Skip to main content
Top
Published in: Virchows Archiv 3/2012

01-09-2012 | Review and Perspectives

Understanding molecular mechanisms in peritoneal dissemination of colorectal cancer

Future possibilities for personalised treatment by use of biomarkers

Authors: E. M. V. de Cuba, R. Kwakman, M. van Egmond, L. J. W. Bosch, H. J. Bonjer, G. A. Meijer, E. A. te Velde

Published in: Virchows Archiv | Issue 3/2012

Login to get access

Abstract

When colorectal cancer (CRC) metastasizes, this is mostly to the liver via the portal circulation. In addition, 10–25 % of CRC patients eventually show metastases in the peritoneum. A selection of these patients is treated with cytoreductive surgery (CRS) and hyperthermic intra-peritoneal chemotherapy (HIPEC). However, several clinical needs still exist in which biomarkers could play an important role. Relatively little is known about the biology of peritoneal spread of CRC. The development of peritoneal metastases (PM) involves several steps, including: detachment of malignant cells; anoikis evasion; attachment to and invasion of the peritoneal surface ultimately ending in a colonization phase in which the malignant cells thrive in the newly formed niche. In this paper, we provide an overview of molecules associated with peritoneal dissemination and explore the clinical possibilities of these candidate biomarkers. A literature search was conducted using the PubMed database of the U.S. National Library of Medicine and Medline to identify studies on the biological behaviour of PM of CRC. In a series of over 100 studies on PM published between 1990 and 2010, IGF-1, HIF1α, VEGF, EGFR and ITGB1 emerge as the most interesting candidates for possible clinical application. Even though these promising candidate biomarkers have been identified, all of these require extensive further validation prior to clinical application. Yet, the pace of the omics revolution makes that the question is not if, but when biomarkers will be introduced to improve diagnosis and ultimately outcome of patients with PM due to CRC.
Literature
1.
go back to reference Parkin DM, Bray F, Ferlay J, Pisani P (2005) Global cancer statistics, 2002. CA Cancer J Clin 55(2):74–108PubMedCrossRef Parkin DM, Bray F, Ferlay J, Pisani P (2005) Global cancer statistics, 2002. CA Cancer J Clin 55(2):74–108PubMedCrossRef
2.
go back to reference Bird NC, Mangnall D, Majeed AW (2006) Biology of colorectal liver metastases: a review. J Surg Oncol 94(1):68–80PubMedCrossRef Bird NC, Mangnall D, Majeed AW (2006) Biology of colorectal liver metastases: a review. J Surg Oncol 94(1):68–80PubMedCrossRef
3.
go back to reference Koppe MJ, Boerman OC, Oyen WJG, Bleichrodt RP (2006) Peritoneal carcinomatosis of colorectal origin. Ann Surg 243(2):212–222PubMedCrossRef Koppe MJ, Boerman OC, Oyen WJG, Bleichrodt RP (2006) Peritoneal carcinomatosis of colorectal origin. Ann Surg 243(2):212–222PubMedCrossRef
4.
go back to reference Maggiori L, Elias D (2010) Curative treatment of colorectal peritoneal carcinomatosis: current status and future trends. Eur J Surg Oncol 36(7):599–603PubMedCrossRef Maggiori L, Elias D (2010) Curative treatment of colorectal peritoneal carcinomatosis: current status and future trends. Eur J Surg Oncol 36(7):599–603PubMedCrossRef
5.
go back to reference Cao C, Yan TD, Black D, Morris DL (2009) A systematic review and meta-analysis of cytoreductive surgery with perioperative intraperitoneal chemotherapy for peritoneal carcinomatosis of colorectal origin. Ann Surg Oncol 16(8):2152–2165PubMedCrossRef Cao C, Yan TD, Black D, Morris DL (2009) A systematic review and meta-analysis of cytoreductive surgery with perioperative intraperitoneal chemotherapy for peritoneal carcinomatosis of colorectal origin. Ann Surg Oncol 16(8):2152–2165PubMedCrossRef
6.
go back to reference Klaver YLB, Lemmens VEPP, Creemers GJ, Rutten HJT, Nienhuijs SW, de Hingh IHJT (2011) Population-based survival of patients with peritoneal carcinomatosis from colorectal origin in the era of increasing use of palliative chemotherapy. Ann Oncol 22(10):2250–2256PubMedCrossRef Klaver YLB, Lemmens VEPP, Creemers GJ, Rutten HJT, Nienhuijs SW, de Hingh IHJT (2011) Population-based survival of patients with peritoneal carcinomatosis from colorectal origin in the era of increasing use of palliative chemotherapy. Ann Oncol 22(10):2250–2256PubMedCrossRef
7.
go back to reference Sugarbaker PH (1995) Patient selection and treatment of peritoneal carcinomatosis from colorectal and appendiceal cancer. World J Surg 19(2):235–240PubMedCrossRef Sugarbaker PH (1995) Patient selection and treatment of peritoneal carcinomatosis from colorectal and appendiceal cancer. World J Surg 19(2):235–240PubMedCrossRef
8.
go back to reference Sugarbaker PH, Jablonski KA (1995) Prognostic features of 51 colorectal and 130 appendiceal cancer patients with peritoneal carcinomatosis treated by cytoreductive surgery and intraperitoneal chemotherapy. Ann Surg 221(2):124–132PubMedCrossRef Sugarbaker PH, Jablonski KA (1995) Prognostic features of 51 colorectal and 130 appendiceal cancer patients with peritoneal carcinomatosis treated by cytoreductive surgery and intraperitoneal chemotherapy. Ann Surg 221(2):124–132PubMedCrossRef
9.
go back to reference Cavaliere F, De Simone M, Virzì S et al (2011) Prognostic factors and oncologic outcome in 146 patients with colorectal peritoneal carcinomatosis treated with cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy: Italian multicenter study S.I.T.I.L.O. Eur J Surg Oncol 37(2):148–154PubMedCrossRef Cavaliere F, De Simone M, Virzì S et al (2011) Prognostic factors and oncologic outcome in 146 patients with colorectal peritoneal carcinomatosis treated with cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy: Italian multicenter study S.I.T.I.L.O. Eur J Surg Oncol 37(2):148–154PubMedCrossRef
10.
go back to reference Verwaal VJ (2003) Randomized trial of cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy and palliative surgery in patients with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol 21(20):3737–3743PubMedCrossRef Verwaal VJ (2003) Randomized trial of cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy and palliative surgery in patients with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol 21(20):3737–3743PubMedCrossRef
11.
go back to reference Abdalla EK, Vauthey J-N, Ellis LM, Ellis V, Pollock R, Broglio KR, Hess K, Curley SA (2004) Recurrence and outcomes following hepatic resection, radiofrequency ablation, and combined resection/ablation for colorectal liver metastases. Annals of Surgery 239(6):818–825, discussion 825–7PubMedCrossRef Abdalla EK, Vauthey J-N, Ellis LM, Ellis V, Pollock R, Broglio KR, Hess K, Curley SA (2004) Recurrence and outcomes following hepatic resection, radiofrequency ablation, and combined resection/ablation for colorectal liver metastases. Annals of Surgery 239(6):818–825, discussion 825–7PubMedCrossRef
12.
go back to reference Verwaal VJ, Zoetmulder FAN (2004) Follow-up of patients treated by cytoreduction and chemotherapy for peritoneal carcinomatosis of colorectal origin. Eur J Surg Oncol (EJSO) 30(3):280–285CrossRef Verwaal VJ, Zoetmulder FAN (2004) Follow-up of patients treated by cytoreduction and chemotherapy for peritoneal carcinomatosis of colorectal origin. Eur J Surg Oncol (EJSO) 30(3):280–285CrossRef
13.
