Skip to main content
Top
Published in: Clinical & Experimental Metastasis 6/2015

01-08-2015 | Review

Molecular targets and pathways involved in liver metastasis of colorectal cancer

Authors: Ulrich H. Weidle, Fabian Birzele, Achim Krüger

Published in: Clinical & Experimental Metastasis | Issue 6/2015

Login to get access

Abstract

We here summarize the current view of molecular mechanisms involved in the dissemination process of colorectal cancer cells to the liver as deduced from preclinical models. We focus on molecular aspects of the current understanding of the biology of liver metastases formation and survival, both being crucial for identification and validation of possible therapeutic targets and review the latest findings elucidating some features of the liver as a metastatic niche. In more detail, we outline the role of proteases and of major pathways such asc-MET signaling and its modulation by factors such as MACC1 and TIMP1, as well as Notch and TGFβ signaling. The relevance of these signalling pathways during tumor-stroma interactions in this context will be addressed. In addition, the functional role and validation of targets such as PRL3, Trop-2, L1CAM, S100A4, S100P, CD133, LIPC, and APOBEC3G are summarized.
Literature
1.
go back to reference Kinzler KW, Vogelstein B (1996) Lessons from hereditary colorectal cancer. Cell 87:159–170PubMed Kinzler KW, Vogelstein B (1996) Lessons from hereditary colorectal cancer. Cell 87:159–170PubMed
2.
go back to reference Velculescu VE, Vogelstein B, Kinzler KW (2000) Analysing uncharted transcriptomes with SAGE. Trends Genet 16:423–425PubMed Velculescu VE, Vogelstein B, Kinzler KW (2000) Analysing uncharted transcriptomes with SAGE. Trends Genet 16:423–425PubMed
3.
go back to reference Markowitz SD, Bertagnolli MM (2009) Molecular origins of cancer: molecular basis of colorectal cancer. N Engl J Med 361:2449–2460PubMedCentralPubMed Markowitz SD, Bertagnolli MM (2009) Molecular origins of cancer: molecular basis of colorectal cancer. N Engl J Med 361:2449–2460PubMedCentralPubMed
4.
go back to reference Hölzl D, Eckel R, Engel J (2009) Colorectal cancer metastasis. Frequency, prognosis and consequences. Chirurg 80:331–340 Hölzl D, Eckel R, Engel J (2009) Colorectal cancer metastasis. Frequency, prognosis and consequences. Chirurg 80:331–340
5.
go back to reference Hess KR, Varadhachary GR, Taylor SH, Wei W, Raber MN, Lenz R, Abbruzzese JL (2006) Metastatic patterns in adenocarcinoma. Cancer 106:1624–1633PubMed Hess KR, Varadhachary GR, Taylor SH, Wei W, Raber MN, Lenz R, Abbruzzese JL (2006) Metastatic patterns in adenocarcinoma. Cancer 106:1624–1633PubMed
6.
go back to reference Wan L, Pantel K, Kang Y (2013) Tumor metastasis: moving new biological insights into the clinic. Nat Med 19:1450–1564PubMed Wan L, Pantel K, Kang Y (2013) Tumor metastasis: moving new biological insights into the clinic. Nat Med 19:1450–1564PubMed
7.
go back to reference Deneve E, Rietdorf S, Ramos J et al (2013) Capture of viable circulating tumor cells in the liver of colorectal cancer patients. Clin Chem 59:1384–1392PubMed Deneve E, Rietdorf S, Ramos J et al (2013) Capture of viable circulating tumor cells in the liver of colorectal cancer patients. Clin Chem 59:1384–1392PubMed
8.
go back to reference MacDonald IC, Groom AC, Chambers AF (2002) Cancer spread and micrometastasis development: quantitative approaches for in vivo models. BioEssays 24:885–893PubMed MacDonald IC, Groom AC, Chambers AF (2002) Cancer spread and micrometastasis development: quantitative approaches for in vivo models. BioEssays 24:885–893PubMed
9.
go back to reference Padget S (1889) The distribution of secondary growths in cancer of the breast. Lancet 1:571–573 Padget S (1889) The distribution of secondary growths in cancer of the breast. Lancet 1:571–573
10.
go back to reference Sceneay J, Smith MJ, Möller H (2013) The pre-metastatic niche: finding common ground. Cancer Metastasis Rev 32:449–464PubMed Sceneay J, Smith MJ, Möller H (2013) The pre-metastatic niche: finding common ground. Cancer Metastasis Rev 32:449–464PubMed
11.
go back to reference Nguyen DX, Bos PD, Massague J (2009) Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer 9:274–284PubMed Nguyen DX, Bos PD, Massague J (2009) Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer 9:274–284PubMed
12.
13.
go back to reference Brabletz T, Jung A, Hermann K, Günther K, Hohenberger W, Kirchner T (1998) Nuclear overexpression of the oncoprotein beta-catenin in colorectal cancer is localized predominantly at the invasion front. Pathol Res Pract 194:701–704PubMed Brabletz T, Jung A, Hermann K, Günther K, Hohenberger W, Kirchner T (1998) Nuclear overexpression of the oncoprotein beta-catenin in colorectal cancer is localized predominantly at the invasion front. Pathol Res Pract 194:701–704PubMed
14.
go back to reference Brabletz T, Jung A, Reu S, Porzner M, Hlubek F, Kunz-Schughart LA, Knuechel R, Kirchner T (2001) Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci USA 98:10356–10361PubMedCentralPubMed Brabletz T, Jung A, Reu S, Porzner M, Hlubek F, Kunz-Schughart LA, Knuechel R, Kirchner T (2001) Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci USA 98:10356–10361PubMedCentralPubMed
15.
go back to reference Kirchner T, Brabletz T (2000) Patterning and nuclear beta-catenin expression in the colonic adenoma-carcinoma sequence. Analogies with human gastrulation. Am J Pathol 157:1113–1121PubMedCentralPubMed Kirchner T, Brabletz T (2000) Patterning and nuclear beta-catenin expression in the colonic adenoma-carcinoma sequence. Analogies with human gastrulation. Am J Pathol 157:1113–1121PubMedCentralPubMed
16.
go back to reference Placke T, Kopp HG, Salih HR (1996) The wolf in sheeps clothing: platelet derived “pseudo-self” impairs cancer cell “missing self” recognition by NK cells. Oncoimmunology 1:557–559 Placke T, Kopp HG, Salih HR (1996) The wolf in sheeps clothing: platelet derived “pseudo-self” impairs cancer cell “missing self” recognition by NK cells. Oncoimmunology 1:557–559
17.
go back to reference Bayon LG, Izquierdo MA, Sirovich I, van Rooijen N, Beelen RH, Meijer S (1996) Role of Kupffer cells in arresting circulating tumor cells and controlling metastatic growth in the liver. Hepatology 23:1224–1231PubMed Bayon LG, Izquierdo MA, Sirovich I, van Rooijen N, Beelen RH, Meijer S (1996) Role of Kupffer cells in arresting circulating tumor cells and controlling metastatic growth in the liver. Hepatology 23:1224–1231PubMed
18.
go back to reference Luo DZ, Vermijlen D, Ahisali B, Triantis V, Plakoutsi G, Braet F, Vanderkerken K, Wisse E (2000) On the biology of pit cells, the liver-specific NK cells. World J Gastroenterol 6:1–11PubMed Luo DZ, Vermijlen D, Ahisali B, Triantis V, Plakoutsi G, Braet F, Vanderkerken K, Wisse E (2000) On the biology of pit cells, the liver-specific NK cells. World J Gastroenterol 6:1–11PubMed
19.
