C977–87.PubMed
547.
go back to reference Mitchell MJ, King MR. Computational and experimental models of cancer cell response to fluid shear stress. Front Oncol. 2013;3:44.PubMedCentralPubMed Mitchell MJ, King MR. Computational and experimental models of cancer cell response to fluid shear stress. Front Oncol. 2013;3:44.PubMedCentralPubMed
548.
549.
go back to reference Ruffell B et al. Lymphocytes in cancer development: polarization towards pro-tumor immunity. Cytokine Growth Factor Rev. 2010;21(1):3–10.PubMedCentralPubMed Ruffell B et al. Lymphocytes in cancer development: polarization towards pro-tumor immunity. Cytokine Growth Factor Rev. 2010;21(1):3–10.PubMedCentralPubMed
550.
go back to reference Garcia‐Lora A, Algarra I, Garrido F. MHC class I antigens, immune surveillance, and tumor immune escape. J Cell Physiol. 2003;195(3):346–55.PubMed Garcia‐Lora A, Algarra I, Garrido F. MHC class I antigens, immune surveillance, and tumor immune escape. J Cell Physiol. 2003;195(3):346–55.PubMed
551.
go back to reference Luo J-L, Kamata H, Karin M. IKK/NF-κB signaling: balancing life and death—a new approach to cancer therapy. J Clin Invest. 2005;115(10):2625–32.PubMedCentralPubMed Luo J-L, Kamata H, Karin M. IKK/NF-κB signaling: balancing life and death—a new approach to cancer therapy. J Clin Invest. 2005;115(10):2625–32.PubMedCentralPubMed
552.
go back to reference Kim S et al. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature. 2009;457(7225):102–6.PubMedCentralPubMed Kim S et al. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature. 2009;457(7225):102–6.PubMedCentralPubMed
553.
go back to reference Bonecchi R et al. Chemokines and chemokine receptors: an overview. Front Biosci (Landmark edition). 2008;14:540–51. Bonecchi R et al. Chemokines and chemokine receptors: an overview. Front Biosci (Landmark edition). 2008;14:540–51.
554.
go back to reference Lee BJ et al. Tissue factor is involved in retinoblastoma cell proliferation via both the Akt and extracellular signal-regulated kinase pathways. Oncol Rep. 2011;26(3):665.PubMed Lee BJ et al. Tissue factor is involved in retinoblastoma cell proliferation via both the Akt and extracellular signal-regulated kinase pathways. Oncol Rep. 2011;26(3):665.PubMed
555.
go back to reference Liu Y et al. Tissue factor-activated coagulation cascade in the tumor microenvironment is critical for tumor progression and an effective target for therapy. Cancer Res. 2011;71(20):6492–502.PubMed Liu Y et al. Tissue factor-activated coagulation cascade in the tumor microenvironment is critical for tumor progression and an effective target for therapy. Cancer Res. 2011;71(20):6492–502.PubMed
556.
go back to reference Versteeg HH et al. Tissue factor and cancer metastasis: the role of intracellular and extracellular signaling pathways. Mol Med. 2004;10(1–6):6.PubMedCentralPubMed Versteeg HH et al. Tissue factor and cancer metastasis: the role of intracellular and extracellular signaling pathways. Mol Med. 2004;10(1–6):6.PubMedCentralPubMed
557.
558.
go back to reference Welsh J et al. Tissue factor expression determines tumour cell coagulation kinetics. Int J Lab Hematol. 2012;34(4):396–402.PubMed Welsh J et al. Tissue factor expression determines tumour cell coagulation kinetics. Int J Lab Hematol. 2012;34(4):396–402.PubMed
559.
go back to reference Breij EC et al. An antibody-drug conjugate that targets tissue factor exhibits potent therapeutic activity against a broad range of solid tumors. Cancer Res. 2014;74(4):1214–26.PubMed Breij EC et al. An antibody-drug conjugate that targets tissue factor exhibits potent therapeutic activity against a broad range of solid tumors. Cancer Res. 2014;74(4):1214–26.PubMed
560.
