Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-m8qmq Total loading time: 0 Render date: 2024-04-19T16:21:51.724Z Has data issue: false hasContentIssue false

14 - Role of Inflammation in Metastatic Progression

from STROMAL CELLS/EXTRACELLULAR MATRIX

Published online by Cambridge University Press:  05 June 2012

Sunhwa Kim
Affiliation:
University of Pittsburgh School of Medicine, United States
Michael Karin
Affiliation:
University of California, San Diego, 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

EXTRINSIC VERSUS INTRINSIC MEDIATORS OF INFLAMMATION

A link between chronic inflammation and cancer has been suspected since the nineteenth century, when Rudolf Virchow first noted that malignant tumors arise at regions of chronic inflammation and contain inflammatory infiltrates [1–5]. However, the sources of inflammation in tumors that are not associated with a chronic inflammation remain incompletely understood. Recently, it became apparent that inflammation can be evoked not only by extrinsic mediators but also by intrinsic mediators (endogenous molecules) (Figure 14.1). For instance, it has been established that necrotic cell death results in the release of molecules normally stored within cells, such as high-mobility group box 1 (HMGB1) and interleukin (IL)-1α, that act as potent inflammatory mediators [6, 7]. Such molecules may responsible for the triggering of tumor-associated inflammation [7].

Toll-like receptors (TLRs), the mammalian homologs of the Drosophila Toll protein, play a crucial role in activation of inflammatory responses and innate host defenses against invading microorganisms by their ability to recognize conserved molecular motifs of microbial origin, also known as pathogen-associated molecular patterns (PAMs) [8–10]. A number of intrinsic mediators have been shown to be capable of engaging TLR family members and other innate immune receptors and thereby trigger the activation of myeloid and lymphoid cells as well as stimulate the maturation of dendritic cells (DCs) [6, 11–13].

The first mammalian TLR to be identified, TLR4, is the receptor for lipopolysaccharide (LPS), a major cell wall component of Gram-negative bacteria [10].

Type
Chapter
Information
Cancer Metastasis
Biologic Basis and Therapeutics
, pp. 155 - 166
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

Balkwill, F, Coussens, LM (2004). Cancer: an inflammatory link. Nature 431 (7007), 405.CrossRefGoogle Scholar
Balkwill, F, Mantovani, A (2001). Inflammation and cancer: back to Virchow? Lancet 357 (9255), 539.CrossRefGoogle ScholarPubMed
Coussens, LM, Werb, Z (2002). Inflammation and cancer. Nature 420 (6917), 860.CrossRefGoogle ScholarPubMed
Karin, M (2005). Inflammation and cancer: the long reach of Ras. Nat Med 11 (1), 20.CrossRefGoogle Scholar
Karin, M (2006). Nuclear factor-kappaB in cancer development and progression. Nature 441 (7092), 431.CrossRefGoogle ScholarPubMed
Park, JS, Svetkauskaite, D et al. (2004). Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. J Biol Chem 279 (9), 7370.CrossRefGoogle ScholarPubMed
Sakurai, T, He, G et al. (2008). Hepatocyte necrosis induced by oxidative stress and IL-1 alpha release mediate carcinogen-induced compensatory proliferation and liver tumorigenesis. Cancer Cell 14 (2), 156.CrossRefGoogle ScholarPubMed
Medzhitov, R, Preston-Hurlburt, P et al. (1997). A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature 388 (6640), 394.CrossRefGoogle ScholarPubMed
Takeda, K, Kaisho, T et al. (2003). Toll-like receptors. Annu Rev Immunol 21, 335.CrossRefGoogle ScholarPubMed
Takeda, K, Akira, S (2007). Toll-like receptors. Curr Protoc Immunol Chapter 14, Unit 14 12.Google ScholarPubMed