14.
go back to reference Nguyen DX, Bos PD, Massagué J (2009) Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer 9(4):274–284PubMedCrossRef Nguyen DX, Bos PD, Massagué J (2009) Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer 9(4):274–284PubMedCrossRef
15.
go back to reference van der Wal JBC, Jeekel J (2007) Biology of the peritoneum in normal homeostasis and after surgical trauma. Colorectal Dis 9(Suppl 2):9–13PubMed van der Wal JBC, Jeekel J (2007) Biology of the peritoneum in normal homeostasis and after surgical trauma. Colorectal Dis 9(Suppl 2):9–13PubMed
16.
go back to reference Yonemura Y, Endou Y, Fujita H, Fushida S, Bandou E, Taniguchi K, Miwa K, Sugiyama K, Sasaki T (2000) Role of MMP-7 in the formation of peritoneal dissemination in gastric cancer. Gastric Cancer 3(2):63–70PubMedCrossRef Yonemura Y, Endou Y, Fujita H, Fushida S, Bandou E, Taniguchi K, Miwa K, Sugiyama K, Sasaki T (2000) Role of MMP-7 in the formation of peritoneal dissemination in gastric cancer. Gastric Cancer 3(2):63–70PubMedCrossRef
17.
go back to reference Yilmaz M, Christofori G (2009) EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev 28(1–2):15–33PubMedCrossRef Yilmaz M, Christofori G (2009) EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev 28(1–2):15–33PubMedCrossRef
18.
go back to reference Paschos KA, Canovas D, Bird NC (2009) The role of cell adhesion molecules in the progression of colorectal cancer and the development of liver metastasis. Cell Signal 21(5):665–674PubMedCrossRef Paschos KA, Canovas D, Bird NC (2009) The role of cell adhesion molecules in the progression of colorectal cancer and the development of liver metastasis. Cell Signal 21(5):665–674PubMedCrossRef
19.
go back to reference Guarino M, Rubino B, Ballabio G (2007) The role of epithelial–mesenchymal transition in cancer pathology. Pathology 39(3):305–318PubMedCrossRef Guarino M, Rubino B, Ballabio G (2007) The role of epithelial–mesenchymal transition in cancer pathology. Pathology 39(3):305–318PubMedCrossRef
20.
go back to reference Terauchi M, Kajiyama H, Yamashita M et al (2007) Possible involvement of TWIST in enhanced peritoneal metastasis of epithelial ovarian carcinoma. Clin Exp Metastasis 24(5):329–339PubMedCrossRef Terauchi M, Kajiyama H, Yamashita M et al (2007) Possible involvement of TWIST in enhanced peritoneal metastasis of epithelial ovarian carcinoma. Clin Exp Metastasis 24(5):329–339PubMedCrossRef
21.
go back to reference Ma PC, Maulik G, Christensen J, Salgia R (2003) c-Met: structure, functions and potential for therapeutic inhibition. Cancer Metastasis Rev 22(4):309–325PubMedCrossRef Ma PC, Maulik G, Christensen J, Salgia R (2003) c-Met: structure, functions and potential for therapeutic inhibition. Cancer Metastasis Rev 22(4):309–325PubMedCrossRef
22.
go back to reference Gentile A, D'Alessandro L, Lazzari L, Martinoglio B, Bertotti A, Mira A, Lanzetti L, Comoglio PM, Medico E (2008) Met-driven invasive growth involves transcriptional regulation of Arhgap12. Oncogene 27(42):5590–5598PubMedCrossRef Gentile A, D'Alessandro L, Lazzari L, Martinoglio B, Bertotti A, Mira A, Lanzetti L, Comoglio PM, Medico E (2008) Met-driven invasive growth involves transcriptional regulation of Arhgap12. Oncogene 27(42):5590–5598PubMedCrossRef
23.
go back to reference Boccaccio C, Comoglio PM (2006) Invasive growth: a MET-driven genetic programme for cancer and stem cells. Nat Rev Cancer 6(8):637–645PubMedCrossRef Boccaccio C, Comoglio PM (2006) Invasive growth: a MET-driven genetic programme for cancer and stem cells. Nat Rev Cancer 6(8):637–645PubMedCrossRef
24.
go back to reference Osada S, Matsui S, Komori S et al (2010) Effect of hepatocyte growth factor on progression of liver metastasis in colorectal cancer. Hepatogastroenterology 57(97):76–80PubMed Osada S, Matsui S, Komori S et al (2010) Effect of hepatocyte growth factor on progression of liver metastasis in colorectal cancer. Hepatogastroenterology 57(97):76–80PubMed
25.
go back to reference Sawada K, Radjabi AR, Shinomiya N et al (2007) c-Met overexpression is a prognostic factor in ovarian cancer and an effective target for inhibition of peritoneal dissemination and invasion. Cancer Res 67(4):1670–1679PubMedCrossRef Sawada K, Radjabi AR, Shinomiya N et al (2007) c-Met overexpression is a prognostic factor in ovarian cancer and an effective target for inhibition of peritoneal dissemination and invasion. Cancer Res 67(4):1670–1679PubMedCrossRef
26.
go back to reference Davies RL, Grosse VA, Kucherlapati R, Bothwell M (1980) Genetic analysis of epidermal growth factor action: assignment of human epidermal growth factor receptor gene to chromosome 7. Proc Natl Acad Sci U S A 77(7):4188–4192PubMedCrossRef Davies RL, Grosse VA, Kucherlapati R, Bothwell M (1980) Genetic analysis of epidermal growth factor action: assignment of human epidermal growth factor receptor gene to chromosome 7. Proc Natl Acad Sci U S A 77(7):4188–4192PubMedCrossRef
27.
go back to reference Cowden Dahl KD, Symowicz J, Ning Y, Gutierrez E, Fishman DA, Adley BP, Stack MS, Hudson LG (2008) Matrix metalloproteinase 9 is a mediator of epidermal growth factor-dependent E-cadherin loss in ovarian carcinoma cells. Cancer Res 68(12):4606–4613PubMedCrossRef Cowden Dahl KD, Symowicz J, Ning Y, Gutierrez E, Fishman DA, Adley BP, Stack MS, Hudson LG (2008) Matrix metalloproteinase 9 is a mediator of epidermal growth factor-dependent E-cadherin loss in ovarian carcinoma cells. Cancer Res 68(12):4606–4613PubMedCrossRef
28.
go back to reference Nicosia SV, Bai W, Cheng JQ, Coppola D, Kruk PA (2003) Oncogenic pathways implicated in ovarian epithelial cancer. Hematol Oncol Clin North Am 17(4):927–943PubMedCrossRef Nicosia SV, Bai W, Cheng JQ, Coppola D, Kruk PA (2003) Oncogenic pathways implicated in ovarian epithelial cancer. Hematol Oncol Clin North Am 17(4):927–943PubMedCrossRef
29.