go back to reference Wellner UF, Keck T, Brabletz T (2010) Liver metastases: pathogenesis and oncogenesis. Chirurg 81:551–556PubMed Wellner UF, Keck T, Brabletz T (2010) Liver metastases: pathogenesis and oncogenesis. Chirurg 81:551–556PubMed
21.
go back to reference Paschos KA, Majeed AW, Bird NC (2014) Natural history of hepatic metastases from colorectal-pathological pathways with clinical significance. World J Gastroenterol 20:3719–3737PubMedCentralPubMed Paschos KA, Majeed AW, Bird NC (2014) Natural history of hepatic metastases from colorectal-pathological pathways with clinical significance. World J Gastroenterol 20:3719–3737PubMedCentralPubMed
22.
go back to reference Giavazzi R, Jessup JM, Campbell DE, Walker SM, Fidler IJ (1986) Experimental nude mouse model of human colorectal cancer liver metastases. J Natl Cancer Inst 77:1303–1308PubMed Giavazzi R, Jessup JM, Campbell DE, Walker SM, Fidler IJ (1986) Experimental nude mouse model of human colorectal cancer liver metastases. J Natl Cancer Inst 77:1303–1308PubMed
23.
go back to reference Morikawa K, Walker SM, Jessup JM, Fidler IJ (1988) In vivo selection of highly metastatic cells from surgical specimens of different human primary colon carcinomas in nude mice. Cancer Res 48:1943–1948PubMed Morikawa K, Walker SM, Jessup JM, Fidler IJ (1988) In vivo selection of highly metastatic cells from surgical specimens of different human primary colon carcinomas in nude mice. Cancer Res 48:1943–1948PubMed
24.
go back to reference Fidler IJ (1991) Orthotopic implantation of human colon carcinomas into nude mice provides a valuable model for the biology and therapy of metastasis. Cancer Metastasis Rev 10:229–243PubMed Fidler IJ (1991) Orthotopic implantation of human colon carcinomas into nude mice provides a valuable model for the biology and therapy of metastasis. Cancer Metastasis Rev 10:229–243PubMed
25.
go back to reference Morikawa K, Walker SM, Nakajima M, Pathak S, Jessup JM, Fidler IJ (1988) Influence of organ environment on the growth, selection and metastasis of human colon carcinoma cells in nude mice. Cancer Res 48:6863–6871PubMed Morikawa K, Walker SM, Nakajima M, Pathak S, Jessup JM, Fidler IJ (1988) Influence of organ environment on the growth, selection and metastasis of human colon carcinoma cells in nude mice. Cancer Res 48:6863–6871PubMed
26.
go back to reference Fidler IJ, Wilmanns C, Staroselsky A, Radinsky R, Dong Z, Fan D (1994) Modulation of tumor cell response to chemotherapy by the organ environment. Cancer Metastasis Rev 13:209–243PubMed Fidler IJ, Wilmanns C, Staroselsky A, Radinsky R, Dong Z, Fan D (1994) Modulation of tumor cell response to chemotherapy by the organ environment. Cancer Metastasis Rev 13:209–243PubMed
27.
go back to reference Radinski R, Risin S, Fan D, Dong Z, Bielenberg D, Bucana CD, Fidler IJ (1995) Level and function of epidermal growth factor receptor predict the metastatic potential of human colon carcinoma cells. Clin Cancer Res 1:19–31 Radinski R, Risin S, Fan D, Dong Z, Bielenberg D, Bucana CD, Fidler IJ (1995) Level and function of epidermal growth factor receptor predict the metastatic potential of human colon carcinoma cells. Clin Cancer Res 1:19–31
28.
go back to reference Kitadai Y, Bucana CD, Ellis LM, Anzai H, Tahara E, Fidler IJ (1995) In situ mRNA hybridization technique for analysis of metastasis-related genes in human colon carcinoma cells. Am J Pathol 147:1238–1247PubMedCentralPubMed Kitadai Y, Bucana CD, Ellis LM, Anzai H, Tahara E, Fidler IJ (1995) In situ mRNA hybridization technique for analysis of metastasis-related genes in human colon carcinoma cells. Am J Pathol 147:1238–1247PubMedCentralPubMed
29.
go back to reference Takahashi Y, Llis LM, Wilson MR, Bucana CD, Kitadai Y (1996) Progressive upregulation of metastasis-related genes in human colon cancer cells implanted into the cecum of nude mice. Oncol Res 8:163–169PubMed Takahashi Y, Llis LM, Wilson MR, Bucana CD, Kitadai Y (1996) Progressive upregulation of metastasis-related genes in human colon cancer cells implanted into the cecum of nude mice. Oncol Res 8:163–169PubMed
30.
go back to reference Kitadai Y, Sasaki T, Kuwai T, Nakamura T, Bucana CD, Fidler IJ (2006) Targeting the expression of platelet-derived growth factor receptor by reactive stroma inhibits growth and metastasis of human colon carcinoma. Am J Pathol 169:2054–2065PubMedCentralPubMed Kitadai Y, Sasaki T, Kuwai T, Nakamura T, Bucana CD, Fidler IJ (2006) Targeting the expression of platelet-derived growth factor receptor by reactive stroma inhibits growth and metastasis of human colon carcinoma. Am J Pathol 169:2054–2065PubMedCentralPubMed
31.
go back to reference Kuwai T, Nakamura T, Sasaki T, Kitadai Y, Kim JS, Langley RR, Fan D, Wang X, Do KA, Kim SJ, Fidler IJ (2008) Targeting EGFR, VEGFR, and PDGFR on colon cancer cells is required for therapy. Clin Exp Metastasis 25:477–489PubMed Kuwai T, Nakamura T, Sasaki T, Kitadai Y, Kim JS, Langley RR, Fan D, Wang X, Do KA, Kim SJ, Fidler IJ (2008) Targeting EGFR, VEGFR, and PDGFR on colon cancer cells is required for therapy. Clin Exp Metastasis 25:477–489PubMed
32.
go back to reference Mao W, Irby R, Coppola D, Fu L, Wloch M, Turner J, Yu H, Garcia R, Jove R, Yeatman TJ (1997) Activation of c-Src by receptor tyrosine kinases in human colon cancer cells with high metastatic potential. Oncogene 15:3083–3090PubMed Mao W, Irby R, Coppola D, Fu L, Wloch M, Turner J, Yu H, Garcia R, Jove R, Yeatman TJ (1997) Activation of c-Src by receptor tyrosine kinases in human colon cancer cells with high metastatic potential. Oncogene 15:3083–3090PubMed
33.
go back to reference Irby RB, Mao W, Coppola D, Kang J, Loubeau JM, Trudeau W, Karl R, Fujita DJ, Jove R, Yeatman TH (1999) Activating SRC mutation in a subset of advanced human colon cancers. Nat Genet 21:187–190PubMed Irby RB, Mao W, Coppola D, Kang J, Loubeau JM, Trudeau W, Karl R, Fujita DJ, Jove R, Yeatman TH (1999) Activating SRC mutation in a subset of advanced human colon cancers. Nat Genet 21:187–190PubMed
34.
go back to reference Brinckerhoff CE, Matrisian LM (2002) Matrix metalloproteinases: a tail of a frog that became a prince. Nat Rev Mol Cell Biol 390:91–97 Brinckerhoff CE, Matrisian LM (2002) Matrix metalloproteinases: a tail of a frog that became a prince. Nat Rev Mol Cell Biol 390:91–97
35.