Asea, A, Rehli, M et al. (2002). Novel signal transduction pathway utilized by extracellular HSP70: role of toll-like receptor (TLR) 2 and TLR4. J Biol Chem 277 (17), 15028.CrossRefGoogle ScholarPubMed
Kariko, K, Ni, H et al. (2004). mRNA is an endogenous ligand for Toll-like receptor 3. J Biol Chem 279 (13), 12542.CrossRefGoogle ScholarPubMed
Ohashi, K, Burkart, V et al. (2000). Cutting edge: heat shock protein 60 is a putative endogenous ligand of the toll-like receptor-4 complex. J Immunol 164 (2), 558.CrossRefGoogle ScholarPubMed
Takeuchi, O, Sato, S et al. (2002). Cutting edge: role of Toll-like receptor 1 in mediating immune response to microbial lipoproteins. J Immunol 169 (1), 10.CrossRefGoogle ScholarPubMed
Takeuchi, O, Hoshino, K et al. (1999). Differential roles of TLR2 and TLR4 in recognition of gram-negative and gram-positive bacterial cell wall components. Immunity 11 (4), 443.CrossRefGoogle ScholarPubMed
Alexopoulou, L, Holt, AC et al. (2001). Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature 413 (6857), 732.CrossRefGoogle ScholarPubMed
Hayashi, F, Smith, KD et al. (2001). The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature 410 (6832), 1099.CrossRefGoogle ScholarPubMed
Takeuchi, O, Kawai, T et al. (2001). Discrimination of bacterial lipoproteins by Toll-like receptor 6. Int Immunol 13 (7), 933.CrossRefGoogle ScholarPubMed
Lund, JM, Alexopoulou, L et al. (2004). Recognition of single-stranded RNA viruses by Toll-like receptor 7. Proc Natl Acad Sci U S A 101 (15), 5598.CrossRefGoogle ScholarPubMed
Hemmi, H, Takeuchi, O et al. (2000). A Toll-like receptor recognizes bacterial DNA. Nature 408 (6813), 740.CrossRefGoogle ScholarPubMed
Akira, S, Takeda, K (2004). Toll-like receptor signalling. Nat Rev Immunol 4 (7), 499.CrossRefGoogle ScholarPubMed
Kopp, E, Medzhitov, R (2003). Recognition of microbial infection by Toll-like receptors. Curr Opin Immunol 15 (4), 396.CrossRefGoogle ScholarPubMed
Tsan, MF, Gao, B (2004). Cytokine function of heat shock proteins. Am J Physiol Cell Physiol 286 (4), C739.CrossRefGoogle ScholarPubMed
Wallin, RP, Lundqvist, A et al. (2002). Heat-shock proteins as activators of the innate immune system. Trends Immunol 23 (3), 130.CrossRefGoogle ScholarPubMed
Zhao, Y, Yokota, K et al. (2007). Helicobacter pylori heat-shock protein 60 induces interleukin-8 via a Toll-like receptor (TLR)2 and mitogen-activated protein (MAP) kinase pathway in human monocytes. J Med Microbiol 56 (Pt 2), 154.CrossRefGoogle Scholar
Smiley, ST, King, JA et al. (2001). Fibrinogen stimulates macrophage chemokine secretion through toll-like receptor 4. J Immunol 167 (5), 2887.CrossRefGoogle ScholarPubMed
Guillot, L, Balloy, V et al. (2002). Cutting edge: the immunostimulatory activity of the lung surfactant protein-A involves Toll-like receptor 4. J Immunol 168 (12), 5989.CrossRefGoogle ScholarPubMed
Okamura, Y, Watari, M et al. (2001). The extra domain A of fibronectin activates Toll-like receptor 4. J Biol Chem 276 (13), 10229.CrossRefGoogle ScholarPubMed
Saito, S, Yamaji, N et al. (1999). The fibronectin extra domain A activates matrix metalloproteinase gene expression by an interleukin-1-dependent mechanism. J Biol Chem 274 (43), 30756.CrossRefGoogle ScholarPubMed
Johnson, GB, Brunn, GJ et al. (2002). Receptor-mediated monitoring of tissue well-being via detection of soluble heparan sulfate by Toll-like receptor 4. J Immunol 168 (10), 5233.CrossRefGoogle ScholarPubMed
Termeer, C, Benedix, F et al. (2002). Oligosaccharides of hyaluronan activate dendritic cells via toll-like receptor 4. J Exp Med 195 (1), 99.CrossRefGoogle ScholarPubMed