go back to reference Belloc C et al. The effect of platelets on invasiveness and protease production of human mammary tumor cells. Int J Cancer. 1995;60(3):413–7.PubMed Belloc C et al. The effect of platelets on invasiveness and protease production of human mammary tumor cells. Int J Cancer. 1995;60(3):413–7.PubMed
561.
go back to reference Erpenbeck L et al. Inhibition of platelet GPIbα and promotion of melanoma metastasis. J Invest Dermatol. 2009;130(2):576–86.PubMed Erpenbeck L et al. Inhibition of platelet GPIbα and promotion of melanoma metastasis. J Invest Dermatol. 2009;130(2):576–86.PubMed
562.
go back to reference Shau H, Roth M, Golub S. Regulation of natural killer function by nonlymphoid cells. Nat Immun. 1992;12(4–5):235–49. Shau H, Roth M, Golub S. Regulation of natural killer function by nonlymphoid cells. Nat Immun. 1992;12(4–5):235–49.
563.
go back to reference Kopp H-G, Placke T, Salih HR. Platelet-derived transforming growth factor-β down-regulates NKG2D thereby inhibiting natural killer cell antitumor reactivity. Cancer Res. 2009;69(19):7775–83.PubMed Kopp H-G, Placke T, Salih HR. Platelet-derived transforming growth factor-β down-regulates NKG2D thereby inhibiting natural killer cell antitumor reactivity. Cancer Res. 2009;69(19):7775–83.PubMed
564.
go back to reference Bambace N, Holmes C. The platelet contribution to cancer progression. J Thromb Haemost. 2011;9(2):237–49.PubMed Bambace N, Holmes C. The platelet contribution to cancer progression. J Thromb Haemost. 2011;9(2):237–49.PubMed
565.
go back to reference Placke T et al. Platelet-derived MHC class I confers a pseudonormal phenotype to cancer cells that subverts the antitumor reactivity of natural killer immune cells. Cancer Res. 2012;72(2):440–8.PubMed Placke T et al. Platelet-derived MHC class I confers a pseudonormal phenotype to cancer cells that subverts the antitumor reactivity of natural killer immune cells. Cancer Res. 2012;72(2):440–8.PubMed
566.
go back to reference Palumbo J.S. Mechanisms linking tumor cell-associated procoagulant function to tumor dissemination. in Seminars in thrombosis and hemostasis. 2008. © Thieme Medical Publishers. Palumbo J.S. Mechanisms linking tumor cell-associated procoagulant function to tumor dissemination. in Seminars in thrombosis and hemostasis. 2008. © Thieme Medical Publishers.
567.
go back to reference Coupland LA, Chong BH, Parish CR. Platelets and p-selectin control tumor cell metastasis in an organ-specific manner and independently of NK cells. Cancer Res. 2012;72(18):4662–71.PubMed Coupland LA, Chong BH, Parish CR. Platelets and p-selectin control tumor cell metastasis in an organ-specific manner and independently of NK cells. Cancer Res. 2012;72(18):4662–71.PubMed
568.
go back to reference Jurasz P. D. Alonso‐Escolano, and M.W. Radomski, Platelet–cancer interactions: mechanisms and pharmacology of tumour cell‐induced platelet aggregation. Br J Pharmacol. 2004;143(7):819–26.PubMedCentralPubMed Jurasz P. D. Alonso‐Escolano, and M.W. Radomski, Platelet–cancer interactions: mechanisms and pharmacology of tumour cell‐induced platelet aggregation. Br J Pharmacol. 2004;143(7):819–26.PubMedCentralPubMed
569.
go back to reference Toliopoulos IK et al. Resveratrol diminishes platelet aggregation and increases susceptibility of K562 tumor cells to natural killer cells. Indian J Biochem Biophys. 2013;50(1):14–8.PubMed Toliopoulos IK et al. Resveratrol diminishes platelet aggregation and increases susceptibility of K562 tumor cells to natural killer cells. Indian J Biochem Biophys. 2013;50(1):14–8.PubMed
570.
go back to reference Amirkhosravi A et al. Inhibition of tumor cell-induced platelet aggregation and lung metastasis by the oral GpIIb/IIIa antagonist XV454. Thromb Haemost. 2003;90(3):549–54.PubMed Amirkhosravi A et al. Inhibition of tumor cell-induced platelet aggregation and lung metastasis by the oral GpIIb/IIIa antagonist XV454. Thromb Haemost. 2003;90(3):549–54.PubMed
571.