go back to reference Overall CM, Lopez-Otin C (2007) Strategies for MMP inhibition in cancer: innovations for the post-trial era. Nat Rev Cancer 2:657–672 Overall CM, Lopez-Otin C (2007) Strategies for MMP inhibition in cancer: innovations for the post-trial era. Nat Rev Cancer 2:657–672
36.
go back to reference Krüger A (2009) Functional genetic mouse models: promising tools for investigation of the proteolytic internet. Biol Chem 390:91–97PubMed Krüger A (2009) Functional genetic mouse models: promising tools for investigation of the proteolytic internet. Biol Chem 390:91–97PubMed
37.
go back to reference Krüger A, Kates RE, Edwards DR (2009) Avoiding spam in the proteolytic internet: future strategies for anti-metastatic MMP inhibition. Biochim Biophys Acta 1803:95–102PubMed Krüger A, Kates RE, Edwards DR (2009) Avoiding spam in the proteolytic internet: future strategies for anti-metastatic MMP inhibition. Biochim Biophys Acta 1803:95–102PubMed
38.
go back to reference Han J, Gao B, Jin X, Li Z, Sun Y, Song B (2012) Small interfering RNA down-regulation of β-catenin inhibits invasion of colon cancer cells in vitro. Med Sci Monit 18:BR273–BR280PubMedCentralPubMed Han J, Gao B, Jin X, Li Z, Sun Y, Song B (2012) Small interfering RNA down-regulation of β-catenin inhibits invasion of colon cancer cells in vitro. Med Sci Monit 18:BR273–BR280PubMedCentralPubMed
39.
go back to reference Damodharan U, Ganesan R, Radhakrishnan UC (2011) Expression of MMP2 and MMP9 gelatinases in human colon cancer cells. Appl Biochem Biotechnol 165:1245–1252PubMed Damodharan U, Ganesan R, Radhakrishnan UC (2011) Expression of MMP2 and MMP9 gelatinases in human colon cancer cells. Appl Biochem Biotechnol 165:1245–1252PubMed
40.
go back to reference Krüger A, Soeltl R, Sopov I, Kopitz C, Artl M, Magdolen V, Harbeck N, Gänsbacher B, Schmitt M (2001) Hydroxamate-type matrix metalloprotease inhibitor batimastat promotes liver metastasis. Cancer Res 61:1272–1275PubMed Krüger A, Soeltl R, Sopov I, Kopitz C, Artl M, Magdolen V, Harbeck N, Gänsbacher B, Schmitt M (2001) Hydroxamate-type matrix metalloprotease inhibitor batimastat promotes liver metastasis. Cancer Res 61:1272–1275PubMed
41.
go back to reference Clinchi B, Fransson A, Druvefors B, Hellsten A, Hakansson A, Gustavson B, Sjödahl R, Hakanson L (2007) Preoperative interleukin-6 production by mononuclear blood cells predicts survival after radical surgery for colorectal carcinoma. Cancer 109:1742–1749 Clinchi B, Fransson A, Druvefors B, Hellsten A, Hakansson A, Gustavson B, Sjödahl R, Hakanson L (2007) Preoperative interleukin-6 production by mononuclear blood cells predicts survival after radical surgery for colorectal carcinoma. Cancer 109:1742–1749
42.
go back to reference Gerg M, Kopitz C, Schaten S et al (2008) Distinct functionality of tumor cell-derived gelatinases during formation of liver metastases. Mol Cancer Res 6:341–351PubMed Gerg M, Kopitz C, Schaten S et al (2008) Distinct functionality of tumor cell-derived gelatinases during formation of liver metastases. Mol Cancer Res 6:341–351PubMed
43.
go back to reference Arlt M, Kopitz C, Pennington C et al (2002) Increase in gelatinase-specificity of matrix metalloprotease inhibitors correlates with antimetastatic efficacy in a T-cell lymphoma model. Cancer Res 62:5543–5550PubMed Arlt M, Kopitz C, Pennington C et al (2002) Increase in gelatinase-specificity of matrix metalloprotease inhibitors correlates with antimetastatic efficacy in a T-cell lymphoma model. Cancer Res 62:5543–5550PubMed
44.
go back to reference Seubert B, Grünwald B, Kobuch J et al (2014) TIMP1 creates a pre-metastatic niche in the liver through SDF/CXCR4-dependent neutrophil recruitment in mice. Hepatology 61:238–248PubMed Seubert B, Grünwald B, Kobuch J et al (2014) TIMP1 creates a pre-metastatic niche in the liver through SDF/CXCR4-dependent neutrophil recruitment in mice. Hepatology 61:238–248PubMed
45.
go back to reference Moller Sorensen N, Veigaard Sorensen I, Ornbierg Würtz S et al (2008) Biology and potential clinical implications of tissue inhibitor of metalloproteinases-1 in colorectal cancer treatment. Scand J Gastroenterol 43:774–786PubMed Moller Sorensen N, Veigaard Sorensen I, Ornbierg Würtz S et al (2008) Biology and potential clinical implications of tissue inhibitor of metalloproteinases-1 in colorectal cancer treatment. Scand J Gastroenterol 43:774–786PubMed
46.
go back to reference Holten-Andersen MN, Stephens RW, Nielsen HJ et al (2000) High preoperative plasma tissue inhibitor of metalloproteinases-1 levels are associated with short survival of patients with colorectal cancer. Clin Cancer Res 6:4292–4299PubMed Holten-Andersen MN, Stephens RW, Nielsen HJ et al (2000) High preoperative plasma tissue inhibitor of metalloproteinases-1 levels are associated with short survival of patients with colorectal cancer. Clin Cancer Res 6:4292–4299PubMed
48.
go back to reference Peinado H, Lavotshkin S, Lyden D (2011) The secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. Semin Cancer Biol 21:139–146PubMed Peinado H, Lavotshkin S, Lyden D (2011) The secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. Semin Cancer Biol 21:139–146PubMed
49.
go back to reference Gherardi E, Birchmeier W, Birchmeier C, Vande Woude G (2012) Targeting MET in cancer: rationale and progress. Nat Rev Cancer 12:89–103PubMed Gherardi E, Birchmeier W, Birchmeier C, Vande Woude G (2012) Targeting MET in cancer: rationale and progress. Nat Rev Cancer 12:89–103PubMed
50.
go back to reference Liu Y, Yu X-F, Zou J, Luo Z-H (2015) Prognostic value of c-MET in colorectal cancer: a meta-analysis. World J Gastroenterol 21:3706–3710PubMedCentralPubMed Liu Y, Yu X-F, Zou J, Luo Z-H (2015) Prognostic value of c-MET in colorectal cancer: a meta-analysis. World J Gastroenterol 21:3706–3710PubMedCentralPubMed
51.
go back to reference Liu Y, Li Q, Zhu L (2012) Expression of hepatocyte growth factor and c-MET in colon cancer: correlation with clinicopathological features and overall survival. Tumori 98:105–112PubMed Liu Y, Li Q, Zhu L (2012) Expression of hepatocyte growth factor and c-MET in colon cancer: correlation with clinicopathological features and overall survival. Tumori 98:105–112PubMed
52.