Biragyn, A, Ruffini, PA et al. (2002). Toll-like receptor 4-dependent activation of dendritic cells by beta-defensin 2. Science 298 (5595), 1025.CrossRefGoogle ScholarPubMed
Wakasugi, K, Schimmel, P (1999). Two distinct cytokines released from a human aminoacyl-tRNA synthetase. Science 284 (5411), 147.CrossRefGoogle ScholarPubMed
Kim, S, Takahashi, H et al. (2009). Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature 457 (7225), 102.CrossRefGoogle ScholarPubMed
Wight, TN (2002). Versican: a versatile extracellular matrix proteoglycan in cell biology. Curr Opin Cell Biol 14 (5), 617.CrossRefGoogle ScholarPubMed
Campana, L, Bosurgi, L et al. (2008). HMGB1: a two-headed signal regulating tumor progression and immunity. Curr Opin Immunol 20 (5), 518.CrossRefGoogle ScholarPubMed
Hanahan, D, Weinberg, RA (2000). The hallmarks of cancer. Cell 100 (1), 57.CrossRefGoogle ScholarPubMed
Hussain, SP, Hofseth, LJ et al. (2003). Radical causes of cancer. Nat Rev Cancer 3 (4), 276.CrossRefGoogle Scholar
Jaiswal, M, LaRusso, NF et al. (2000). Inflammatory cytokines induce DNA damage and inhibit DNA repair in cholangiocarcinoma cells by a nitric oxide-dependent mechanism. Cancer Res 60 (1), 184.Google ScholarPubMed
Jaiswal, M, LaRusso, NF et al. (2001). Nitric oxide in gastrointestinal epithelial cell carcinogenesis: linking inflammation to oncogenesis. Am J Physiol Gastrointest Liver Physiol 281 (3), G626.CrossRefGoogle ScholarPubMed
Greten, FR, Karin, M (2004). The IKK/NF-kappaB activation pathway – a target for prevention and treatment of cancer. Cancer Lett 206 (2), 193.CrossRefGoogle ScholarPubMed
Meira, LB, Bugni, JM et al. (2008). DNA damage induced by chronic inflammation contributes to colon carcinogenesis in mice. J Clin Invest 118 (7), 2516.Google ScholarPubMed
Arnott, CH, Scott, KA et al. (2002). Tumour necrosis factor-alpha mediates tumour promotion via a PKC alpha- and AP-1-dependent pathway. Oncogene 21 (31), 4728.CrossRefGoogle Scholar
Balkwill, F (2002). Tumor necrosis factor or tumor promoting factor? Cytokine Growth Factor Rev 13 (2), 135.CrossRefGoogle ScholarPubMed
Arnott, CH, Scott, KA et al. (2004). Expression of both TNF-alpha receptor subtypes is essential for optimal skin tumour development. Oncogene 23 (10), 1902.CrossRefGoogle ScholarPubMed
Knight, B, Yeoh, GC et al. (2000). Impaired preneoplastic changes and liver tumor formation in tumor necrosis factor receptor type 1 knockout mice. J Exp Med 192 (12), 1809.CrossRefGoogle ScholarPubMed
Pikarsky, E, Porat, RM et al. (2004). NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature 431 (7007), 461.CrossRefGoogle ScholarPubMed
Greten, FR, Eckmann, L et al. (2004). IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 118 (3), 285.CrossRefGoogle Scholar
Grivennikov, S, Karin, M (2008). Autocrine IL-6 signaling: a key event in tumorigenesis? Cancer Cell 13 (1), 7.CrossRefGoogle ScholarPubMed
Lin, EY, Nguyen, AV et al. (2001). Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy. J Exp Med 193 (6), 727.CrossRefGoogle ScholarPubMed
Balkwill, F, Charles, KA et al. (2005). Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell 7 (3), 211.CrossRefGoogle ScholarPubMed
Bui, JD, Schreiber, RD (2007). Cancer immunosurveillance, immunoediting and inflammation: independent or interdependent processes? Curr Opin Immunol 19 (2), 203.CrossRefGoogle ScholarPubMed
Karin, M, Cao, Y et al. (2002). NF-kappaB in cancer: from innocent bystander to major culprit. Nat Rev Cancer 2 (4), 301.CrossRefGoogle ScholarPubMed
Old, LJ (1988). Tumor necrosis factor. Sci Am 258 (5), 59.CrossRefGoogle ScholarPubMed