572.
go back to reference Strell C, Entschladen F. Extravasation of leukocytes in comparison to tumor cells. Cell Commun Signal. 2008;6(10):5. Strell C, Entschladen F. Extravasation of leukocytes in comparison to tumor cells. Cell Commun Signal. 2008;6(10):5.
573.
go back to reference Schlüter K et al. Organ-specific metastatic tumor cell adhesion and extravasation of colon carcinoma cells with different metastatic potential. Am J Pathol. 2006;169(3):1064–73.PubMedCentralPubMed Schlüter K et al. Organ-specific metastatic tumor cell adhesion and extravasation of colon carcinoma cells with different metastatic potential. Am J Pathol. 2006;169(3):1064–73.PubMedCentralPubMed
575.
go back to reference Weis SM, Cheresh DA. Pathophysiological consequences of VEGF-induced vascular permeability. Nature. 2005;437(7058):497–504.PubMed Weis SM, Cheresh DA. Pathophysiological consequences of VEGF-induced vascular permeability. Nature. 2005;437(7058):497–504.PubMed
576.
go back to reference Colmone A et al. Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells. Science. 2008;322(5909):1861–5.PubMed Colmone A et al. Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells. Science. 2008;322(5909):1861–5.PubMed
577.
go back to reference Kienast Y et al. Real-time imaging reveals the single steps of brain metastasis formation. Nat Med. 2010;16(1):116–22.PubMed Kienast Y et al. Real-time imaging reveals the single steps of brain metastasis formation. Nat Med. 2010;16(1):116–22.PubMed
578.
go back to reference Heyder C et al. Visualization of tumor cell extravasation. Contrib Microbiol. 2006;13:200–8.PubMed Heyder C et al. Visualization of tumor cell extravasation. Contrib Microbiol. 2006;13:200–8.PubMed
579.
go back to reference Kim J-E et al. RGD peptides released from βig-h3, a TGF-β-induced cell-adhesive molecule, mediate apoptosis. Oncogene. 2003;22(13):2045–53.PubMed Kim J-E et al. RGD peptides released from βig-h3, a TGF-β-induced cell-adhesive molecule, mediate apoptosis. Oncogene. 2003;22(13):2045–53.PubMed
580.
go back to reference Padua D et al. TGFβ primes breast tumors for lung metastasis seeding through angiopoietin-like 4. Cell. 2008;133(1):66–77.PubMedCentralPubMed Padua D et al. TGFβ primes breast tumors for lung metastasis seeding through angiopoietin-like 4. Cell. 2008;133(1):66–77.PubMedCentralPubMed
581.
go back to reference Lu X, Kang Y. Hypoxia and hypoxia-inducible factors: master regulators of metastasis. Clin Cancer Res. 2010;16(24):5928–35.PubMedCentralPubMed Lu X, Kang Y. Hypoxia and hypoxia-inducible factors: master regulators of metastasis. Clin Cancer Res. 2010;16(24):5928–35.PubMedCentralPubMed
582.
go back to reference Fidler IJ, Poste G. The “seed and soil” hypothesis revisited. Lancet Oncol. 2008;9(8):808.PubMed Fidler IJ, Poste G. The “seed and soil” hypothesis revisited. Lancet Oncol. 2008;9(8):808.PubMed
583.
go back to reference Fidler IJ et al. The brain microenvironment and cancer metastasis. Mol Cells. 2010;30(2):93–8.PubMed Fidler IJ et al. The brain microenvironment and cancer metastasis. Mol Cells. 2010;30(2):93–8.PubMed
584.
go back to reference Trinh VA, Hwu W-J. Chemoprevention for brain metastases. Curr Oncol Rep. 2012;14(1):63–9.PubMed Trinh VA, Hwu W-J. Chemoprevention for brain metastases. Curr Oncol Rep. 2012;14(1):63–9.PubMed
585.
go back to reference Woodhouse EC, Chuaqui RF, Liotta LA. General mechanisms of metastasis. Cancer. 1997;80(S8):1529–37.PubMed Woodhouse EC, Chuaqui RF, Liotta LA. General mechanisms of metastasis. Cancer. 1997;80(S8):1529–37.PubMed
586.