go back to reference Luraghi P, Schelter F, Krüger A, Boccaccio C (2012) The MET oncogene as a therapeutic target in cancer invasive growth. Front Pharmacol 3:164PubMedCentralPubMed Luraghi P, Schelter F, Krüger A, Boccaccio C (2012) The MET oncogene as a therapeutic target in cancer invasive growth. Front Pharmacol 3:164PubMedCentralPubMed
53.
go back to reference Kopitz C, Gerg M, Bandapalli OR et al (2007) Tissue inhibitor of metalloproteinases-1 promotes liver metastasis by induction of hepatocyte growth factor signaling. Cancer Res 67:8615–8623PubMed Kopitz C, Gerg M, Bandapalli OR et al (2007) Tissue inhibitor of metalloproteinases-1 promotes liver metastasis by induction of hepatocyte growth factor signaling. Cancer Res 67:8615–8623PubMed
55.
go back to reference Schelter F, Grandl M, Seubert B et al (2011) Tumor cell-derived Timp-1 is necessary for maintaining metastasis-promoting Met signaling via inhibition of Adam-10. Clin Exp Metastasis 28:793–802PubMed Schelter F, Grandl M, Seubert B et al (2011) Tumor cell-derived Timp-1 is necessary for maintaining metastasis-promoting Met signaling via inhibition of Adam-10. Clin Exp Metastasis 28:793–802PubMed
56.
go back to reference Schrötzlmair F, Kopitz C, Halbgewachs B (2010) Tissue inhibitor of metalloproteinases-1-induced scattered liver metastasis is mediated by host-derived urokinase type plasminogen activator. J Cell Mol Med 14:2760–2770PubMedCentralPubMed Schrötzlmair F, Kopitz C, Halbgewachs B (2010) Tissue inhibitor of metalloproteinases-1-induced scattered liver metastasis is mediated by host-derived urokinase type plasminogen activator. J Cell Mol Med 14:2760–2770PubMedCentralPubMed
57.
go back to reference Schelter F, Halbgewachs B, Bäumler P et al (2010) Tissue inhibitor of metalloproteinases-1 induced scattered liver metastasis is mediated by hypoxia-inducible factor-1α. Clin Exp Metastasis 28:91–99PubMed Schelter F, Halbgewachs B, Bäumler P et al (2010) Tissue inhibitor of metalloproteinases-1 induced scattered liver metastasis is mediated by hypoxia-inducible factor-1α. Clin Exp Metastasis 28:91–99PubMed
58.
go back to reference Schelter F, Gerg M, Halbgewachs B et al (2010) Identification of survival-independent metastasis enhancing role of hypoxia-inducible factor-1 alpha with a hypoxia-tolerant tumor cell line. J Biol Chem 285:26182–26189PubMedCentralPubMed Schelter F, Gerg M, Halbgewachs B et al (2010) Identification of survival-independent metastasis enhancing role of hypoxia-inducible factor-1 alpha with a hypoxia-tolerant tumor cell line. J Biol Chem 285:26182–26189PubMedCentralPubMed
59.
go back to reference Jung KK, Liu XW, Chirco R, Fridman R, Kim HR (2006) Identification of CD63 as a tissue inhibitor of metalloproteinase-1 interacting cell surface protein. EMBO J 25:3934–3942PubMedCentralPubMed Jung KK, Liu XW, Chirco R, Fridman R, Kim HR (2006) Identification of CD63 as a tissue inhibitor of metalloproteinase-1 interacting cell surface protein. EMBO J 25:3934–3942PubMedCentralPubMed
60.
go back to reference Toricelli M, Melo FH, Peres GB, Siulva DC, Jasiulionis MG (2013) Timp1 interacts with beta-1 integrin and CD63 along melanoma genesis and confers anoikis resistance by activating PI3 K signaling pathway independently of Akt phosphorylation. Mol Cancer 12:51 Toricelli M, Melo FH, Peres GB, Siulva DC, Jasiulionis MG (2013) Timp1 interacts with beta-1 integrin and CD63 along melanoma genesis and confers anoikis resistance by activating PI3 K signaling pathway independently of Akt phosphorylation. Mol Cancer 12:51
61.
go back to reference Puissegur MP, Mazure NM, Bertero P et al (2011) miR-210 is overexpressed in late stages of lung cancer and mediates mitochondrial alterations associated with modulation of HIF-1 activity. Cell Death Differ 18:465–478PubMedCentralPubMed Puissegur MP, Mazure NM, Bertero P et al (2011) miR-210 is overexpressed in late stages of lung cancer and mediates mitochondrial alterations associated with modulation of HIF-1 activity. Cell Death Differ 18:465–478PubMedCentralPubMed
62.
go back to reference Cui A, Seubert B, Stahl E et al (2014) Tissue inhibitor of metalloproteinases-1 induces a protumorigenic increase of miR-210 in lung adenocarcinoma cells and their exosomes. Oncogene in press Cui A, Seubert B, Stahl E et al (2014) Tissue inhibitor of metalloproteinases-1 induces a protumorigenic increase of miR-210 in lung adenocarcinoma cells and their exosomes. Oncogene in press
63.
go back to reference Cui H, Grosso S, Schelter F, Mari B, Krueger A (2012) On the prometastatic stress response in cancer therapies: evidence for a positive cooperation between TIMP-1, HIF-1α and miR-210. Front Pharmacol 3:134PubMedCentralPubMed Cui H, Grosso S, Schelter F, Mari B, Krueger A (2012) On the prometastatic stress response in cancer therapies: evidence for a positive cooperation between TIMP-1, HIF-1α and miR-210. Front Pharmacol 3:134PubMedCentralPubMed
64.
go back to reference Mason SD, Joyce JA (2011) Proteolytic networks in cancer. Trends Cell Biol 21:228–237PubMed Mason SD, Joyce JA (2011) Proteolytic networks in cancer. Trends Cell Biol 21:228–237PubMed
65.
go back to reference Sorensen NM, Byström P, Christensen IJ et al (2007) TIMP-1 is significantly associated with objective response and survival in metastatic colorectal cancer patients receiving a combination of irinotecan, 5-fluoruracil, and folinic acid. Clin Cancer Res 13:4117–4122PubMed Sorensen NM, Byström P, Christensen IJ et al (2007) TIMP-1 is significantly associated with objective response and survival in metastatic colorectal cancer patients receiving a combination of irinotecan, 5-fluoruracil, and folinic acid. Clin Cancer Res 13:4117–4122PubMed
66.
go back to reference Stein U, Walther W, Arlt F et al (2009) MACC1, a newly identified key regulator of HGF-MET signaling, predicts colon cancer metastasis. Nat Med 15:59–67PubMed Stein U, Walther W, Arlt F et al (2009) MACC1, a newly identified key regulator of HGF-MET signaling, predicts colon cancer metastasis. Nat Med 15:59–67PubMed
67.
go back to reference Isella C, Mellano A, Galimi F et al (2013) MACC1 mRNA levels predict cancer recurrence after resection of colorectal cancer liver metastases. Ann Surg 257:1089–1095PubMed Isella C, Mellano A, Galimi F et al (2013) MACC1 mRNA levels predict cancer recurrence after resection of colorectal cancer liver metastases. Ann Surg 257:1089–1095PubMed
68.
go back to reference Stein U, Burock S, Herrmann P et al (2012) Circulating MACC1 transcripts in colorectal cancer patient plasma predict metastasis and prognosis. PLoS ONE 7:e49249PubMedCentralPubMed Stein U, Burock S, Herrmann P et al (2012) Circulating MACC1 transcripts in colorectal cancer patient plasma predict metastasis and prognosis. PLoS ONE 7:e49249PubMedCentralPubMed
69.