Luo, JL, Maeda, S et al. (2004). Inhibition of NF-kappaB in cancer cells converts inflammation-induced tumor growth mediated by TNFalpha to TRAIL-mediated tumor regression. Cancer Cell 6 (3), 297.CrossRefGoogle ScholarPubMed
Hofseth, LJ (2008). Nitric oxide as a target of complementary and alternative medicines to prevent and treat inflammation and cancer. Cancer Lett 268 (1), 10.CrossRefGoogle ScholarPubMed
Sawa, T, Ohshima, H (2006). Nitrative DNA damage in inflammation and its possible role in carcinogenesis. Nitric Oxide 14 (2), 91.CrossRefGoogle ScholarPubMed
Gordon, S (2003). Alternative activation of macrophages. Nat Rev Immunol 3 (1), 23.CrossRefGoogle ScholarPubMed
Mantovani, A, Sica, A et al. (2005). Macrophage polarization comes of age. Immunity 23 (4), 344.CrossRefGoogle ScholarPubMed
Mantovani, A, Sica, A et al. (2004). The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 25 (12), 677.CrossRefGoogle Scholar
Verreck, FA, de Boer, T et al. (2004). Human IL-23-producing type 1 macrophages promote but IL-10-producing type 2 macrophages subvert immunity to (myco)bacteria. Proc Natl Acad Sci U S A 101 (13), 4560.CrossRefGoogle ScholarPubMed
Mantovani, A, Bottazzi, B et al. (1992). The origin and function of tumor-associated macrophages. Immunol Today 13 (7), 265.CrossRefGoogle ScholarPubMed
Mantovani, A, Sozzani, S et al. (2002). Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol 23 (11), 549.CrossRefGoogle ScholarPubMed
Pollard, JW (2004). Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer 4 (1), 71.CrossRefGoogle ScholarPubMed
Coussens, LM, Tinkle, CL et al. (2000). MMP-9 supplied by bone marrow-derived cells contributes to skin carcinogenesis. Cell 103 (3), 481.CrossRefGoogle ScholarPubMed
Luo, JL, Tan, W et al. (2007). Nuclear cytokine-activated IKKalpha controls prostate cancer metastasis by repressing Maspin. Nature 446 (7136), 690.CrossRefGoogle ScholarPubMed
Sparmann, A, Bar-Sagi, D (2004). Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell 6 (5), 447.CrossRefGoogle Scholar
Yu, H, Kortylewski, M et al. (2007). Crosstalk between cancer and immune cells: role of STAT3 in the tumour microenvironment. Nat Rev Immunol 7 (1), 41.CrossRefGoogle ScholarPubMed
Rius, J, Guma, M et al. (2008). NF-kappaB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1alpha. Nature 453 (7196), 807.CrossRefGoogle ScholarPubMed
Zinkernagel, AS, Johnson, RS et al. (2007). Hypoxia inducible factor (HIF) function in innate immunity and infection. J Mol Med 85 (12), 1339.CrossRefGoogle ScholarPubMed
Rossi, D, Zlotnik, A (2000). The biology of chemokines and their receptors. Annu Rev Immunol 18, 217.CrossRefGoogle ScholarPubMed
Mantovani, A, Muzio, M et al. (2001). Macrophage control of inflammation: negative pathways of regulation of inflammatory cytokines. Novartis Found Symp 234, 120.Google ScholarPubMed
Norgauer, J, Metzner, B et al. (1996). Expression and growth-promoting function of the IL-8 receptor beta in human melanoma cells. J Immunol 156 (3), 1132.Google ScholarPubMed
Ottaiano, A, Franco, R et al. (2006). Overexpression of both CXC chemokine receptor 4 and vascular endothelial growth factor proteins predicts early distant relapse in stage II-III colorectal cancer patients. Clin Cancer Res 12 (9), 2795.CrossRefGoogle ScholarPubMed
Richmond, A, Thomas, HG (1986). Purification of melanoma growth stimulatory activity. J Cell Physiol 129 (3), 375.CrossRefGoogle ScholarPubMed
Havell, EA, Fiers, W et al. (1988). The antitumor function of tumor necrosis factor (TNF), I. Therapeutic action of TNF against an established murine sarcoma is indirect, immunologically dependent, and limited by severe toxicity. J Exp Med 167 (3), 1067.CrossRefGoogle Scholar