go back to reference Davidson B, Goldberg I, Gotlieb WH, Kopolovic J, Ben-Baruch G, Nesland JM, Berner A, Bryne M, Reich R (1999) High levels of MMP-2, MMP-9, MT1-MMP and TIMP-2 mRNA correlate with poor survival in ovarian carcinoma. Clin Exp Metastasis 17(10):799–808PubMedCrossRef Davidson B, Goldberg I, Gotlieb WH, Kopolovic J, Ben-Baruch G, Nesland JM, Berner A, Bryne M, Reich R (1999) High levels of MMP-2, MMP-9, MT1-MMP and TIMP-2 mRNA correlate with poor survival in ovarian carcinoma. Clin Exp Metastasis 17(10):799–808PubMedCrossRef
30.
go back to reference Kamat AA, Fletcher M, Gruman LM, Mueller P, Lopez A, Landen CN, Han L, Gershenson DM, Sood AK (2006) The clinical relevance of stromal matrix metalloproteinase expression in ovarian cancer. Clin Cancer Res 12(6):1707–1714PubMedCrossRef Kamat AA, Fletcher M, Gruman LM, Mueller P, Lopez A, Landen CN, Han L, Gershenson DM, Sood AK (2006) The clinical relevance of stromal matrix metalloproteinase expression in ovarian cancer. Clin Cancer Res 12(6):1707–1714PubMedCrossRef
31.
go back to reference Gentile A, Trusolino L, Comoglio PM (2008) The Met tyrosine kinase receptor in development and cancer. Cancer Metastasis Rev 27(1):85–94PubMedCrossRef Gentile A, Trusolino L, Comoglio PM (2008) The Met tyrosine kinase receptor in development and cancer. Cancer Metastasis Rev 27(1):85–94PubMedCrossRef
32.
go back to reference Benvenuti S, Comoglio PM (2007) The MET receptor tyrosine kinase in invasion and metastasis. J Cell Physiol 213(2):316–325PubMedCrossRef Benvenuti S, Comoglio PM (2007) The MET receptor tyrosine kinase in invasion and metastasis. J Cell Physiol 213(2):316–325PubMedCrossRef
33.
go back to reference Holloway SE, Beck AW, Girard L, Jaber MR, Barnett CC Jr, Brekken RA, Fleming JB (2005) Increased expression of Cyr61 (CCN1) identified in peritoneal metastases from human pancreatic cancer. J Am Coll Surg 200(3):371–377PubMedCrossRef Holloway SE, Beck AW, Girard L, Jaber MR, Barnett CC Jr, Brekken RA, Fleming JB (2005) Increased expression of Cyr61 (CCN1) identified in peritoneal metastases from human pancreatic cancer. J Am Coll Surg 200(3):371–377PubMedCrossRef
34.
go back to reference Sawada K, Mitra AK, Radjabi AR et al (2008) Loss of E-cadherin promotes ovarian cancer metastasis via 5-integrin, which is a therapeutic target. Cancer Res 68(7):2329–2339PubMedCrossRef Sawada K, Mitra AK, Radjabi AR et al (2008) Loss of E-cadherin promotes ovarian cancer metastasis via 5-integrin, which is a therapeutic target. Cancer Res 68(7):2329–2339PubMedCrossRef
35.
go back to reference Douma S, Van Laar T, Zevenhoven J, Meuwissen R, Van Garderen E, Peeper DS (2004) Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature 430(7003):1034–1039PubMedCrossRef Douma S, Van Laar T, Zevenhoven J, Meuwissen R, Van Garderen E, Peeper DS (2004) Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature 430(7003):1034–1039PubMedCrossRef
36.
go back to reference Chiarugi P, Giannoni E (2008) Anoikis: a necessary death program for anchorage-dependent cells. Biochem Pharmacol 76(11):1352–1364PubMedCrossRef Chiarugi P, Giannoni E (2008) Anoikis: a necessary death program for anchorage-dependent cells. Biochem Pharmacol 76(11):1352–1364PubMedCrossRef
37.
38.
go back to reference Gimond C, van Der Flier A, van Delft S, Brakebusch C, Kuikman I, Collard JG, Fässler R, Sonnenberg A (1999) Induction of cell scattering by expression of beta1 integrins in beta1-deficient epithelial cells requires activation of members of the rho family of GTPases and downregulation of cadherin and catenin function. J Cell Biol 147(6):1325–1340PubMedCrossRef Gimond C, van Der Flier A, van Delft S, Brakebusch C, Kuikman I, Collard JG, Fässler R, Sonnenberg A (1999) Induction of cell scattering by expression of beta1 integrins in beta1-deficient epithelial cells requires activation of members of the rho family of GTPases and downregulation of cadherin and catenin function. J Cell Biol 147(6):1325–1340PubMedCrossRef
39.
go back to reference Weinberg RA (2006) The biology of cancer. 1st ed. 850. Garland Science, Taylor & Francis Group, LLC. New York, NY USA Weinberg RA (2006) The biology of cancer. 1st ed. 850. Garland Science, Taylor & Francis Group, LLC. New York, NY USA
40.
go back to reference Dong Y, Tan OL, Loessner D, Stephens C, Walpole C, Boyle GM, Parsons PG, Clements JA (2010) Kallikrein-related peptidase 7 promotes multicellular aggregation via the 5 1 integrin pathway and paclitaxel chemoresistance in serous epithelial ovarian carcinoma. Cancer Res 70(7):2624–2633PubMedCrossRef Dong Y, Tan OL, Loessner D, Stephens C, Walpole C, Boyle GM, Parsons PG, Clements JA (2010) Kallikrein-related peptidase 7 promotes multicellular aggregation via the 5 1 integrin pathway and paclitaxel chemoresistance in serous epithelial ovarian carcinoma. Cancer Res 70(7):2624–2633PubMedCrossRef
41.
go back to reference Stamey TA, Yang N, Hay AR, McNeal JE, Freiha FS, Redwine E (1987) Prostate-specific antigen as a serum marker for adenocarcinoma of the prostate. N Engl J Med 317(15):909–916PubMedCrossRef Stamey TA, Yang N, Hay AR, McNeal JE, Freiha FS, Redwine E (1987) Prostate-specific antigen as a serum marker for adenocarcinoma of the prostate. N Engl J Med 317(15):909–916PubMedCrossRef
42.
go back to reference Bhaskar V, Zhang D, Fox M, Seto P, Wong MHL, Wales PE, Powers D, Chao DT, DuBridge RB, Ramakrishnan V (2007) A function blocking anti-mouse integrin alpha5beta1 antibody inhibits angiogenesis and impedes tumor growth in vivo. J Transl Med 5:61PubMedCrossRef Bhaskar V, Zhang D, Fox M, Seto P, Wong MHL, Wales PE, Powers D, Chao DT, DuBridge RB, Ramakrishnan V (2007) A function blocking anti-mouse integrin alpha5beta1 antibody inhibits angiogenesis and impedes tumor growth in vivo. J Transl Med 5:61PubMedCrossRef
43.