go back to reference Talmadge JE, Fidler IJ. AACR centennial series: the biology of cancer metastasis: historical perspective. Cancer Res. 2010;70(14):5649–69.PubMedCentralPubMed Talmadge JE, Fidler IJ. AACR centennial series: the biology of cancer metastasis: historical perspective. Cancer Res. 2010;70(14):5649–69.PubMedCentralPubMed
587.
go back to reference Mathot L, Stenninger J. Behavior of seeds and soil in the mechanism of metastasis: a deeper understanding. Cancer Sci. 2012;103(4):626–31.PubMed Mathot L, Stenninger J. Behavior of seeds and soil in the mechanism of metastasis: a deeper understanding. Cancer Sci. 2012;103(4):626–31.PubMed
588.
go back to reference Langley RR, Fidler IJ. Tumor cell-organ microenvironment interactions in the pathogenesis of cancer metastasis. Endocr Rev. 2007;28(3):297–321.PubMed Langley RR, Fidler IJ. Tumor cell-organ microenvironment interactions in the pathogenesis of cancer metastasis. Endocr Rev. 2007;28(3):297–321.PubMed
589.
go back to reference Fidler IJ, Kripke ML. Genomic analysis of primary tumors does not address the prevalence of metastatic cells in the population. Nat Genet. 2003;34(1):23–23.PubMed Fidler IJ, Kripke ML. Genomic analysis of primary tumors does not address the prevalence of metastatic cells in the population. Nat Genet. 2003;34(1):23–23.PubMed
590.
go back to reference Ruoslahti E. Vascular zip codes in angiogenesis and metastasis. Biochem Soc Trans. 2004;32:397–402.PubMed Ruoslahti E. Vascular zip codes in angiogenesis and metastasis. Biochem Soc Trans. 2004;32:397–402.PubMed
591.
go back to reference Müller A et al. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001;410(6824):50–6.PubMed Müller A et al. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001;410(6824):50–6.PubMed
592.
593.
go back to reference Naumov GN et al. Persistence of solitary mammary carcinoma cells in a secondary site: a possible contributor to dormancy. Cancer Res. 2002;62(7):2162–8.PubMed Naumov GN et al. Persistence of solitary mammary carcinoma cells in a secondary site: a possible contributor to dormancy. Cancer Res. 2002;62(7):2162–8.PubMed
594.
595.
go back to reference Udagawa T. Tumor dormancy of primary and secondary cancers. Apmis. 2008;116(7‐8):615–28.PubMed Udagawa T. Tumor dormancy of primary and secondary cancers. Apmis. 2008;116(7‐8):615–28.PubMed
596.
go back to reference Almog N. Molecular mechanisms underlying tumor dormancy. Cancer Lett. 2010;294(2):139–46.PubMed Almog N. Molecular mechanisms underlying tumor dormancy. Cancer Lett. 2010;294(2):139–46.PubMed
597.
go back to reference Baeriswyl V. and Christofori G. The angiogenic switch in carcinogenesis. in Seminars in cancer biology. 2009. Elsevier. Baeriswyl V. and Christofori G. The angiogenic switch in carcinogenesis. in Seminars in cancer biology. 2009. Elsevier.
598.
go back to reference Watnick RS et al. RETRACTED: Ras modulates Myc activity to repress thrombospondin-1 expression and increase tumor angiogenesis. Cancer Cell. 2003;3(3):219–31.PubMed Watnick RS et al. RETRACTED: Ras modulates Myc activity to repress thrombospondin-1 expression and increase tumor angiogenesis. Cancer Cell. 2003;3(3):219–31.PubMed
599.
go back to reference Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 1996;86(3):p. 353–364.PubMed Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 1996;86(3):p. 353–364.PubMed
600.
go back to reference Hensel JA, Flaig TW, Theodorescu D. Clinical opportunities and challenges in targeting tumour dormancy. Nat Rev Clin Oncol. 2012;10(1):41–51.PubMed Hensel JA, Flaig TW, Theodorescu D. Clinical opportunities and challenges in targeting tumour dormancy. Nat Rev Clin Oncol. 2012;10(1):41–51.PubMed
601.