Galban, S, Fan, J et al. (2003). von Hippel-Lindau protein-mediated repression of tumor necrosis factor alpha translation revealed through use of cDNA arrays. Mol Cell Biol 23 (7), 2316.CrossRefGoogle ScholarPubMed
Tracey, KJ, Lowry, SF et al. (1986). Cachectin/tumor necrosis factor mediates changes of skeletal muscle plasma membrane potential. J Exp Med 164 (4), 1368.CrossRefGoogle ScholarPubMed
Anasagasti, MJ, Olaso, E et al. (1997). Interleukin 1-dependent and -independent mouse melanoma metastases. J Natl Cancer Inst 89 (9), 645.CrossRefGoogle ScholarPubMed
Apte, RN, Voronov, E (2002). Interleukin-1–a major pleiotropic cytokine in tumor-host interactions. Semin Cancer Biol 12 (4), 277.CrossRefGoogle ScholarPubMed
Song, X, Voronov, E et al. (2003). Differential effects of IL-1 alpha and IL-1 beta on tumorigenicity patterns and invasiveness. J Immunol 171 (12), 6448.CrossRefGoogle ScholarPubMed
Dinarello, CA, Schindler, R (1990). Dissociation of transcription from translation of human IL-1-beta: the induction of steady state mRNA by adherence or recombinant C5a in the absence of translation. Prog Clin Biol Res 349, 195.Google ScholarPubMed
Greten, FR, Arkan, MC et al. (2007). NF-kappaB is a negative regulator of IL-1beta secretion as revealed by genetic and pharmacological inhibition of IKKbeta. Cell 130 (5), 918.CrossRefGoogle ScholarPubMed
Chen, CJ, Kono, H et al. (2007). Identification of a key pathway required for the sterile inflammatory response triggered by dying cells. Nat Med 13 (7), 851.CrossRefGoogle ScholarPubMed
Harris, TB, Ferrucci, L et al. (1999). Associations of elevated interleukin-6 and C-reactive protein levels with mortality in the elderly. Am J Med 106 (5), 506.CrossRefGoogle ScholarPubMed
Kiecolt-Glaser, JK, Preacher, KJ et al. (2003). Chronic stress and age-related increases in the proinflammatory cytokine IL-6. Proc Natl Acad Sci U S A 100 (15), 9090.CrossRefGoogle ScholarPubMed
Gallucci, M, Amici, GP et al. (2007). Associations of the plasma interleukin 6 (IL-6) levels with disability and mortality in the elderly in the Treviso Longeva (Trelong) study. Arch Gerontol Geriatr 44 Suppl 1, 193.CrossRefGoogle ScholarPubMed
Ershler, WB, Keller, ET (2000). Age-associated increased interleukin-6 gene expression, late-life diseases, and frailty. Annu Rev Med 51, 245.CrossRefGoogle ScholarPubMed
Trikha, M, Corringham, R et al. (2003). Targeted anti-interleukin-6 monoclonal antibody therapy for cancer: a review of the rationale and clinical evidence. Clin Cancer Res 9 (13), 4653.Google ScholarPubMed
Honemann, D, Chatterjee, M et al. (2001). The IL-6 receptor antagonist SANT-7 overcomes bone marrow stromal cell-mediated drug resistance of multiple myeloma cells. Int J Cancer 93 (5), 674.CrossRefGoogle ScholarPubMed
Naugler, WE, Sakurai, T et al. (2007). Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science 317 (5834), 121.CrossRefGoogle ScholarPubMed
Choy, H, Milas, L (2003). Enhancing radiotherapy with cyclooxygenase-2 enzyme inhibitors: a rational advance? J Natl Cancer Inst 95 (19), 1440.CrossRefGoogle ScholarPubMed
Gasparini, G, Longo, R et al. (2003). Inhibitors of cyclo-oxygenase 2: a new class of anticancer agents? Lancet Oncol 4 (10), 605.CrossRefGoogle ScholarPubMed
Baek, SJ, Eling, TE (2006). Changes in gene expression contribute to cancer prevention by COX inhibitors. Prog Lipid Res 45 (1), 1.CrossRefGoogle ScholarPubMed
Harmey, JH, Bucana, CD et al. (2002). Lipopolysaccharide-induced metastatic growth is associated with increased angiogenesis, vascular permeability and tumor cell invasion. Int J Cancer 101 (5), 415.CrossRefGoogle ScholarPubMed