go back to reference Stein U, Dahlmann M, Walther W (2010) MACC1 – more then metastasis? Facts and predictions about a novel gene. J Mol Med 88:11–18PubMed Stein U, Dahlmann M, Walther W (2010) MACC1 – more then metastasis? Facts and predictions about a novel gene. J Mol Med 88:11–18PubMed
70.
go back to reference Arlt F, Stein U (2009) Colon cancer metastasis: MACC1 and Met as metastatic pacemakers. Int J Biochem Cell Biol 41:2356–2359PubMed Arlt F, Stein U (2009) Colon cancer metastasis: MACC1 and Met as metastatic pacemakers. Int J Biochem Cell Biol 41:2356–2359PubMed
71.
go back to reference Zhang Y, Wang Z, Chen M et al (2012) MicroRNA-143 targets MACC1 to inhibit cell invasion and migration in colorectal cancer. Mol Cancer 11:23PubMedCentralPubMed Zhang Y, Wang Z, Chen M et al (2012) MicroRNA-143 targets MACC1 to inhibit cell invasion and migration in colorectal cancer. Mol Cancer 11:23PubMedCentralPubMed
72.
go back to reference Ostman A, Hellberg C, Böhmer FD (2006) Protein-tyrosine phosphatases and cancer. Nat Rev Cancer 6:307–320PubMed Ostman A, Hellberg C, Böhmer FD (2006) Protein-tyrosine phosphatases and cancer. Nat Rev Cancer 6:307–320PubMed
73.
go back to reference Bessette DC, Qui D, Pallen C (2008) PRL PTPs: mediators and markers of cancer prognosis. Cancer Metastasis Rev 27:231–252PubMed Bessette DC, Qui D, Pallen C (2008) PRL PTPs: mediators and markers of cancer prognosis. Cancer Metastasis Rev 27:231–252PubMed
74.
go back to reference Lee SK, Han YM, Yun J et al (2012) Phosphatase of regenerating liver-3 promotes migration and invasion by upregulating matrix metalloproteinases-7 in human colorectal cancer cells. Int J Cancer 131:E190–E203PubMed Lee SK, Han YM, Yun J et al (2012) Phosphatase of regenerating liver-3 promotes migration and invasion by upregulating matrix metalloproteinases-7 in human colorectal cancer cells. Int J Cancer 131:E190–E203PubMed
75.
go back to reference Al-Aidaroos AQ, Yuen HD, Guo K (2013) Metastasis-associated PRL-3 induces EGFR activation and addiction in cancer cells. J Clin Invest 123:3459–3471PubMedCentralPubMed Al-Aidaroos AQ, Yuen HD, Guo K (2013) Metastasis-associated PRL-3 induces EGFR activation and addiction in cancer cells. J Clin Invest 123:3459–3471PubMedCentralPubMed
76.
go back to reference Al-Aidaroos AQ, Zeng Q (2010) PRL3 phosphatase and cancer metastasis. J Cell Biochem 111:1087–1098PubMed Al-Aidaroos AQ, Zeng Q (2010) PRL3 phosphatase and cancer metastasis. J Cell Biochem 111:1087–1098PubMed
77.
go back to reference Saha S, Bardelli A, Buckhaults P et al (2001) A phosphatase associated with metastasis of colorectal cancer. Science 294:1343–1346PubMed Saha S, Bardelli A, Buckhaults P et al (2001) A phosphatase associated with metastasis of colorectal cancer. Science 294:1343–1346PubMed
78.
go back to reference Bardelli A, Saha S, Sager JA et al (2003) PRL-3 expression in metastatic cancers. Clin Cancer Res 9:5607–5615PubMed Bardelli A, Saha S, Sager JA et al (2003) PRL-3 expression in metastatic cancers. Clin Cancer Res 9:5607–5615PubMed
79.
go back to reference Zeng Q, Dong JM, Guo K et al (2003) PRL-3 and PRL-1 promote cell migration, invasion and metastasis. Cancer Res 63:2716–2722PubMed Zeng Q, Dong JM, Guo K et al (2003) PRL-3 and PRL-1 promote cell migration, invasion and metastasis. Cancer Res 63:2716–2722PubMed
80.
go back to reference Jiang Y, Liu XQ, Rajput A et al (2011) Phosphatase PRL-3 is a direct regulatory target of TGFβ in colon cancer metastasis. Cancer Res 71:234–244PubMedCentralPubMed Jiang Y, Liu XQ, Rajput A et al (2011) Phosphatase PRL-3 is a direct regulatory target of TGFβ in colon cancer metastasis. Cancer Res 71:234–244PubMedCentralPubMed
81.
go back to reference Guo K, Tang JP, Tan CP, Wang H, Zeng Q (2008) Monoclonal antibodies target intracellular PRL phosphatases to inhibit cancer metastases in mice. Cancer Biol Ther 7:750–757PubMed Guo K, Tang JP, Tan CP, Wang H, Zeng Q (2008) Monoclonal antibodies target intracellular PRL phosphatases to inhibit cancer metastases in mice. Cancer Biol Ther 7:750–757PubMed
82.
go back to reference Guo K, Tang JP, Al-Aidaroos AQ et al (2012) Engineering the first chimeric antibody in targeting intracellular PRL-3 oncoprotein for cancer therapy in mice. Oncotarget 3:158–171PubMedCentralPubMed Guo K, Tang JP, Al-Aidaroos AQ et al (2012) Engineering the first chimeric antibody in targeting intracellular PRL-3 oncoprotein for cancer therapy in mice. Oncotarget 3:158–171PubMedCentralPubMed
83.
go back to reference Weidle UH, Eggle D, Klostermann S (2009) L1-CAM as a target for treatment of cancer with monoclonal antibodies. Anticancer Res 29:4919–4931PubMed Weidle UH, Eggle D, Klostermann S (2009) L1-CAM as a target for treatment of cancer with monoclonal antibodies. Anticancer Res 29:4919–4931PubMed
84.
go back to reference Gavert S, Conacci-Sorell M, Gast D et al (2005) L1, a novel target of beta-catenin signaling, transforms cells and is expressed at the invasive front of colon tumors. J Cell Biol 168:633–642PubMedCentralPubMed Gavert S, Conacci-Sorell M, Gast D et al (2005) L1, a novel target of beta-catenin signaling, transforms cells and is expressed at the invasive front of colon tumors. J Cell Biol 168:633–642PubMedCentralPubMed
85.
go back to reference Gerg M, Kopitz C, Schaten S et al (2008) Distinct functionality of tumor-cell derived gelatinases during formation of liver metastases. Mol Cancer Res 6:341–351PubMed Gerg M, Kopitz C, Schaten S et al (2008) Distinct functionality of tumor-cell derived gelatinases during formation of liver metastases. Mol Cancer Res 6:341–351PubMed
86.
go back to reference Weinspach D, Seubert B, Schaten S et al (2014) Role of L1 cell adhesion molecule (L1CAM) in the metastatic cascade: promotion of dissemination, colonization and metastatic growth. Clin Exp Metastasis 31:87–100PubMed Weinspach D, Seubert B, Schaten S et al (2014) Role of L1 cell adhesion molecule (L1CAM) in the metastatic cascade: promotion of dissemination, colonization and metastatic growth. Clin Exp Metastasis 31:87–100PubMed
87.