go back to reference Barkan D et al. Inhibition of metastatic outgrowth from single dormant tumor cells by targeting the cytoskeleton. Cancer Res. 2008;68(15):6241–50.PubMedCentralPubMed Barkan D et al. Inhibition of metastatic outgrowth from single dormant tumor cells by targeting the cytoskeleton. Cancer Res. 2008;68(15):6241–50.PubMedCentralPubMed
602.
go back to reference Bragado P. et al., Microenvironments dictating tumor cell dormancy, in Minimal residual disease and circulating tumor cells in breast cancer. 2012, Springer. p. 25-39. Bragado P. et al., Microenvironments dictating tumor cell dormancy, in Minimal residual disease and circulating tumor cells in breast cancer. 2012, Springer. p. 25-39.
603.
go back to reference Aguirre-Ghiso JA et al. Urokinase receptor and fibronectin regulate the ERKMAPK to p38MAPK activity ratios that determine carcinoma cell proliferation or dormancy in vivo. Mol Biol Cell. 2001;12(4):863–79.PubMedCentralPubMed Aguirre-Ghiso JA et al. Urokinase receptor and fibronectin regulate the ERKMAPK to p38MAPK activity ratios that determine carcinoma cell proliferation or dormancy in vivo. Mol Biol Cell. 2001;12(4):863–79.PubMedCentralPubMed
604.
go back to reference Allgayer H, Aguirre‐ghiso JA. The urokinase receptor (u‐PAR)—a link between tumor cell dormancy and minimal residual disease in bone marrow? Apmis. 2008;116(7‐8):602–14.PubMedCentralPubMed Allgayer H, Aguirre‐ghiso JA. The urokinase receptor (u‐PAR)—a link between tumor cell dormancy and minimal residual disease in bone marrow? Apmis. 2008;116(7‐8):602–14.PubMedCentralPubMed
605.
go back to reference Kren A et al. Increased tumor cell dissemination and cellular senescence in the absence of β1‐integrin function. EMBO J. 2007;26(12):2832–42.PubMedCentralPubMed Kren A et al. Increased tumor cell dissemination and cellular senescence in the absence of β1‐integrin function. EMBO J. 2007;26(12):2832–42.PubMedCentralPubMed
606.
go back to reference Teng MW et al. Immune-mediated dormancy: an equilibrium with cancer. J Leukocyte Biol. 2008;84(4):988–93.PubMed Teng MW et al. Immune-mediated dormancy: an equilibrium with cancer. J Leukocyte Biol. 2008;84(4):988–93.PubMed
607.
go back to reference Moserle L, Amadori A, Indraccolo S. The angiogenic switch: implications in the regulation of tumor dormancy. Curr Mol Med. 2009;9(8):935–41.PubMed Moserle L, Amadori A, Indraccolo S. The angiogenic switch: implications in the regulation of tumor dormancy. Curr Mol Med. 2009;9(8):935–41.PubMed
608.
go back to reference Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22:329–60.PubMed Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22:329–60.PubMed
609.
go back to reference Chambers AF. Influence of diet on metastasis and tumor dormancy. Clin Exp Metastasis. 2009;26(1):61–6.PubMed Chambers AF. Influence of diet on metastasis and tumor dormancy. Clin Exp Metastasis. 2009;26(1):61–6.PubMed
610.
go back to reference Gewirtz DA. Autophagy, senescence and tumor dormancy in cancer therapy. Autophagy. 2009;5(8):1232–4.PubMed Gewirtz DA. Autophagy, senescence and tumor dormancy in cancer therapy. Autophagy. 2009;5(8):1232–4.PubMed
611.
go back to reference Troyanovsky B et al. Angiomotin: an angiostatin binding protein that regulates endothelial cell migration and tube formation. J Cell Biol. 2001;152(6):1247–54.PubMedCentralPubMed Troyanovsky B et al. Angiomotin: an angiostatin binding protein that regulates endothelial cell migration and tube formation. J Cell Biol. 2001;152(6):1247–54.PubMedCentralPubMed
612.