Pidgeon, GP, Harmey, JH et al. (1999). The role of endotoxin/lipopolysaccharide in surgically induced tumour growth in a murine model of metastatic disease. Br J Cancer 81 (8), 1311.CrossRefGoogle Scholar
Taketomi, A, Takenaka, K et al. (1997). Circulating intercellular adhesion molecule-1 in patients with hepatocellular carcinoma before and after hepatic resection. Hepatogastroenterology 44 (14), 477.Google ScholarPubMed
Wilson, J and Balkwill, F (2002). The role of cytokines in the epithelial cancer microenvironment. Semin Cancer Biol 12 (2), 113.CrossRefGoogle ScholarPubMed
Song, K, Chen, Y et al. (2000). Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an inhibitor of autoimmune inflammation and cell cycle progression. J Exp Med 191 (7), 1095.CrossRefGoogle ScholarPubMed
Ashkenazi, A, Pai, RC et al. (1999). Safety and antitumor activity of recombinant soluble Apo2 ligand. J Clin Invest 104 (2), 155.CrossRefGoogle ScholarPubMed
Smyth, MJ, Cretney, E et al. (2001). Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) contributes to interferon gamma-dependent natural killer cell protection from tumor metastasis. J Exp Med 193 (6), 661.CrossRefGoogle ScholarPubMed
Dranoff, G (2004). Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer 4 (1), 11.CrossRefGoogle ScholarPubMed
Dranoff, G (1997). Interferon alfa versus chemotherapy for chronic myeloid leukemia: a meta-analysis of seven randomized trials: Chronic Myeloid Leukemia Trialists' Collaborative Group. J Natl Cancer Inst 89 (21), 1616.Google Scholar
Fyfe, G, Fisher, RI et al. (1995). Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J Clin Oncol 13 (3), 688.CrossRefGoogle ScholarPubMed
Rosenberg, SA, Lotze, MT et al. (1993). Prospective randomized trial of high-dose interleukin-2 alone or in conjunction with lymphokine-activated killer cells for the treatment of patients with advanced cancer. J Natl Cancer Inst 85 (8), 622.CrossRefGoogle ScholarPubMed
Mach, N, Dranoff, G (2000). Cytokine-secreting tumor cell vaccines. Curr Opin Immunol 12 (5), 571.CrossRefGoogle ScholarPubMed
Hiratsuka, S, Nakamura, K et al. (2002). MMP9 induction by vascular endothelial growth factor receptor-1 is involved in lung-specific metastasis. Cancer Cell 2 (4), 289.CrossRefGoogle ScholarPubMed
Kaplan, RN, Riba, RD et al. (2005). VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 438 (7069), 820.CrossRefGoogle ScholarPubMed
Pirinen, R, Leinonen, T et al. (2005). Versican in nonsmall cell lung cancer: relation to hyaluronan, clinicopathologic factors, and prognosis. Hum Pathol 36 (1), 44.CrossRefGoogle ScholarPubMed
Ricciardelli, C, Russell, DL et al. (2007). Formation of hyaluronan- and versican-rich pericellular matrix by prostate cancer cells promotes cell motility. J Biol Chem 282 (14), 10814.CrossRefGoogle ScholarPubMed
Yee, AJ, Akens, M et al. (2007). The effect of versican G3 domain on local breast cancer invasiveness and bony metastasis. Breast Cancer Res 9 (4), R47.CrossRefGoogle Scholar
Rahmani, M, Wong, BW et al. (2006). Versican: signaling to transcriptional control pathways. Can J Physiol Pharmacol 84 (1), 77.CrossRefGoogle ScholarPubMed
Zheng, PS, Wen, J et al. (2004). Versican/PG-M G3 domain promotes tumor growth and angiogenesis. Faseb J 18 (6), 754.CrossRefGoogle Scholar
Schaefer, L, Babelova, A et al. (2005). The matrix component biglycan is proinflammatory and signals through Toll-like receptors 4 and 2 in macrophages. J Clin Invest 115 (8), 2223.CrossRefGoogle ScholarPubMed