go back to reference Ramakrishnan V, Bhaskar V, Law DA et al (2006) Preclinical evaluation of an anti-alpha5beta1 integrin antibody as a novel anti-angiogenic agent. J Exp Ther Oncol 5(4):273–286PubMed Ramakrishnan V, Bhaskar V, Law DA et al (2006) Preclinical evaluation of an anti-alpha5beta1 integrin antibody as a novel anti-angiogenic agent. J Exp Ther Oncol 5(4):273–286PubMed
44.
go back to reference Oosterling SJ, van der Bij GJ, Bögels M, Raa ST, Post JA, Meijer GA, Beelen RHJ, Egmond MV (2008) Anti-β1 integrin antibody reduces surgery-induced adhesion of colon carcinoma cells to traumatized peritoneal surfaces. Ann Surg 247(1):85–94PubMedCrossRef Oosterling SJ, van der Bij GJ, Bögels M, Raa ST, Post JA, Meijer GA, Beelen RHJ, Egmond MV (2008) Anti-β1 integrin antibody reduces surgery-induced adhesion of colon carcinoma cells to traumatized peritoneal surfaces. Ann Surg 247(1):85–94PubMedCrossRef
45.
go back to reference Talieri M, Mathioudaki K, Prezas P, Alexopoulou DK, Diamandis EP, Xynopoulos D, Ardavanis A, Arnogiannaki N, Scorilas A (2009) Clinical significance of kallikrein-related peptidase 7 (KLK7) in colorectal cancer. Thromb Haemost. doi:10.1160/TH08-07-0471 Talieri M, Mathioudaki K, Prezas P, Alexopoulou DK, Diamandis EP, Xynopoulos D, Ardavanis A, Arnogiannaki N, Scorilas A (2009) Clinical significance of kallikrein-related peptidase 7 (KLK7) in colorectal cancer. Thromb Haemost. doi:10.​1160/​TH08-07-0471
46.
go back to reference Talieri M, Li L, Zheng Y, Alexopoulou DK, Soosaipillai A, Scorilas A, Xynopoulos D, Diamandis EP (2009) The use of kallikrein-related peptidases as adjuvant prognostic markers in colorectal cancer. Br J Canc 100(10):1659–1665 Talieri M, Li L, Zheng Y, Alexopoulou DK, Soosaipillai A, Scorilas A, Xynopoulos D, Diamandis EP (2009) The use of kallikrein-related peptidases as adjuvant prognostic markers in colorectal cancer. Br J Canc 100(10):1659–1665
47.
go back to reference Kim LC, Song L, Haura EB (2009) Src kinases as therapeutic targets for cancer. Nat Rev Clin Oncol 6(10):587–595PubMedCrossRef Kim LC, Song L, Haura EB (2009) Src kinases as therapeutic targets for cancer. Nat Rev Clin Oncol 6(10):587–595PubMedCrossRef
48.
go back to reference Hofmann C, Lippert E, Falk W, Schölmerich J, Rogler G, Obermeier F (2009) Primary human colonic epithelial cells are transiently protected from anoikis by a Src-dependent mechanism. Biochem Biophys Res Commun 390(3):908–914PubMedCrossRef Hofmann C, Lippert E, Falk W, Schölmerich J, Rogler G, Obermeier F (2009) Primary human colonic epithelial cells are transiently protected from anoikis by a Src-dependent mechanism. Biochem Biophys Res Commun 390(3):908–914PubMedCrossRef
49.
go back to reference Windham TC, Parikh NU, Siwak DR, Summy JM, McConkey DJ, Kraker AJ, Gallick GE (2002) Src activation regulates anoikis in human colon tumor cell lines. Oncogene 21(51):7797–7807PubMedCrossRef Windham TC, Parikh NU, Siwak DR, Summy JM, McConkey DJ, Kraker AJ, Gallick GE (2002) Src activation regulates anoikis in human colon tumor cell lines. Oncogene 21(51):7797–7807PubMedCrossRef
50.
go back to reference Sakamoto M, Takamura M, Ino Y, Miura A, Genda T, Hirohashi S (2001) Involvement of c-Src in carcinoma cell motility and metastasis. Jpn J Cancer Res 92(9):941–946PubMedCrossRef Sakamoto M, Takamura M, Ino Y, Miura A, Genda T, Hirohashi S (2001) Involvement of c-Src in carcinoma cell motility and metastasis. Jpn J Cancer Res 92(9):941–946PubMedCrossRef
51.
go back to reference Felding-Habermann B (2003) Integrin adhesion receptors in tumor metastasis. Clin Exp Metastasis 20(3):203–213PubMedCrossRef Felding-Habermann B (2003) Integrin adhesion receptors in tumor metastasis. Clin Exp Metastasis 20(3):203–213PubMedCrossRef
53.
go back to reference Guilford P, Hopkins J, Harraway J, McLeod M, McLeod N, Harawira P, Taite H, Scoular R, Miller A, Reeve AE (1998) E-cadherin germline mutations in familial gastric cancer. Nature 392(6674):402–405PubMedCrossRef Guilford P, Hopkins J, Harraway J, McLeod M, McLeod N, Harawira P, Taite H, Scoular R, Miller A, Reeve AE (1998) E-cadherin germline mutations in familial gastric cancer. Nature 392(6674):402–405PubMedCrossRef
54.
go back to reference Birchmeier W, Weidner KM, Hülsken J, Behrens J (1993) Molecular mechanisms leading to cell junction (cadherin) deficiency in invasive carcinomas. Semin Cancer Biol 4(4):231–239PubMed Birchmeier W, Weidner KM, Hülsken J, Behrens J (1993) Molecular mechanisms leading to cell junction (cadherin) deficiency in invasive carcinomas. Semin Cancer Biol 4(4):231–239PubMed
55.
go back to reference Pocard M, Debruyne P, Bras-Gonçalves R, Mareel M, Dutrillaux B, Poupon MF (2001) Single alteration of p53 or E-cadherin genes can alter the surgical resection benefit in an experimental model of colon cancer. Dis Colon Rectum 44(8):1106–1112PubMedCrossRef Pocard M, Debruyne P, Bras-Gonçalves R, Mareel M, Dutrillaux B, Poupon MF (2001) Single alteration of p53 or E-cadherin genes can alter the surgical resection benefit in an experimental model of colon cancer. Dis Colon Rectum 44(8):1106–1112PubMedCrossRef
56.
go back to reference Ziprin P, Ridgway PF, Peck DH, Darzi AW (2003) Laparoscopic enhancement of tumour cell binding to the peritoneum is inhibited by anti-intercellular adhesion molecule-1 monoclonal antibody. Surg Endosc 17(11):1812–1817PubMedCrossRef Ziprin P, Ridgway PF, Peck DH, Darzi AW (2003) Laparoscopic enhancement of tumour cell binding to the peritoneum is inhibited by anti-intercellular adhesion molecule-1 monoclonal antibody. Surg Endosc 17(11):1812–1817PubMedCrossRef
57.
go back to reference Alkhamesi NA, Roberts G, Ziprin P, Peck DH (2007) Induction of proteases in peritoneal carcinomatosis, the role of ICAM-1/CD43 interaction. Biomark Insights 2:377–384PubMed Alkhamesi NA, Roberts G, Ziprin P, Peck DH (2007) Induction of proteases in peritoneal carcinomatosis, the role of ICAM-1/CD43 interaction. Biomark Insights 2:377–384PubMed
58.
go back to reference Balzar M, Winter MJ, de Boer CJ, Not Available SVL (1999) The biology of the 17-1A antigen (Ep-CAM). J Mol Med 77(10):699–712PubMedCrossRef Balzar M, Winter MJ, de Boer CJ, Not Available SVL (1999) The biology of the 17-1A antigen (Ep-CAM). J Mol Med 77(10):699–712PubMedCrossRef
59.
go back to reference Xie X, Wang C-Y, Cao Y-X, Wang W, Zhuang R, Chen L-H, Dang N-N, Fang L, Jin B-Q (2005) Expression pattern of epithelial cell adhesion molecule on normal and malignant colon tissues. WJG 11(3):344–347PubMed Xie X, Wang C-Y, Cao Y-X, Wang W, Zhuang R, Chen L-H, Dang N-N, Fang L, Jin B-Q (2005) Expression pattern of epithelial cell adhesion molecule on normal and malignant colon tissues. WJG 11(3):344–347PubMed
60.
go back to reference Bellone S, Siegel ER, Cocco E, Cargnelutti M, Silasi D-A, Azodi M, Schwartz PE, Rutherford TJ, Pecorelli S, Santin AD (2009) Overexpression of epithelial cell adhesion molecule in primary, metastatic, and recurrent/chemotherapy-resistant epithelial ovarian cancer. Int J Gynecol Canc 19(5):860–866CrossRef Bellone S, Siegel ER, Cocco E, Cargnelutti M, Silasi D-A, Azodi M, Schwartz PE, Rutherford TJ, Pecorelli S, Santin AD (2009) Overexpression of epithelial cell adhesion molecule in primary, metastatic, and recurrent/chemotherapy-resistant epithelial ovarian cancer. Int J Gynecol Canc 19(5):860–866CrossRef
61.
go back to reference Lessan K, Aguiar DJ, Oegema T, Siebenson L, Skubitz AP (1999) CD44 and beta1 integrin mediate ovarian carcinoma cell adhesion to peritoneal mesothelial cells. Am J Pathol 154(5):1525–1537PubMedCrossRef Lessan K, Aguiar DJ, Oegema T, Siebenson L, Skubitz AP (1999) CD44 and beta1 integrin mediate ovarian carcinoma cell adhesion to peritoneal mesothelial cells. Am J Pathol 154(5):1525–1537PubMedCrossRef
62.
63.
go back to reference Mulder JW, Wielenga VJ, Polak MM, van den Berg FM, Adolf GR, Herrlich P, Pals ST, Offerhaus GJ (1995) Expression of mutant p53 protein and CD44 variant proteins in colorectal tumorigenesis. Gut 36(1):76–80PubMedCrossRef Mulder JW, Wielenga VJ, Polak MM, van den Berg FM, Adolf GR, Herrlich P, Pals ST, Offerhaus GJ (1995) Expression of mutant p53 protein and CD44 variant proteins in colorectal tumorigenesis. Gut 36(1):76–80PubMedCrossRef
64.
go back to reference Cannistra SA, Kansas GS, Niloff J, DeFranzo B, Kim Y, Ottensmeier C (1993) Binding of ovarian cancer cells to peritoneal mesothelium in vitro is partly mediated by CD44H. Cancer Res 53(16):3830–3838PubMed Cannistra SA, Kansas GS, Niloff J, DeFranzo B, Kim Y, Ottensmeier C (1993) Binding of ovarian cancer cells to peritoneal mesothelium in vitro is partly mediated by CD44H. Cancer Res 53(16):3830–3838PubMed
65.
go back to reference Kajiyama H, Shibata K, Terauchi M, Ino K, Nawa A, Kikkawa F (2008) Involvement of SDF-1alpha/CXCR4 axis in the enhanced peritoneal metastasis of epithelial ovarian carcinoma. Int J Cancer 122(1):91–99PubMedCrossRef Kajiyama H, Shibata K, Terauchi M, Ino K, Nawa A, Kikkawa F (2008) Involvement of SDF-1alpha/CXCR4 axis in the enhanced peritoneal metastasis of epithelial ovarian carcinoma. Int J Cancer 122(1):91–99PubMedCrossRef
66.
go back to reference Gubbels JAA, Belisle J, Onda M et al (2006) Mesothelin-MUC16 binding is a high affinity, N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors. Mol Cancer 5(1):50PubMedCrossRef Gubbels JAA, Belisle J, Onda M et al (2006) Mesothelin-MUC16 binding is a high affinity, N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors. Mol Cancer 5(1):50PubMedCrossRef
67.
go back to reference Kataoka H, Tanaka H, Nagaike K, Uchiyama S, Itoh H (2003) Role of cancer cell–stroma interaction in invasive growth of cancer cells. Hum Cell 16(1):1–14PubMedCrossRef Kataoka H, Tanaka H, Nagaike K, Uchiyama S, Itoh H (2003) Role of cancer cell–stroma interaction in invasive growth of cancer cells. Hum Cell 16(1):1–14PubMedCrossRef
68.
go back to reference Vihinen P, Kähäri V-M (2002) Matrix metalloproteinases in cancer: prognostic markers and therapeutic targets. Int J Cancer 99(2):157–166PubMedCrossRef Vihinen P, Kähäri V-M (2002) Matrix metalloproteinases in cancer: prognostic markers and therapeutic targets. Int J Cancer 99(2):157–166PubMedCrossRef
69.
go back to reference Yamada T, Oshima T, Yoshihara K et al (2010) Overexpression of MMP-13 gene in colorectal cancer with liver metastasis. Anticancer Res 30(7):2693–2699PubMed Yamada T, Oshima T, Yoshihara K et al (2010) Overexpression of MMP-13 gene in colorectal cancer with liver metastasis. Anticancer Res 30(7):2693–2699PubMed
70.
go back to reference Sc EJCB, D GRP, Sc RGB, Sc TRB, Sc MMB, Sc PZB (2010) Investigation of tumor–peritoneal interactions in the pathogenesis of peritoneal metastases using a novel ex vivo peritoneal model1. J Surg Res 164(2):e265–e272 Sc EJCB, D GRP, Sc RGB, Sc TRB, Sc MMB, Sc PZB (2010) Investigation of tumor–peritoneal interactions in the pathogenesis of peritoneal metastases using a novel ex vivo peritoneal model1. J Surg Res 164(2):e265–e272
71.
go back to reference Peng L, Xing X, Li W, Qu L, Meng L, Lian S, Jiang B, Wu J, Shou C (2009) PRL-3 promotes the motility, invasion, and metastasis of LoVo colon cancer cells through PRL-3-integrin β1-ERK1/2 and-MMP2 signaling. Mol Cancer 8(1):110PubMedCrossRef Peng L, Xing X, Li W, Qu L, Meng L, Lian S, Jiang B, Wu J, Shou C (2009) PRL-3 promotes the motility, invasion, and metastasis of LoVo colon cancer cells through PRL-3-integrin β1-ERK1/2 and-MMP2 signaling. Mol Cancer 8(1):110PubMedCrossRef
72.
go back to reference Aparicio T, Kermorgant S, Dessirier V, Lewin MJ, Lehy T (1999) Matrix metalloproteinase inhibition prevents colon cancer peritoneal carcinomatosis development and prolongs survival in rats. Carcinogenesis 20(8):1445–1451PubMedCrossRef Aparicio T, Kermorgant S, Dessirier V, Lewin MJ, Lehy T (1999) Matrix metalloproteinase inhibition prevents colon cancer peritoneal carcinomatosis development and prolongs survival in rats. Carcinogenesis 20(8):1445–1451PubMedCrossRef
73.
go back to reference Barbolina MV, Adley BP, Shea LD, Stack MS (2008) Wilms tumor gene protein 1 is associated with ovarian cancer metastasis and modulates cell invasion. Cancer 112(7):1632–1641PubMedCrossRef Barbolina MV, Adley BP, Shea LD, Stack MS (2008) Wilms tumor gene protein 1 is associated with ovarian cancer metastasis and modulates cell invasion. Cancer 112(7):1632–1641PubMedCrossRef
74.
go back to reference Varghese S, Burness M, Xu H, Beresnev T, Pingpank J, Alexander HR (2007) Site-specific gene expression profiles and novel molecular prognostic factors in patients with lower gastrointestinal adenocarcinoma diffusely metastatic to liver or peritoneum. Ann Surg Oncol 14(12):3460–3471PubMedCrossRef Varghese S, Burness M, Xu H, Beresnev T, Pingpank J, Alexander HR (2007) Site-specific gene expression profiles and novel molecular prognostic factors in patients with lower gastrointestinal adenocarcinoma diffusely metastatic to liver or peritoneum. Ann Surg Oncol 14(12):3460–3471PubMedCrossRef
75.
go back to reference Hojilla CV, Mohammed FF, Khokha R (2003) Matrix metalloproteinases and their tissue inhibitors direct cell fate during cancer development. Br J Cancer 89(10):1817–1821PubMedCrossRef Hojilla CV, Mohammed FF, Khokha R (2003) Matrix metalloproteinases and their tissue inhibitors direct cell fate during cancer development. Br J Cancer 89(10):1817–1821PubMedCrossRef
76.
go back to reference NCBI (ed) TIMP2 TIMP metallopeptidase inhibitor 2 [Homo sapiens]. Gene, NCBI. NCBI (ed) TIMP2 TIMP metallopeptidase inhibitor 2 [Homo sapiens]. Gene, NCBI.
77.
go back to reference Raa ST, Oosterling SJ, van der Kaaij NP, van den Tol MP, Beelen RHJ, Meijer S, van Eijck CHJ, van der Sijp JRM, van Egmond M, Jeekel J (2005) Surgery promotes implantation of disseminated tumor cells, but does not increase growth of tumor cell clusters. J Surg Oncol 92(2):124–129PubMedCrossRef Raa ST, Oosterling SJ, van der Kaaij NP, van den Tol MP, Beelen RHJ, Meijer S, van Eijck CHJ, van der Sijp JRM, van Egmond M, Jeekel J (2005) Surgery promotes implantation of disseminated tumor cells, but does not increase growth of tumor cell clusters. J Surg Oncol 92(2):124–129PubMedCrossRef
78.
go back to reference van der Bij GJ, Oosterling SJ, Beelen RHJ, Meijer S, Coffey JC, van Egmond M (2009) The perioperative period is an underutilized window of therapeutic opportunity in patients with colorectal cancer. Ann Surg 249(5):727–734PubMedCrossRef van der Bij GJ, Oosterling SJ, Beelen RHJ, Meijer S, Coffey JC, van Egmond M (2009) The perioperative period is an underutilized window of therapeutic opportunity in patients with colorectal cancer. Ann Surg 249(5):727–734PubMedCrossRef
79.
go back to reference Bhowmick NA, Neilson EG, Moses HL (2004) Stromal fibroblasts in cancer initiation and progression. Nature 432(7015):332–337PubMedCrossRef Bhowmick NA, Neilson EG, Moses HL (2004) Stromal fibroblasts in cancer initiation and progression. Nature 432(7015):332–337PubMedCrossRef
80.
go back to reference Kjeldsen T, Andersen AS, Wiberg FC, Rasmussen JS, Schäffer L, Balschmidt P, Møller KB, Møller NP (1991) The ligand specificities of the insulin receptor and the insulin-like growth factor I receptor reside in different regions of a common binding site. Proc Natl Acad Sci U S A 88(10):4404–4408PubMedCrossRef Kjeldsen T, Andersen AS, Wiberg FC, Rasmussen JS, Schäffer L, Balschmidt P, Møller KB, Møller NP (1991) The ligand specificities of the insulin receptor and the insulin-like growth factor I receptor reside in different regions of a common binding site. Proc Natl Acad Sci U S A 88(10):4404–4408PubMedCrossRef
81.
go back to reference Fuchs CS, Goldberg RM, Sargent DJ, Meyerhardt JA, Wolpin BM, Green EM, Pitot HC, Pollak M (2008) Plasma insulin-like growth factors, insulin-like binding protein-3, and outcome in metastatic colorectal cancer: results from Intergroup Trial N9741. Clin Cancer Res 14(24):8263–8269PubMedCrossRef Fuchs CS, Goldberg RM, Sargent DJ, Meyerhardt JA, Wolpin BM, Green EM, Pitot HC, Pollak M (2008) Plasma insulin-like growth factors, insulin-like binding protein-3, and outcome in metastatic colorectal cancer: results from Intergroup Trial N9741. Clin Cancer Res 14(24):8263–8269PubMedCrossRef
82.
go back to reference Goldberg RM (2003) A randomized controlled trial of fluorouracil plus leucovorin, irinotecan, and oxaliplatin combinations in patients with previously untreated metastatic colorectal cancer. J Clin Oncol 22(1):23–30PubMedCrossRef Goldberg RM (2003) A randomized controlled trial of fluorouracil plus leucovorin, irinotecan, and oxaliplatin combinations in patients with previously untreated metastatic colorectal cancer. J Clin Oncol 22(1):23–30PubMedCrossRef
83.
go back to reference Greijer AE, Delis-van Diemen PM, Fijneman RJA, Giles RH, Voest EE, Hinsbergh VWM, Meijer GA (2008) Presence of HIF-1 and related genes in normal mucosa, adenomas and carcinomas of the colorectum. Virchows Arch 452(5):535–544PubMedCrossRef Greijer AE, Delis-van Diemen PM, Fijneman RJA, Giles RH, Voest EE, Hinsbergh VWM, Meijer GA (2008) Presence of HIF-1 and related genes in normal mucosa, adenomas and carcinomas of the colorectum. Virchows Arch 452(5):535–544PubMedCrossRef
84.
go back to reference Nijkamp MW, Hoogwater FJH, Steller EJA, Westendorp BF, van der Meulen TA, Leenders MWH, Rinkes IHMB, Kranenburg O (2010) CD95 is a key mediator of invasion and accelerated outgrowth of mouse colorectal liver metastases following radiofrequency ablation. J Hepatol 53(6):1069–1077PubMedCrossRef Nijkamp MW, Hoogwater FJH, Steller EJA, Westendorp BF, van der Meulen TA, Leenders MWH, Rinkes IHMB, Kranenburg O (2010) CD95 is a key mediator of invasion and accelerated outgrowth of mouse colorectal liver metastases following radiofrequency ablation. J Hepatol 53(6):1069–1077PubMedCrossRef
85.