go back to reference Lim IT, Brown S, Mobashery S (2004) A convenient synthesis of a selective gelatinase inhibitor as an antimetastatic agent. J Org Chem 69:3572–3573PubMed Lim IT, Brown S, Mobashery S (2004) A convenient synthesis of a selective gelatinase inhibitor as an antimetastatic agent. J Org Chem 69:3572–3573PubMed
88.
go back to reference Coutelle O, Nyakatura G, Tsudien S et al (1998) The neural cell adhesion molecule L1: genomic organisation and differential splicing is conserved between man and the pufferfish fugu. Gene 208:7–15PubMed Coutelle O, Nyakatura G, Tsudien S et al (1998) The neural cell adhesion molecule L1: genomic organisation and differential splicing is conserved between man and the pufferfish fugu. Gene 208:7–15PubMed
89.
go back to reference De Angelis E, Brummendorf T, Cheng L, Lemmon V, Kenwrick S (2001) Alternative use of a mini exon of the L1 gene affects L1 binding to neural ligands. J Biol Chem 276:32738–32742PubMed De Angelis E, Brummendorf T, Cheng L, Lemmon V, Kenwrick S (2001) Alternative use of a mini exon of the L1 gene affects L1 binding to neural ligands. J Biol Chem 276:32738–32742PubMed
90.
go back to reference Kamiguchi H, Long KE, Pendergast M, Schaefer AW, Rapoport I, Kirchhausen T, Lemmon V (1998) The neural cell adhesion molecule L1 interacts with the AP-2 adaptor and is endocytosed via its clathrin-mediated pathway. J Neurosc 18:5311–5321 Kamiguchi H, Long KE, Pendergast M, Schaefer AW, Rapoport I, Kirchhausen T, Lemmon V (1998) The neural cell adhesion molecule L1 interacts with the AP-2 adaptor and is endocytosed via its clathrin-mediated pathway. J Neurosc 18:5311–5321
91.
go back to reference Hauser S, Bickel L, Weinspach D et al (2011) Full-length L1CAM and not its ∆2∆27 splice variant promotes metastasis through induction of gelatinase expression. PLoS One 6:e18989PubMedCentralPubMed Hauser S, Bickel L, Weinspach D et al (2011) Full-length L1CAM and not its ∆2∆27 splice variant promotes metastasis through induction of gelatinase expression. PLoS One 6:e18989PubMedCentralPubMed
92.
go back to reference Gast D, Riedle S, Kiefel H et al (2008) The RGD integrin binding site in human L1-CAM is important for nuclear signaling. Exp Cell Res 314:2411–2418PubMed Gast D, Riedle S, Kiefel H et al (2008) The RGD integrin binding site in human L1-CAM is important for nuclear signaling. Exp Cell Res 314:2411–2418PubMed
93.
go back to reference Gavert N, Ben-Shmuel A, Lemmon V, Brabletz T, Ben-Zeév A (2010) Nuclear factor-kappa B signaling and ezrin are essential for L1-mediated metastasis of colon cancer cells. J Cell Sci 123:2135–2143PubMed Gavert N, Ben-Shmuel A, Lemmon V, Brabletz T, Ben-Zeév A (2010) Nuclear factor-kappa B signaling and ezrin are essential for L1-mediated metastasis of colon cancer cells. J Cell Sci 123:2135–2143PubMed
94.
go back to reference Gast D, Riedle S, Issa Y et al (2008) The cytoplasmic part of L1-CAM controls growth and gene expression in human tumors that is reversed by therapeutic antibodies. Oncogene 27:1281–1289PubMed Gast D, Riedle S, Issa Y et al (2008) The cytoplasmic part of L1-CAM controls growth and gene expression in human tumors that is reversed by therapeutic antibodies. Oncogene 27:1281–1289PubMed
95.
go back to reference Arlt M, Novak-Hofer I, Gast D et al (2006) Efficient inhibition of intra-peritoneal tumor growth and dissemination of human ovarian carcinoma cells in nude mice by anti-L1-cell adhesion molecule monoclonal antibody treatment. Cancer Res 66:936–943PubMed Arlt M, Novak-Hofer I, Gast D et al (2006) Efficient inhibition of intra-peritoneal tumor growth and dissemination of human ovarian carcinoma cells in nude mice by anti-L1-cell adhesion molecule monoclonal antibody treatment. Cancer Res 66:936–943PubMed
96.
go back to reference Wolterink S, Moldenhauer G, Fogel M et al (2010) Therapeutic antibodies to human L1CAM: functional characterization and application in a mouse model for ovarian carcinoma. Cancer Res 70:2504–2515PubMed Wolterink S, Moldenhauer G, Fogel M et al (2010) Therapeutic antibodies to human L1CAM: functional characterization and application in a mouse model for ovarian carcinoma. Cancer Res 70:2504–2515PubMed
97.
go back to reference Schäfer H, Dieckmann C, Kornijenko O et al (2012) Combined treatment of L1CAM antibodies and cytotstatic drugs improve the therapeutic response to pancreatic and ovarian carcinoma. Cancer Lett 319:66–82PubMed Schäfer H, Dieckmann C, Kornijenko O et al (2012) Combined treatment of L1CAM antibodies and cytotstatic drugs improve the therapeutic response to pancreatic and ovarian carcinoma. Cancer Lett 319:66–82PubMed
98.
go back to reference Kaifi JT, Reichelt U, Quaas A et al (2007) L1 is associated with micrometastatic spread and poor outcome in colorectal cancer. Mod Pathol 20:1183–1190PubMed Kaifi JT, Reichelt U, Quaas A et al (2007) L1 is associated with micrometastatic spread and poor outcome in colorectal cancer. Mod Pathol 20:1183–1190PubMed
99.
go back to reference Boo YJ, Park JM, Kim J et al (2007) L1 expression as a marker for poor diagnosis, tumor progression, and short survival in patients with colorectal cancer. Ann Surg Oncol 14:1703–1711PubMed Boo YJ, Park JM, Kim J et al (2007) L1 expression as a marker for poor diagnosis, tumor progression, and short survival in patients with colorectal cancer. Ann Surg Oncol 14:1703–1711PubMed
100.
go back to reference Gavert N, Sheffer M, Raveh S et al (2007) Expression of L1-CAM and ADAM 10 in human colon cancer cells induces metastasis. Cancer Res 67:7703–7712PubMed Gavert N, Sheffer M, Raveh S et al (2007) Expression of L1-CAM and ADAM 10 in human colon cancer cells induces metastasis. Cancer Res 67:7703–7712PubMed
101.
go back to reference Gavert N, Conacci-Sorrell M, Gast D et al (2005) L1, a novel target of beta-catenin signaling, transforms cells and is expressed at the invasive front of colon cancers. J Cell Biol 168:633–642PubMedCentralPubMed Gavert N, Conacci-Sorrell M, Gast D et al (2005) L1, a novel target of beta-catenin signaling, transforms cells and is expressed at the invasive front of colon cancers. J Cell Biol 168:633–642PubMedCentralPubMed
102.
go back to reference Gavert N, Ben-Shmuel A, Raveh S, Ben-Zeév A (2008) L1-CAM in cancerous tissues. Expert Opin Biol Ther 8:1749–1757PubMed Gavert N, Ben-Shmuel A, Raveh S, Ben-Zeév A (2008) L1-CAM in cancerous tissues. Expert Opin Biol Ther 8:1749–1757PubMed
103.
go back to reference Trerotola M, Cantanelli P, Guerra E et al (2013) Upregulation of Trop-2 quantitatively stimulates human cancer growth. Oncogene 32:222–233PubMed Trerotola M, Cantanelli P, Guerra E et al (2013) Upregulation of Trop-2 quantitatively stimulates human cancer growth. Oncogene 32:222–233PubMed
104.
go back to reference Wang J, Day R, Dong Y, Weintraub SJ, Michel L (2008) Identification of Trop-2 as an oncogene and an attractive target in colon cancers. Mol Cancer Ther 7:280–285PubMed Wang J, Day R, Dong Y, Weintraub SJ, Michel L (2008) Identification of Trop-2 as an oncogene and an attractive target in colon cancers. Mol Cancer Ther 7:280–285PubMed
105.
go back to reference Cubas R, Zhang S, Li M, Chen C, Yao Q (2010) Trop2 expression contributes to tumor pathogenesis by activating the ERK MAPK pathway. Mol Cancer 9:253PubMedCentralPubMed Cubas R, Zhang S, Li M, Chen C, Yao Q (2010) Trop2 expression contributes to tumor pathogenesis by activating the ERK MAPK pathway. Mol Cancer 9:253PubMedCentralPubMed
106.
go back to reference Guerra E, Trerotola M, Aloisi AL et al (2013) The Trop-2 signaling network in cancer growth. Oncogene 32:1594–1600PubMed Guerra E, Trerotola M, Aloisi AL et al (2013) The Trop-2 signaling network in cancer growth. Oncogene 32:1594–1600PubMed
107.
go back to reference Ohmachi T, Taneka Miori K, Inoue H, Yanaga K, Mori M (2006) Clinical significance of TROP2 expression in colorectal cancer. Clin Cancer Res 12:3057–3063PubMed Ohmachi T, Taneka Miori K, Inoue H, Yanaga K, Mori M (2006) Clinical significance of TROP2 expression in colorectal cancer. Clin Cancer Res 12:3057–3063PubMed
108.
go back to reference Fang YH, Lu ZH, Wang GQ et al (2007) Elevated expression of MMP7, TROP2, and Survivin are associated with survival, disease recurrence, and liver metastasis of colon cancer. Int J Colorectal Dis 24:875–884 Fang YH, Lu ZH, Wang GQ et al (2007) Elevated expression of MMP7, TROP2, and Survivin are associated with survival, disease recurrence, and liver metastasis of colon cancer. Int J Colorectal Dis 24:875–884
109.
go back to reference Alberti S, Trerotola G, Vaca R et al (2007) TROP2 is a major determinant of growth and metastatic spreading of human cancer. J Clin Oncol 25(18S):10510 Alberti S, Trerotola G, Vaca R et al (2007) TROP2 is a major determinant of growth and metastatic spreading of human cancer. J Clin Oncol 25(18S):10510
110.
go back to reference Donato R, Cannon BR, Sorci G et al (2013) Functions of S100 proteins. Curr Mol Med 1:24–57 Donato R, Cannon BR, Sorci G et al (2013) Functions of S100 proteins. Curr Mol Med 1:24–57
111.
go back to reference Sack U, Walther W, Scuderio D et al (2011) S100A4-induced cell motility and metastasis is restricted by the Wnt/β-catenin pathway inhibitor calcimycin in colon cancer cells. Mol Biol Cell 22:3344–3354PubMedCentralPubMed Sack U, Walther W, Scuderio D et al (2011) S100A4-induced cell motility and metastasis is restricted by the Wnt/β-catenin pathway inhibitor calcimycin in colon cancer cells. Mol Biol Cell 22:3344–3354PubMedCentralPubMed
112.
go back to reference Cho YG, Kim CJ, Nam SW (2005) Overexpression of S100A4 is closely associated with progression of colorectal cancer. World J Gastroenterol 11:4852–4856PubMedCentralPubMed Cho YG, Kim CJ, Nam SW (2005) Overexpression of S100A4 is closely associated with progression of colorectal cancer. World J Gastroenterol 11:4852–4856PubMedCentralPubMed
113.
go back to reference Boye K, Nesland JM, Sandstad B, Maelandsmo GM, Flatmark K (2007) Nuclear S100A4 is a novel prognostic marker in colorectal cancer. Eur J Cancer 46:2919–2925 Boye K, Nesland JM, Sandstad B, Maelandsmo GM, Flatmark K (2007) Nuclear S100A4 is a novel prognostic marker in colorectal cancer. Eur J Cancer 46:2919–2925
114.
go back to reference Dahlmann M, Sack U, Herrmann P et al (2012) Systemic shRNA mediated knock down of S100A4 in colorectal cancer xenografted mice reduces metastasis formation. Oncotarget 3:783–797PubMedCentralPubMed Dahlmann M, Sack U, Herrmann P et al (2012) Systemic shRNA mediated knock down of S100A4 in colorectal cancer xenografted mice reduces metastasis formation. Oncotarget 3:783–797PubMedCentralPubMed
115.
go back to reference Stein U, Arlt F, Walther W et al (2006) The metastasis-associated gene S100A4 is a novel target of beta-catenin/T-cell factor signaling in colon cancer. Gastroenterology 131:1486–1500PubMed Stein U, Arlt F, Walther W et al (2006) The metastasis-associated gene S100A4 is a novel target of beta-catenin/T-cell factor signaling in colon cancer. Gastroenterology 131:1486–1500PubMed
116.
go back to reference Ding Q, Chang CJ, Xie X et al (2011) APOBEC3G promotes liver metastasis in an orthotopic mouse model of colorectal cancer and predicts human hepatic metastasis. J Clin Invest 121:4526–4536PubMedCentralPubMed Ding Q, Chang CJ, Xie X et al (2011) APOBEC3G promotes liver metastasis in an orthotopic mouse model of colorectal cancer and predicts human hepatic metastasis. J Clin Invest 121:4526–4536PubMedCentralPubMed
117.
go back to reference Wang Q, Zhang YN, Lin GL et al (2012) S100P, a potential novel prognostic marker in colorectal cancer. Oncol Rep 28:303–310PubMed Wang Q, Zhang YN, Lin GL et al (2012) S100P, a potential novel prognostic marker in colorectal cancer. Oncol Rep 28:303–310PubMed
118.
go back to reference Chandramouli A, Mercado-Pimentel ME, Hutchinson A et al (2010) The induction of S100P expression by the prostaglandin E2 (PGE2/EP4 receptor signaling pathway in colon cancer cells. Cancer Biol Ther 28:303–310 Chandramouli A, Mercado-Pimentel ME, Hutchinson A et al (2010) The induction of S100P expression by the prostaglandin E2 (PGE2/EP4 receptor signaling pathway in colon cancer cells. Cancer Biol Ther 28:303–310
119.
go back to reference Dong L, Wang F, Yin X et al (2014) Overexpression of S100P promotes colorectal cancer metastasis and decreases chemosensitivity to 5-FU in vitro. Mol Cell Biochem 389:257–264PubMed Dong L, Wang F, Yin X et al (2014) Overexpression of S100P promotes colorectal cancer metastasis and decreases chemosensitivity to 5-FU in vitro. Mol Cell Biochem 389:257–264PubMed
120.
go back to reference Wong H, Schotz MC (2002) The lipase gene family. J Lipid Res 43:993–999PubMed Wong H, Schotz MC (2002) The lipase gene family. J Lipid Res 43:993–999PubMed
121.
go back to reference Nomura DK, Long JZ, Niessen S et al (2010) Monoacylglycerol lipase regulates a fatty acid network that promotes cancer pathogenesis. Cell 140:49–61PubMedCentralPubMed Nomura DK, Long JZ, Niessen S et al (2010) Monoacylglycerol lipase regulates a fatty acid network that promotes cancer pathogenesis. Cell 140:49–61PubMedCentralPubMed
122.
go back to reference Notarnicola M, Messa C, Caruso MG (2012) A significant role of lipogenic enzymes in colorectal cancer. Anticancer Res 32:2585–2590PubMed Notarnicola M, Messa C, Caruso MG (2012) A significant role of lipogenic enzymes in colorectal cancer. Anticancer Res 32:2585–2590PubMed
123.
go back to reference Silinsky J, Grimes C, Driscoll T et al (2013) CD 133+ and CXCR4+ colon cancer cells as a marker for lymph node metastasis. J Surg Res 185:113–118PubMed Silinsky J, Grimes C, Driscoll T et al (2013) CD 133+ and CXCR4+ colon cancer cells as a marker for lymph node metastasis. J Surg Res 185:113–118PubMed
124.
go back to reference Shmelkov SV, Butler JM, Hooper AT et al (2008) CD133 expression is not restricted to stem cells, and both CD133+ and CD133− colon cancer cells initiate tumors. J Clin Invest 118:2111–2120PubMedCentralPubMed Shmelkov SV, Butler JM, Hooper AT et al (2008) CD133 expression is not restricted to stem cells, and both CD133+ and CD133− colon cancer cells initiate tumors. J Clin Invest 118:2111–2120PubMedCentralPubMed
125.
go back to reference Jaszczur M, Bertram JG, Pham P, Scharff MD, Goodman MF (2013) AID and Apobec3G haphazard deamination and mutational diversity. Cell Mol Life Sci 70:3089–3108PubMedCentralPubMed Jaszczur M, Bertram JG, Pham P, Scharff MD, Goodman MF (2013) AID and Apobec3G haphazard deamination and mutational diversity. Cell Mol Life Sci 70:3089–3108PubMedCentralPubMed
126.
go back to reference Huang J, Liang Z, Yang B, Tian H, Ma J, Zhang H (2007) Derepression of microRNA-mediated protein translation inhibition by apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3G (APOBEC3G) and its family members. J Biol Chem 282:33632–33640PubMed Huang J, Liang Z, Yang B, Tian H, Ma J, Zhang H (2007) Derepression of microRNA-mediated protein translation inhibition by apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3G (APOBEC3G) and its family members. J Biol Chem 282:33632–33640PubMed
127.
go back to reference Gallois-Montbrun S, Kramer B, Swanson CM et al (2007) Antiviral protein APOBEC3G localizes to ribonucleoprotein complexes found in P bodies and stress granules. J Virol 81:2165–2178PubMedCentralPubMed Gallois-Montbrun S, Kramer B, Swanson CM et al (2007) Antiviral protein APOBEC3G localizes to ribonucleoprotein complexes found in P bodies and stress granules. J Virol 81:2165–2178PubMedCentralPubMed
128.
go back to reference Ranganathan P, Weaver KL, Capobianco AJ (2011) Notch signalling in solid tumors: a little bit of everything but not all the time. Nat Rev Cancer 11:338–351PubMed Ranganathan P, Weaver KL, Capobianco AJ (2011) Notch signalling in solid tumors: a little bit of everything but not all the time. Nat Rev Cancer 11:338–351PubMed
129.
go back to reference Sonoshita M, Aoki M, Fuwa H et al (2011) Suppression of colon cancer metastasis by Aes through inhibition of Notch signaling. Cancer Cell 19:125–137PubMed Sonoshita M, Aoki M, Fuwa H et al (2011) Suppression of colon cancer metastasis by Aes through inhibition of Notch signaling. Cancer Cell 19:125–137PubMed
130.
go back to reference Katz LH, Li Y, Chen J-S et al (2013) Targeting TGFβ signaling in cancer. Exp Opin Ther Targets 17:743–760 Katz LH, Li Y, Chen J-S et al (2013) Targeting TGFβ signaling in cancer. Exp Opin Ther Targets 17:743–760
131.
132.
go back to reference Zhang B, Halder SK, Kashikar ND et al (2010) Antimetastatic role of Smad4 signaling in colorectal cancer. Gastroenterology 138:969–980PubMedCentralPubMed Zhang B, Halder SK, Kashikar ND et al (2010) Antimetastatic role of Smad4 signaling in colorectal cancer. Gastroenterology 138:969–980PubMedCentralPubMed
133.
go back to reference Halder SK, Rachakonda G, Deane NG, Datta PK (2008) Smad7 induces hepatic metastasis in colorectal cancer. Br J Cancer 99:957–965PubMedCentralPubMed Halder SK, Rachakonda G, Deane NG, Datta PK (2008) Smad7 induces hepatic metastasis in colorectal cancer. Br J Cancer 99:957–965PubMedCentralPubMed
134.
go back to reference Calon A, Espinet E, Palomo-Ponce S et al (2012) Dependency of colorectal cancer on a TGF-β-driven program in stromal cells for metastasis initiation. Cancer Cell 22:571–584PubMedCentralPubMed Calon A, Espinet E, Palomo-Ponce S et al (2012) Dependency of colorectal cancer on a TGF-β-driven program in stromal cells for metastasis initiation. Cancer Cell 22:571–584PubMedCentralPubMed
135.
go back to reference Markowitz SD, Bertagnolli MM (2009) Molecular origins of cancer: molecular basis of colorectal cancer. N Engl J Med 361:2449–2460PubMedCentralPubMed Markowitz SD, Bertagnolli MM (2009) Molecular origins of cancer: molecular basis of colorectal cancer. N Engl J Med 361:2449–2460PubMedCentralPubMed
136.
go back to reference Shibayama M, Maak M, Nitsche U et al (2011) Prediction of metastasis and recurrence in colorectal cancer based on gene expression analysis: ready for the clinic? Cancers 3:2858–2869PubMedCentralPubMed Shibayama M, Maak M, Nitsche U et al (2011) Prediction of metastasis and recurrence in colorectal cancer based on gene expression analysis: ready for the clinic? Cancers 3:2858–2869PubMedCentralPubMed
137.
go back to reference Taketo MM (2011) Reflections on the spread of metastasis to cancer prevention. Cancer Prev Res 4:324–328 Taketo MM (2011) Reflections on the spread of metastasis to cancer prevention. Cancer Prev Res 4:324–328
138.
go back to reference Ye L-C, Liu T-S, Ren L et al (2013) Randomized controlled clinical trial of cetuximab plus chemotherapy for patients with KRAS unresectable colorectal liver-limited metastasis. J Clin Oncol 31:1931–1938PubMed Ye L-C, Liu T-S, Ren L et al (2013) Randomized controlled clinical trial of cetuximab plus chemotherapy for patients with KRAS unresectable colorectal liver-limited metastasis. J Clin Oncol 31:1931–1938PubMed
Metadata
Title
Molecular targets and pathways involved in liver metastasis of colorectal cancer
Authors
Ulrich H. Weidle
Fabian Birzele
Achim Krüger
Publication date
01-08-2015
Publisher
Springer Netherlands
Published in
Clinical & Experimental Metastasis / Issue 6/2015
Print ISSN: 0262-0898
Electronic ISSN: 1573-7276
DOI
https://doi.org/10.1007/s10585-015-9732-3

Other articles of this Issue 6/2015

Clinical & Experimental Metastasis 6/2015 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