Brown, LF, Dvorak, AM et al. (1989). Leaky vessels, fibrin deposition, and fibrosis: a sequence of events common to solid tumors and to many other types of disease. Am Rev Respir Dis 140 (4), 1104.CrossRefGoogle Scholar
Dvorak, HF (1986). Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med 315 (26), 1650.Google Scholar
Massague, J, Blain, SW et al. (2000). TGFbeta signaling in growth control, cancer, and heritable disorders. Cell 103 (2), 295.CrossRefGoogle ScholarPubMed
Bierie, B, Moses, HL (2006). Tumour microenvironment: TGFbeta: the molecular Jekyll and Hyde of cancer. Nat Rev Cancer 6 (7), 506.CrossRefGoogle Scholar
Dumont, N, Arteaga, CL (2003). Targeting the TGF beta signaling network in human neoplasia. Cancer Cell 3 (6), 531.CrossRefGoogle ScholarPubMed
Padua, D, Zhang, XH et al. (2008). TGFbeta primes breast tumors for lung metastasis seeding through angiopoietin-like 4. Cell 133 (1), 66.CrossRefGoogle ScholarPubMed
Alberti, C (2008). Genetic and microenvironmental implications in prostate cancer progression and metastasis. Eur Rev Med Pharmacol Sci 12 (3), 167.Google ScholarPubMed
Yang, J, Mani, SA et al. (2004). Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117 (7), 927.CrossRefGoogle ScholarPubMed
Leptin, M, Grunewald, B (1990). Cell shape changes during gastrulation in Drosophila. Development 110 (1), 73.Google ScholarPubMed
Thisse, B, el Messal, M et al. (1987). The twist gene: isolation of a Drosophila zygotic gene necessary for the establishment of dorsoventral pattern. Nucleic Acids Res 15 (8), 3439.CrossRefGoogle ScholarPubMed
Chen, ZF, Behringer, RR (1995). Twist is required in head mesenchyme for cranial neural tube morphogenesis. Genes Dev 9 (6), 686.CrossRefGoogle ScholarPubMed
Soo, K, O'Rourke, MP et al. (2002). Twist function is required for the morphogenesis of the cephalic neural tube and the differentiation of the cranial neural crest cells in the mouse embryo. Dev Biol 247 (2), 251.CrossRefGoogle ScholarPubMed
Hay, ED (1995). An overview of epithelio-mesenchymal transformation. Acta Anat (Basel) 154 (1), 8.CrossRefGoogle ScholarPubMed
Pham, CG, Bubici, C et al. (2007). Upregulation of Twist-1 by NF-kappaB blocks cytotoxicity induced by chemotherapeutic drugs. Mol Cell Biol 27 (11), 3920.CrossRefGoogle ScholarPubMed
Steeg, PS (2003). Metastasis suppressors alter the signal transduction of cancer cells. Nat Rev Cancer 3 (1), 55.CrossRefGoogle ScholarPubMed
Lockett, J, Yin, S et al. (2006). Tumor suppressive maspin and epithelial homeostasis. J Cell Biochem 97 (4), 651.CrossRefGoogle ScholarPubMed
Zou, Z, Anisowicz, A et al. (1994). Maspin, a serpin with tumor-suppressing activity in human mammary epithelial cells. Science 263 (5146), 526.CrossRefGoogle ScholarPubMed
Baron, JA, Sandler, RS (2000). Nonsteroidal anti-inflammatory drugs and cancer prevention. Annu Rev Med 51, 511.CrossRefGoogle ScholarPubMed
Garcia-Rodriguez, , Huerta-Alvarez, C (2001). Reduced risk of colorectal cancer among long-term users of aspirin and nonaspirin nonsteroidal antiinflammatory drugs. Epidemiology 12 (1), 88.CrossRefGoogle ScholarPubMed
Mamytbekova, A, Rezabek, K et al. (1986). Antimetastatic effect of flurbiprofen and other platelet aggregation inhibitors. Neoplasma 33 (4), 417.Google ScholarPubMed
Elder, DJ, Halton, et al. (1997). Induction of apoptotic cell death in human colorectal carcinoma cell lines by a cyclooxygenase-2 (COX-2)-selective nonsteroidal anti-inflammatory drug: independence from COX-2 protein expression. Clin Cancer Res 3 (10), 1679.Google ScholarPubMed
Marx, J (2004). Cancer research. Inflammation and cancer: the link grows stronger. Science 306 (5698), 966.CrossRefGoogle ScholarPubMed

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
×