go back to reference Zhang X et al. Induction of mitochondrial dysfunction as a strategy for targeting tumour cells in metabolically compromised microenvironments. Nat Commun. 2014;5:3295.PubMedCentralPubMed Zhang X et al. Induction of mitochondrial dysfunction as a strategy for targeting tumour cells in metabolically compromised microenvironments. Nat Commun. 2014;5:3295.PubMedCentralPubMed
613.
go back to reference El Touny LH et al. Combined SFK/MEK inhibition prevents metastatic outgrowth of dormant tumor cells. J Clin Invest. 2014;124(1):156–68.PubMedCentralPubMed El Touny LH et al. Combined SFK/MEK inhibition prevents metastatic outgrowth of dormant tumor cells. J Clin Invest. 2014;124(1):156–68.PubMedCentralPubMed
614.
go back to reference Perren TJ et al. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med. 2011;365(26):2484–96.PubMed Perren TJ et al. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med. 2011;365(26):2484–96.PubMed
615.
go back to reference O’Reilly MS et al. Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell. 1994;79(2):315–28.PubMed O’Reilly MS et al. Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell. 1994;79(2):315–28.PubMed
616.
go back to reference O’Reilly MS et al. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell. 1997;88(2):277–85.PubMed O’Reilly MS et al. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell. 1997;88(2):277–85.PubMed
617.
go back to reference Folkman J. Antiangiogenesis in cancer therapy—endostatin and its mechanisms of action. Exp Cell Res. 2006;312(5):594–607.PubMed Folkman J. Antiangiogenesis in cancer therapy—endostatin and its mechanisms of action. Exp Cell Res. 2006;312(5):594–607.PubMed
618.
go back to reference Volpert OV, Lawler J, Bouck NP. A human fibrosarcoma inhibits systemic angiogenesis and the growth of experimental metastases via thrombospondin-1. Proc Natl Acad Sci U S A. 1998;95(11):6343–8.PubMedCentralPubMed Volpert OV, Lawler J, Bouck NP. A human fibrosarcoma inhibits systemic angiogenesis and the growth of experimental metastases via thrombospondin-1. Proc Natl Acad Sci U S A. 1998;95(11):6343–8.PubMedCentralPubMed
619.
go back to reference Hahnfeldt P et al. Tumor development under angiogenic signaling: a dynamical theory of tumor growth, treatment response, and postvascular dormancy. Cancer Res. 1999;59(19):4770–5.PubMed Hahnfeldt P et al. Tumor development under angiogenic signaling: a dynamical theory of tumor growth, treatment response, and postvascular dormancy. Cancer Res. 1999;59(19):4770–5.PubMed
620.
go back to reference Folkman J. Angiogenesis: an organizing principle for drug discovery? Nat Rev Drug Discov. 2007;6(4):273–86.PubMed Folkman J. Angiogenesis: an organizing principle for drug discovery? Nat Rev Drug Discov. 2007;6(4):273–86.PubMed
622.
go back to reference Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1995;1(1):27–31.PubMed Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1995;1(1):27–31.PubMed
623.
go back to reference Benzekry S, Gandolfi A, Hahnfeldt P. Global dormancy of metastases due to systemic inhibition of angiogenesis. PLoS One. 2014;9(1):e84249.PubMedCentralPubMed Benzekry S, Gandolfi A, Hahnfeldt P. Global dormancy of metastases due to systemic inhibition of angiogenesis. PLoS One. 2014;9(1):e84249.PubMedCentralPubMed
624.
go back to reference Livant DL et al. Anti-invasive, antitumorigenic, and antimetastatic activities of the PHSCN sequence in prostate carcinoma. Cancer Res. 2000;60(2):309–20.PubMed Livant DL et al. Anti-invasive, antitumorigenic, and antimetastatic activities of the PHSCN sequence in prostate carcinoma. Cancer Res. 2000;60(2):309–20.PubMed
Metadata
Title
Metastasis review: from bench to bedside
Authors
Ali Mohammad Alizadeh
Sadaf Shiri
Sadaf Farsinejad
Publication date
01-09-2014
Publisher
Springer Netherlands
Published in
Tumor Biology / Issue 9/2014
Print ISSN: 1010-4283
Electronic ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-014-2421-z

Other articles of this Issue 9/2014

Tumor Biology 9/2014 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine