Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-qs9v7 Total loading time: 0 Render date: 2024-07-11T03:18:57.627Z Has data issue: false hasContentIssue false

12 - Apoptosis, Anoikis, and Senescence

from VARIOUS PROPERTIES OF CANCER CELLS

Published online by Cambridge University Press:  05 June 2012

Wen Liu
Affiliation:
Southern Illinois University School of Medicine, United States
Kounosuke Watabe
Affiliation:
Southern Illinois University School of Medicine, United States
David Lyden
Affiliation:
Weill Cornell Medical College, New York
Danny R. Welch
Affiliation:
Weill Cornell Medical College, New York
Bethan Psaila
Affiliation:
Imperial College of Medicine, London
Get access

Summary

APOPTOSIS: A CRITICAL PLAYER IN TUMOR PROGRESSION

In the early 1970s, pioneering work of Fidler and colleagues demonstrated that tumor metastasis is an extremely inefficient process, and fewer than 0.01 percent of tumor cells shed in the circulation system are able to survive for the following metastatic colonization at distant organs [1]. The majority of tumor cells that depart from the primary cancer mass die by encountering the body's natural defense barriers, which include induction of apoptosis and senescence and immunosurveillance. Even the tumor cells that survive and reach the secondary organ often become dormant or senescent, and their growth is significantly limited owing to the condition of the microenvironment at the organ sites.

Apoptosis is the most common form of programmed cell death in vertebrates and has been extensively studied over the past decade; it is commonly considered as an important mechanism that negatively regulates cancer development. Recent evidence strongly supports the notion that the apoptosis mechanism also serves as a safeguard system that prevents the dissemination of malignant cells and metastasis. Inbal and his group, by using lung carcinoma clones, have shown that the inhibition of the expression of DAPK, a positive mediator of apoptosis, favored the metastatic process [2]. On the other hand, Del Bufalo et al. demonstrated that overexpression of the antiapoptotic oncoprotein, Bcl-2, significantly promoted the metastatic potential of human breast cancer cells [3].

Type
Chapter
Information
Cancer Metastasis
Biologic Basis and Therapeutics
, pp. 131 - 147
Publisher: Cambridge University Press
Print publication year: 2011

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Fidler, IJ, Nicolson, GL (1977) Fate of recirculating B16 melanoma metastatic variant cells in parabiotic syngeneic recipients. J Natl Cancer Inst. 58(6): 1867–72.CrossRefGoogle ScholarPubMed
Inbal, B et al. (1997) DAP kinase links the control of apoptosis to metastasis. Nature. 390(6656): 180–4.CrossRefGoogle Scholar
Del Bufalo, D et al. (1997) Bcl-2 overexpression enhances the metastatic potential of a human breast cancer line. FASEB J. 11(12): 947–53.CrossRefGoogle ScholarPubMed
Glinsky, GV et al. (1997) Apoptosis and metastasis: increased apoptosis resistance of metastatic cancer cells is associated with the profound deficiency of apoptosis execution mechanisms. Cancer Lett. 115(2): 185–93.CrossRefGoogle ScholarPubMed
Kerr, JF et al. (1994) Apoptosis. Its significance in cancer and cancer therapy. Cancer. 73: 2013–26.3.0.CO;2-J>CrossRefGoogle ScholarPubMed
Salvesen, GS, Dixit, VM (1997) Caspases: intracellular signaling by proteolysis. Cell. 91(4): 443–6.CrossRefGoogle ScholarPubMed
Zimmermann, KC, Green, DR (2001) How cells die: apoptosis pathways. J Allergy Clin Immunol. 108: S99-S103.CrossRefGoogle ScholarPubMed
Antonsson, B, Martinou, JC (2000) The Bcl-2 protein family. Exp Cell Res. 256: 50–57.CrossRefGoogle ScholarPubMed
Cheng, EH et al. (2001) BCL-2, BCL-X(L) sequester BH3 domain-only molecules preventing BAX- and BAK- mediated mitochondrial apoptosis. Mol Cell. 8: 705–11.CrossRefGoogle ScholarPubMed
Bergman, PJ et al. (1996) Acquisition of apoptosis resistance with chemoresistance and metastatic potential in human breast cancer: Role of bcl-2 and bcl-X, Proc. Am Asso. Cancer Res. 37: 17.Google Scholar
McConkey, D et al. (1996) Apoptosis resistance increases with metastatic potential in cells of the human LNCaP prostate carcinoma line. Cancer Res. 56: 5594–9.Google ScholarPubMed
Furuya, Y et al. (1996) Expression of bcl-2 and the progression of human and rodent prostatic cancers. Clin Cancer Res. 2: 389–98.Google ScholarPubMed
Takaoka, A et al. (1997) Anti-cell death activity promotes pulmonary metastasis of melanoma cells. Oncogene. 14: 2971–7.CrossRefGoogle ScholarPubMed
Wong, CW et al. (2001) Apoptosis: an early event in metastatic inefficiency. Cancer Res. 61: 333–8.Google ScholarPubMed
Martin, SS, Leder, P (2001) Human MCF10A mammary epithelial cells undergo apoptosis following actin depolymerization that is independent of attachment and rescued by Bcl-2. Mol Cell Biol. 21: 6529–36.CrossRefGoogle ScholarPubMed
Pinkas, J et al. (2004) Bcl-2-mdiated cell survival promotes metastasis of EpH4 βMEKDD mammary epithelial cells. Mol Cancer Res. 2: 551–6.Google ScholarPubMed
Martin, SS et al. (2004) A cytoskeleton-based functional genetic screen identifies Bcl-xL as an enhancer of metastasis, but not primary tumor growth. Oncogene. 23: 4641–5.CrossRefGoogle Scholar
Olopade, OI et al. (1997) Overexpression of BCL-x protein in primary breast cancer is associated with high tumor grade and nodal metastases. Cancer J Sci Am. 3: 230–7.Google ScholarPubMed
Fernandez, Y et al. (2002) Inhibition of apoptosis in human breast cancer cells: role in tumor progression to the metastatic state. Int J Cancer. 101: 317–26.CrossRefGoogle ScholarPubMed
Zhang, WG et al. (2005) Maspin overexpression modulates tumor cell apoptosis through the regulation of Bcl-2 family proteins. BMC Cancer. 5: 50.CrossRefGoogle ScholarPubMed
Cher, ML et al. (2003) Maspin expression inhibits osteolysis, tumor growth, and angiogenesis in a model of prostate cancer bone metastasis. Proc Natl Acad Sci U S A 100(13): 7847–52.CrossRefGoogle Scholar
Olson, M, Kornbluth, S (2001) Mitochondria in apoptosis and human disease. Curr Mol Med. 1(1): 91–122.CrossRefGoogle ScholarPubMed
Vousden, KH, Lu, X (2002) Live or let die: the cell's response to p53. Nat Rev Cancer. 2(8): 594–604.CrossRefGoogle ScholarPubMed
Xu, H, el-Gewely, MR (2001) P53-responsive genes and the potential for cancer diagnostics and therapeutics development. Biotechnol Annu Rev. 7: 131–64.CrossRefGoogle ScholarPubMed
Peller, S, Rotter, V (2003) TP53 in hematological cancer: low incidence of mutations with significant clinical relevance. Hum Mutat. 21(3): 277–84.CrossRefGoogle ScholarPubMed
Kastrinakis, WV et al. (1995) Increased incidence of p53 mutations is associated with hepatic metastasis in colorectal neoplastic progression. Oncogene. 11(4): 647–52.Google ScholarPubMed
Silvestrini, R et al. (1996) Validation of p53 accumulation as a predictor of distant metastasis at 10 years of follow-up in 1400 node-negative breast cancers. Clin Cancer Res. 2(12): 2007–13.Google ScholarPubMed
Pan, W et al. (2003) Prognostic use of growth characteristics of early gastric cancer and expression patterns of apoptotic, cell proliferation, and cell adhesion proteins. J Surg Oncol. 82(2): 104–10.CrossRefGoogle ScholarPubMed
Nikiforov, MA et al. (1996) p53 modulation of anchorage independent growth and experimental metastasis. Oncogene. 13(8): 1709–19.Google ScholarPubMed
Qiu, H et al. (2003) Arrest of B16 melanoma cells in the mouse pulmonary microcirculation induces endothelial nitric oxide synthase-dependent nitric oxide release that is cytotoxic to the tumor cells. Am J Pathol. 162(2): 403–12.CrossRefGoogle ScholarPubMed
Wang, HH et al. (2002) Regulation of B16F1 melanoma cell metastasis by inducible functions of the hepatic microvasculature. Eur J Cancer. 38(9): 1261–70.CrossRefGoogle ScholarPubMed
Wang, HH et al. (2000) B16 melanoma cell arrest in the mouse liver induces nitric oxide release and sinusoidal cytotoxicity: a natural hepatic defense against metastasis. Cancer Res. 60(20): 5862–9.Google ScholarPubMed
Lala, PK, Chakraborty, C (2001) Role of nitric oxide in carcinogenesis and tumour progression. Lancet Oncol. 2(3): 149–56.CrossRefGoogle ScholarPubMed
Jadeski, LC et al. (2002) Role of nitric oxide in tumour progression with special reference to a murine breast cancer model. Can J Physiol Pharmacol 80(2): 125–35.CrossRefGoogle ScholarPubMed
Lala, PK, Orucevic, A (1998) Role of nitric oxide in tumor progression: lessons from experimental tumors. Cancer Metastasis Rev. 17(1): 91–106.CrossRefGoogle ScholarPubMed
Holcik, M et al. (2001) XIAP: apoptotic brake and promising therapeutic target. Apoptosis. 6(4): 253–61.CrossRefGoogle ScholarPubMed
Jäättelä, M (1999) Escaping cell death: survival proteins in cancer. Exp Cell Res. 248(1): 30–43.CrossRefGoogle Scholar
Yoshida, H et al. (2001) Survivin, Bcl-2 and matrix metalloproteinase-2 enhance progression of clear cell- and serous-type ovarian carcinomas. Int J Oncol. 19: 537–42.Google ScholarPubMed
Kato, J et al. (2001) Expression of survivin in esophageal cancer: correlation with the prognosis and response to chemotherapy. Int J Cancer. 95: 92–5.3.0.CO;2-9>CrossRefGoogle ScholarPubMed
Ueda, M et al. (2002) Survivin gene expression in endometriosis. J Clin Endocrinol Met. 87: 3452–9.CrossRefGoogle ScholarPubMed
Berezovskaya, O et al. (2005) Increased expression of apoptosis inhibitor protein XIAP contributes to anoikis resistance of circulating human prostate cancer metastasis precursor cells. Cancer Res. 65(6): 2378–86.CrossRefGoogle ScholarPubMed
Quhtit, A et al. (2007) Survivin is not only a death encounter but also a survival protein for invading tumor cells. Front Biosci. 12: 1260–70.Google Scholar
Tran, J et al. (1999) Marked induction of the IAP family antiapoptotic proteins survivin and XIAP by VEGF in vascular endothelial cells. Biochem Biophys ResCommun. 264(3): 781–8.Google Scholar
Tran, J et al. (2002) The role for survivin in chemoresistance of endothelial cells mediated by VEGF. Proc Natl Acad Sci. USA. 99: 4349–54.CrossRefGoogle ScholarPubMed
Downward, J (2003) Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 3(1): 11–22.CrossRefGoogle ScholarPubMed
Wolfman, JC et al. (2002) Cellular N-Ras promotes cell survival by downregulation of Jun N-terminal protein kinase and p38. Mol Cell Biol. 22(5): 1589–606.CrossRefGoogle ScholarPubMed
Varghese, HJ (2002) Activated ras regulates the proliferation/apoptosis balance and early survival of developing micrometastases. Cancer Res. 62(3): 887–91.Google ScholarPubMed
Liao, Y et al. (2000) Modulation of apoptosis, tumorigenesity and metastatic potential with antisense H-ras oligodeoxynucleotides in a high metastatic tumor model of hepatoma: LCI-D20. Hepatogastroenterology. 47(32): 365–70.Google Scholar
Takeuchi, M et al. (2000) The dominant negative H-ras mutant, N116Y, suppresses growth of metastatic human pancreatic cancer cells in the liver of nude mice. Gene Ther. 7(6): 518–26.CrossRefGoogle ScholarPubMed
Akhurst, RJ, Derynck, R (2001) TGF-beta signaling in cancer – a double-edged sword. Trends Cell Biol. 11(11): S44–S51.Google Scholar
Ellenrieder, V et al. (2002) TGFbeta-regulated transcriptional mechanisms in cancer. Int J Gastrointest Cancer. 31(1–3): 61–9.CrossRefGoogle Scholar
Janda, E et al. (2002) Ras and TGFbeta cooperatively regulate epithelial cell plasticity and metastasis: dissection of Ras signaling pathways. J Cell Biol. 156(2): 299–313.CrossRefGoogle ScholarPubMed
Luzzi, KJ et al. (1998) Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am J Pathol. 153(3): 865–73.CrossRefGoogle ScholarPubMed
Cameron, MD et al. (2000) Temporal progression of metastasis in lung: cell survival, dormancy, and location dependence of metastatic inefficiency. Cancer Res. 60(9): 2541–6.Google ScholarPubMed
Naumov, GN et al. (2002) Persistence of solitary mammary carcinoma cells in a secondary site: a possible contributor to dormancy. Cancer Res. 62(7): 2162–8.Google Scholar
Al-Mehdi, AB et al. (2000) Intravascular origin of metastasis from the proliferation of endothelium-attached tumor cells: a new model for metastasis. Nat Med. 6(1): 100–2.CrossRefGoogle ScholarPubMed
Orr, FW, Wang, HH (2001) Tumor cell interactions with the microvasculature: a rate-limiting step in metastasis. Surg Oncol Clin North Am. 10(2): 357–81.Google Scholar
Holmgren, L et al. (1995) Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med. 1(2): 149–53.CrossRefGoogle ScholarPubMed
O'Reilly, MS et al. (1996) Angiostatin induces and sustains dormancy of human primary tumors in mice. Nat Med. 2(6): 689–92.CrossRefGoogle ScholarPubMed
O'Reilly, MS et al. (1997) Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell. 88(2): 277–85.CrossRefGoogle ScholarPubMed
Kim, HR et al. (1999) Cell cycle arrest and inhibition of anoikis by galectin-3 in human breast epithelial cells. Cancer Res. 59: 4148–54.Google ScholarPubMed
Frisch, SM et al. (1994) Disruption of epithelial cell matrix interactions induces apoptosis. J Cell Biol. 124: 619–26.CrossRefGoogle ScholarPubMed
Frisch, SM et al. (2001) Anoikis mechanisms. Curr Opin Cell Biol. 13: 555–62.CrossRefGoogle ScholarPubMed
Thompson, EW et al. (2005) Invasion and metastasis: a role for epithelial-mesenchymal transition? Cancer Res. 65: 5991–5.CrossRefGoogle ScholarPubMed
Nieto, MA et al. (2002) The snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol. 3: 155–66.CrossRefGoogle ScholarPubMed
Cavallaro, U et al. (2004) Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer. 4: 118–32.CrossRefGoogle Scholar
Helbig, G et al. (2003) NF-κB promotes breast cancer cell migration and metastasis by inducing the expression of the chemokine receptor CXCR4. J Biol Chem. 278: 21631–8.CrossRefGoogle ScholarPubMed
Huang, S et al. (2001) Blockade of NF-κB activity in human prostate cancer cells is associated with suppression of angiogenesis, invasion, and metastasis. Oncogene. 20: 4188–97.CrossRefGoogle Scholar
Huang, EJ, Reichardt, LF (2003) Trk receptors: roles in neuronal signal transduction. Annu Rev Biochem. 72: 609–42.CrossRefGoogle ScholarPubMed
Brodeur, GM (2003) Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer. 3: 203–216.CrossRefGoogle ScholarPubMed
Matsumoto, K et al. (1995) Expression of brain-derived neurotrophic factor and p145TrkB affects survival, differentiation and invasiveness of human neuroblastoma cells. Cancer Res. 55: 1798–1806.Google ScholarPubMed
Ho, R et al. (2002) Resistance to chemotherapy mediated by TrkB in neuroblastomas. Cancer Res. 62: 6462–6.Google ScholarPubMed
Jaboin, J et al. (2002) Brain-derived neurotrophic factor activation of TrkB protects neuroblastoma cells from chemotherapy-induced apoptosis via phosphatidylinositol 3′-kinase pathway. Cancer Res. 62: 6756–63.Google ScholarPubMed
Li, Z et al. (2005) Genetic and pharmacologic identification of Akt as a mediator of brain-derived neurotrophic factor/TrkB rescue of neuroblastoma cells from chemotherapy-induced cell death. Cancer Res. 65: 2070–5.CrossRefGoogle ScholarPubMed
Schulte, JH. et al. (2005) Microarray analysis reveals differential gene expression patterns and regulation of single target genes contributing to the opposing phenotype of TrkA and TrkB-expressing neuroblastomas. Oncogene 24: 165–77.CrossRefGoogle ScholarPubMed
Aoyama, M et al. (2001) Human neuroblastomas with unfavorable biologies express high levels of brain-derived neurotrophic factor mRNA and a variety of its variants. Cancer Lett. 164: 51–60.CrossRefGoogle Scholar
Douma, S et al. (2004) Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature. 430: 1034–9.CrossRefGoogle ScholarPubMed
Geiger, TR, Peeper, DS (2005) The neurotrophic receptor TrkB in anoikis resistance and metastasis: a perspective. Cancer Res. 65: 7033–6.CrossRefGoogle ScholarPubMed
Desmet, CJ, Peeper, DS (2005) The neurotrophic receptor TrkB: a drug target in anti-cancer therapy? Cell Mol Life Sci. 63: 755–9.CrossRefGoogle Scholar
Takenaka, Y et al. (2004) Galectin-2 and metastasis. Glycoconjugate J. 19: 543–9.CrossRefGoogle Scholar
Iurisci, I et al. (2000) Concentrations of galectin-3 in the sera of normal controls and cancer patients. ClinCancer Res. 6: 1389–93.Google Scholar
Nangia-Makker, P. et al. (2000) Galectin-3 induces endothelial cell morphogenesis and angiogenesis. Am J Pathol. 156: 899–909.CrossRefGoogle ScholarPubMed
Matarrese, P et al. (2000) Galectin-3 overexpression protects from apoptosis by improving cell adhesion properties. Int J Cancer. 85: 545–54.3.0.CO;2-N>CrossRefGoogle ScholarPubMed
Warfield, PR et al. (1997) Adhesion of human breast carcinoma to extracellular matrix proteins is modulated by galectin-3. Invasion Metastasis. 17: 101–12.Google ScholarPubMed
Thompson, SC et al. (1974) The colony forming efficiency of single cells and cell aggregates from a spontaneous mouse mammary tumour using the lung colony assay. Br J Cancer. 30: 332–6.CrossRefGoogle ScholarPubMed
Raz, A et al. (1980) Distribution of membrane anionic sites on B16 melanoma variants with differing lung colonising potential. Nature. 284: 363–4.CrossRefGoogle ScholarPubMed
Inohara, H et al. (1996) Interactions between galectin-3 and Mac-2-binding protein mediate cell-cell adhesion. Cancer Res. 56: 4530–4.Google ScholarPubMed
Platt, D, Raz, A (1992) Modulation of the lung colonization of B16-F1melanoma cells by citrus pectin. J Natl Cancer Inst. 84: 438–42.CrossRefGoogle ScholarPubMed
Glinsky, VV et al. (2000) Effects of Thomsen-Friedenreich antigen-specific peptide P-30 on beta-galactoside-mediated homotypic aggregation and adhesion to the endothelium of MDA-MB-435 human breast carcinoma cells. Cancer Res. 15(60): 2584–8.Google Scholar
Lotan, R et al. (1994) Expression of galectins on microvessel endothelial cells and their involvement in tumour cell adhesion. Glycoconjugate J. 11: 462–8.CrossRefGoogle ScholarPubMed
Glinsky, VV et al. (2001) The role of Thomsen-Friedenreich antigen in adhesion of human breast and prostate cancer cells to the endothelium. Cancer Res. 61: 4851–7.Google ScholarPubMed
Palade, GE et al. (1953) Fine structure of blood capillaries. J Appl Physiol. 24: 1424–36.Google Scholar
Williams, TM, Lisanti, MP (2005) Caveolin-1 in oncogenic transformation, cancer, and metastasis. Am J Physiol Cell Physiol. 288: C494-C506.CrossRefGoogle ScholarPubMed
Li, L et al. (2003) Caveolin-1 maintains activated Akt in prostate cancer cells through scaffolding domain binding site interactions with and inhibition of serine/threonine protein phosphatases PP1 and PP2A. Mol Cell Biol. 23: 9389–404.CrossRefGoogle ScholarPubMed
Baserga, R et al. (2003) The IGF-1 receptor in cancer biology. Int J Cancer 107: 873–7.CrossRefGoogle ScholarPubMed
Lu, Z et al. (2003) Downregulation of caveolin-1 function by EGF leads to the loss of E-cadherin, increased transcriptional activity of beta-catenin, and enhanced tumor cell invasion. Cancer Cell. 4: 499–515.CrossRefGoogle ScholarPubMed
Han, JH, Sun, PQ (2007) The pathways to tumor suppression via route p38. Trends Biochem Sci. 32(8): 364–71.CrossRefGoogle ScholarPubMed
Taylor, J et al. (2008) Using metastasis suppressor proteins to dissect interactions among cancer cells and their microenvironment. Cancer Metastasis Rev. 27(1): 67–73.CrossRefGoogle ScholarPubMed
Funasaka, T et al. (2007) Down-regulation of phosphoglucose isomerase/autocrine motility factor expression sensitizes human fibrosarcoma cells to oxidative stress leading to cellular senescence. J Biol Chem. 282(50): 36362–9.CrossRefGoogle ScholarPubMed
Bandyopadhyay, S (2006) Interaction of KAI1 on tumor cells with DARC on vascular endothelium leads to metastasis suppression. Nat Med. 8: 933–8.CrossRefGoogle Scholar
Bissell, MJ, Radisky, D (2001) Putting tumours in context. Nat Rev Cancer 1: 46–54.CrossRefGoogle ScholarPubMed
Mueller, MM, Fusenig, NE (2004) Friends or foes – bipolar effects of the tumour stroma in cancer. Nat Rev Cancer. 4: 839–49.CrossRefGoogle ScholarPubMed
Sappino, AP et al. (1988) Smooth-muscle differentiation in stromal cells of malignant and nonmalignant breast tissues. Int J Cancer. 41: 707–12.CrossRefGoogle Scholar
Olumi, AF et al. (1999) Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res. 59: 5002–11.Google ScholarPubMed
Allinen, M et al. (2004) Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell. 6: 17–32.CrossRefGoogle ScholarPubMed
Orimo, A, Weinberg, RA (2007) Heterogeneity of stromal fibroblasts in tumors. Cancer Biol Ther. 6(4): 618–9.CrossRef
Orimo, A, Weinberg, RA (2006) Stromal fibroblasts in cancer: a novel tumor-promoting cell type. Cell Cycle. 5(15): 1597–601.CrossRefGoogle ScholarPubMed
Grum-Schwensen, B et al. (2005) Suppression of tumor development and metastasis formation in mice lacking the S100A4(mts1) gene. Cancer Res. 65: 3772–3780.CrossRefGoogle ScholarPubMed
Hwang, RF et al. (2008) Cancer-associated stromal fibroblasts promote pancreatic tumor progression. Cancer Res. 68(3): 918–26.CrossRefGoogle ScholarPubMed
Sato, N et al. (2004) Gene expression profiling of tumor-stromal interactions between pancreatic cancer cells and stromal fibroblasts. Cancer Res. 64(19): 6950–6.CrossRefGoogle ScholarPubMed
Yang, AD et al. (2006) Vascular endothelial growth factor receptor-1 activation mediates epithelial to mesenchymal transition in human pancreatic carcinoma cells. Cancer Res. 66: 46–51.CrossRefGoogle ScholarPubMed
Hiratsuka, S et al. (2002) MMP9 induction by vascular endothelial growth factor receptor- 1 is involved in lung-specific metastasis. Cancer Cell. 2: 289–300.CrossRefGoogle ScholarPubMed
Kaplan, R et al. (2006) Preparing the “soil”: the premetastatic niche. Cancer Res. 66(23): 11089–93.CrossRefGoogle ScholarPubMed
Bussard, KM et al. (2007) The bone microenvironment in metastasis; what is special about bone? Cancer Metastasis Rev. 27(1): 41–55.CrossRefGoogle Scholar
Lu, X, Kang, YB (2007) Organotropism of breast cancer metastasis. J. Mammary Gland Biol Neoplasia. 12:153–162.CrossRefGoogle ScholarPubMed
Bidard, FC et al. (2008) A “class action” against the microenvironment: do cancer cells cooperate in metastasis?Cancer Metastasis Rev. 27(1): 5–10.CrossRefGoogle Scholar
Miller-Jensen, K et al. (2007) Common effector processing mediates cell-specific response to stimuli. Nature. 448: 604–8.CrossRefGoogle Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×