go back to reference Wu Y, Jin M, Xu H, Shimin Z, He S, Wang L, Zhang Y (2010) Clinicopathologic significance of HIF-1α, CXCR4, and VEGF expression in colon cancer. Clin Dev Immunol 2010:1–10. doi:10.1155/2010/537531 Wu Y, Jin M, Xu H, Shimin Z, He S, Wang L, Zhang Y (2010) Clinicopathologic significance of HIF-1α, CXCR4, and VEGF expression in colon cancer. Clin Dev Immunol 2010:1–10. doi:10.​1155/​2010/​537531
86.
go back to reference Logan-Collins JM, Lowy AM, Robinson-Smith TM, Kumar S, Sussman JJ, James LE, Ahmad SA (2007) VEGF expression predicts survival in patients with peritoneal surface metastases from mucinous adenocarcinoma of the appendix and colon. Ann Surg Oncol 15(3):738–744PubMedCrossRef Logan-Collins JM, Lowy AM, Robinson-Smith TM, Kumar S, Sussman JJ, James LE, Ahmad SA (2007) VEGF expression predicts survival in patients with peritoneal surface metastases from mucinous adenocarcinoma of the appendix and colon. Ann Surg Oncol 15(3):738–744PubMedCrossRef
87.
go back to reference Hurwitz H, Fehrenbacher L, Novotny W et al (2004) Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 350(23):2335–2342PubMedCrossRef Hurwitz H, Fehrenbacher L, Novotny W et al (2004) Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 350(23):2335–2342PubMedCrossRef
88.
go back to reference Shaheen RM, Ahmad SA, Liu W, Reinmuth N, Jung YD, Tseng WW, Drazan KE, Bucana CD, Hicklin DJ, Ellis LM (2001) Inhibited growth of colon cancer carcinomatosis by antibodies to vascular endothelial and epidermal growth factor receptors. Br J Cancer 85(4):584–589PubMedCrossRef Shaheen RM, Ahmad SA, Liu W, Reinmuth N, Jung YD, Tseng WW, Drazan KE, Bucana CD, Hicklin DJ, Ellis LM (2001) Inhibited growth of colon cancer carcinomatosis by antibodies to vascular endothelial and epidermal growth factor receptors. Br J Cancer 85(4):584–589PubMedCrossRef
89.
go back to reference Bruns CJ, Shrader M, Harbison MT, Portera C, Solorzano CC, Jauch K-W, Hicklin DJ, Radinsky R, Ellis LM (2002) Effect of the vascular endothelial growth factor receptor-2 antibody DC101 plus gemcitabine on growth, metastasis and angiogenesis of human pancreatic cancer growing orthotopically in nude mice. Int J Cancer 102(2):101–108PubMedCrossRef Bruns CJ, Shrader M, Harbison MT, Portera C, Solorzano CC, Jauch K-W, Hicklin DJ, Radinsky R, Ellis LM (2002) Effect of the vascular endothelial growth factor receptor-2 antibody DC101 plus gemcitabine on growth, metastasis and angiogenesis of human pancreatic cancer growing orthotopically in nude mice. Int J Cancer 102(2):101–108PubMedCrossRef
90.
go back to reference Bruns CJ, Liu W, Davis DW, Shaheen RM, McConkey DJ, Wilson MR, Bucana CD, Hicklin DJ, Ellis LM (2000) Vascular endothelial growth factor is an in vivo survival factor for tumor endothelium in a murine model of colorectal carcinoma liver metastases. Cancer 89(3):488–499PubMedCrossRef Bruns CJ, Liu W, Davis DW, Shaheen RM, McConkey DJ, Wilson MR, Bucana CD, Hicklin DJ, Ellis LM (2000) Vascular endothelial growth factor is an in vivo survival factor for tumor endothelium in a murine model of colorectal carcinoma liver metastases. Cancer 89(3):488–499PubMedCrossRef
91.
go back to reference Sorensen EW, Gerber SA, Sedlacek AL, Rybalko VY, Chan WM, Lord EM (2009) Omental immune aggregates and tumor metastasis within the peritoneal cavity. Immunol Res. doi:10.1007/s12026-009-8100-2 Sorensen EW, Gerber SA, Sedlacek AL, Rybalko VY, Chan WM, Lord EM (2009) Omental immune aggregates and tumor metastasis within the peritoneal cavity. Immunol Res. doi:10.​1007/​s12026-009-8100-2
92.
go back to reference Gerber SA, Rybalko VY, Bigelow CE, Lugade AA, Foster TH, Frelinger JG, Lord EM (2006) Preferential attachment of peritoneal tumor metastases to omental immune aggregates and possible role of a unique vascular microenvironment in metastatic survival and growth. Am J Pathol 169(5):1739–1752PubMedCrossRef Gerber SA, Rybalko VY, Bigelow CE, Lugade AA, Foster TH, Frelinger JG, Lord EM (2006) Preferential attachment of peritoneal tumor metastases to omental immune aggregates and possible role of a unique vascular microenvironment in metastatic survival and growth. Am J Pathol 169(5):1739–1752PubMedCrossRef
93.
go back to reference Beelen RHJ, Oosterling SJ, van Egmond M, van den Born J, Zareie M (2005) Omental milky spots in peritoneal pathophysiology (spots before your eyes). Perit Dial Int 25(1):30–32PubMed Beelen RHJ, Oosterling SJ, van Egmond M, van den Born J, Zareie M (2005) Omental milky spots in peritoneal pathophysiology (spots before your eyes). Perit Dial Int 25(1):30–32PubMed
94.
95.
go back to reference Krist LF, Koenen H, Calame W, van der Harten JJ, van der Linden JC, Eestermans IL, Meyer S, Beelen RH (1997) Ontogeny of milky spots in the human greater omentum: an immunochemical study. Anat Rec 249(3):399–404PubMedCrossRef Krist LF, Koenen H, Calame W, van der Harten JJ, van der Linden JC, Eestermans IL, Meyer S, Beelen RH (1997) Ontogeny of milky spots in the human greater omentum: an immunochemical study. Anat Rec 249(3):399–404PubMedCrossRef
96.
go back to reference Yildirim A, Aktaş A, Nergiz Y, Akkuş M (2010) Analysis of human omentum-associated lymphoid tissue components with S-100: an immunohistochemical study. Rom J Morphol Embryol 51(4):759–764PubMed Yildirim A, Aktaş A, Nergiz Y, Akkuş M (2010) Analysis of human omentum-associated lymphoid tissue components with S-100: an immunohistochemical study. Rom J Morphol Embryol 51(4):759–764PubMed
97.
go back to reference Yildirim A, Akkus M, Nergiz Y, Yuruker S (2004) Immunohistochemical analysis of CD31, CD36, and CD44 antigens in human omentum. Saudi Med J 25(3):308–312PubMed Yildirim A, Akkus M, Nergiz Y, Yuruker S (2004) Immunohistochemical analysis of CD31, CD36, and CD44 antigens in human omentum. Saudi Med J 25(3):308–312PubMed
98.
go back to reference Babic AM, Kireeva ML, Kolesnikova TV, Lau LF (1998) CYR61, a product of a growth factor-inducible immediate early gene, promotes angiogenesis and tumor growth. Proc Natl Acad Sci U S A 95(11):6355–6360PubMedCrossRef Babic AM, Kireeva ML, Kolesnikova TV, Lau LF (1998) CYR61, a product of a growth factor-inducible immediate early gene, promotes angiogenesis and tumor growth. Proc Natl Acad Sci U S A 95(11):6355–6360PubMedCrossRef
99.
go back to reference Fan Y-F, Huang Z-H (2002) Angiogenesis inhibitor TNP-470 suppresses growth of peritoneal disseminating foci of human colon cancer line Lovo. WJG 8(5):853–856PubMed Fan Y-F, Huang Z-H (2002) Angiogenesis inhibitor TNP-470 suppresses growth of peritoneal disseminating foci of human colon cancer line Lovo. WJG 8(5):853–856PubMed
100.
go back to reference Griffith EC, Su Z, Niwayama S, Ramsay CA, Chang YH, Liu JO (1998) Molecular recognition of angiogenesis inhibitors fumagillin and ovalicin by methionine aminopeptidase 2. Proc Natl Acad Sci U S A 95(26):15183–15188PubMedCrossRef Griffith EC, Su Z, Niwayama S, Ramsay CA, Chang YH, Liu JO (1998) Molecular recognition of angiogenesis inhibitors fumagillin and ovalicin by methionine aminopeptidase 2. Proc Natl Acad Sci U S A 95(26):15183–15188PubMedCrossRef
101.
go back to reference Hines J, Ju R, Dutschman GE, Cheng Y-C, Crews CM (2010) Reversal of TNP-470-induced endothelial cell growth arrest by guanine and guanine nucleosides. J Pharmacol Exp Ther 334(3):729–738PubMedCrossRef Hines J, Ju R, Dutschman GE, Cheng Y-C, Crews CM (2010) Reversal of TNP-470-induced endothelial cell growth arrest by guanine and guanine nucleosides. J Pharmacol Exp Ther 334(3):729–738PubMedCrossRef
102.
go back to reference Yan TD (2006) Systematic review on the efficacy of cytoreductive surgery combined with perioperative intraperitoneal chemotherapy for peritoneal carcinomatosis from colorectal carcinoma. J Clin Oncol 24(24):4011–4019PubMedCrossRef Yan TD (2006) Systematic review on the efficacy of cytoreductive surgery combined with perioperative intraperitoneal chemotherapy for peritoneal carcinomatosis from colorectal carcinoma. J Clin Oncol 24(24):4011–4019PubMedCrossRef
103.
go back to reference van Dam GM, Themelis G, Crane LMA, et al (2011) Intraoperative tumor-specific fluorescence imaging in ovarian cancer by folate receptor-α targeting: first in-human results. Nature Medicine 17:1–6 van Dam GM, Themelis G, Crane LMA, et al (2011) Intraoperative tumor-specific fluorescence imaging in ovarian cancer by folate receptor-α targeting: first in-human results. Nature Medicine 17:1–6
104.
go back to reference Van der Speeten K, Stuart OA, Chang D, Mahteme H, Sugarbaker PH (2011) Changes induced by surgical and clinical factors in the pharmacology of intraperitoneal mitomycin C in 145 patients with peritoneal carcinomatosis. Cancer Chemother Pharmacol 68(1):147–156PubMedCrossRef Van der Speeten K, Stuart OA, Chang D, Mahteme H, Sugarbaker PH (2011) Changes induced by surgical and clinical factors in the pharmacology of intraperitoneal mitomycin C in 145 patients with peritoneal carcinomatosis. Cancer Chemother Pharmacol 68(1):147–156PubMedCrossRef
105.
go back to reference De Roock MD W, De Vriendt MSc V, MD NN, MD PFC, MD PST (2011) KRAS, BRAF, PIK3CA, and PTEN mutations: implications for targeted therapies in metastatic colorectal cancer. Lancet Oncology 12(6):594–603 De Roock MD W, De Vriendt MSc V, MD NN, MD PFC, MD PST (2011) KRAS, BRAF, PIK3CA, and PTEN mutations: implications for targeted therapies in metastatic colorectal cancer. Lancet Oncology 12(6):594–603
106.
go back to reference Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, Beijersbergen RL, Bardelli A, Bernards R (2012) Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 483(7387):100–103PubMedCrossRef Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, Beijersbergen RL, Bardelli A, Bernards R (2012) Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 483(7387):100–103PubMedCrossRef
107.
go back to reference Olmos D, Postel-Vinay S, Molife LR et al (2010) Safety, pharmacokinetics, and preliminary activity of the anti-IGF-1R antibody figitumumab (CP-751,871) in patients with sarcoma and Ewing's sarcoma: a phase 1 expansion cohort study. Lancet Oncol 11(2):129–135PubMedCrossRef Olmos D, Postel-Vinay S, Molife LR et al (2010) Safety, pharmacokinetics, and preliminary activity of the anti-IGF-1R antibody figitumumab (CP-751,871) in patients with sarcoma and Ewing's sarcoma: a phase 1 expansion cohort study. Lancet Oncol 11(2):129–135PubMedCrossRef
108.
go back to reference Haluska P, Worden F, Olmos D, Yin D, Schteingart D, Batzel GN, Paccagnella ML, de Bono JS, Gualberto A, Hammer GD (2010) Safety, tolerability, and pharmacokinetics of the anti-IGF-1R monoclonal antibody figitumumab in patients with refractory adrenocortical carcinoma. Cancer Chemother Pharmacol 65(4):765–773PubMedCrossRef Haluska P, Worden F, Olmos D, Yin D, Schteingart D, Batzel GN, Paccagnella ML, de Bono JS, Gualberto A, Hammer GD (2010) Safety, tolerability, and pharmacokinetics of the anti-IGF-1R monoclonal antibody figitumumab in patients with refractory adrenocortical carcinoma. Cancer Chemother Pharmacol 65(4):765–773PubMedCrossRef
109.
go back to reference Nijkamp MW, van der Bilt JDW, de Bruijn MT, Molenaar IQ, Voest EE, van Diest PJ, Kranenburg O, Borel Rinkes IHM (2009) Accelerated perinecrotic outgrowth of colorectal liver metastases following radiofrequency ablation is a hypoxia-driven phenomenon. Ann Surg 249(5):814–823PubMedCrossRef Nijkamp MW, van der Bilt JDW, de Bruijn MT, Molenaar IQ, Voest EE, van Diest PJ, Kranenburg O, Borel Rinkes IHM (2009) Accelerated perinecrotic outgrowth of colorectal liver metastases following radiofrequency ablation is a hypoxia-driven phenomenon. Ann Surg 249(5):814–823PubMedCrossRef
Metadata
Title
Understanding molecular mechanisms in peritoneal dissemination of colorectal cancer
Future possibilities for personalised treatment by use of biomarkers
Authors
E. M. V. de Cuba
R. Kwakman
M. van Egmond
L. J. W. Bosch
H. J. Bonjer
G. A. Meijer
E. A. te Velde
Publication date
01-09-2012
Publisher
Springer-Verlag
Published in
Virchows Archiv / Issue 3/2012
Print ISSN: 0945-6317
Electronic ISSN: 1432-2307
DOI
https://doi.org/10.1007/s00428-012-1287-y

Other articles of this Issue 3/2012

Virchows Archiv 3/2012 Go to the issue