Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-gtxcr Total loading time: 0 Render date: 2024-04-19T08:26:10.522Z Has data issue: false hasContentIssue false

6 - The Role of Metastasis Suppressor Genes in Metastasis

from GENES

Published online by Cambridge University Press:  05 June 2012

Brunilde Gril
Affiliation:
National Cancer Institute, United States
Russell Szmulewitz
Affiliation:
The University of Chicago, Committee on Cancer Biology and Pritzker School of Medicine, United States
Joshua Collins
Affiliation:
National Cancer Institute, United States
Jennifer Taylor
Affiliation:
The University of Chicago, Committee on Cancer Biology and Pritzker School of Medicine, United States
Carrie Rinker-Schaeffer
Affiliation:
The University of Chicago, Committee on Cancer Biology and Pritzker School of Medicine, United States
Patricia Steeg
Affiliation:
National Cancer Institute, United States
Jean-Claude Marshall
Affiliation:
National Cancer Institute, 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

In the 1970s and 1980s, clever scientific insight and innovation rapidly advanced our understanding of the molecular mechanisms of cancer biology. The discoveries of oncogenes and tumor suppressors, and the elucidation of their functions, greatly aided in studies aimed at a molecular understanding of the etiology of primary tumors. Despite this, cancer biologists had little understanding of the molecular aspects of metastasis. Considering the devastating consequences, scientists were anxious for a breakthrough. The first clue would come from the study of tumor suppressors.

Tumor suppressor genes were identified when it was discovered that their loss of function was critical to tumorigenesis. Prior to their discovery, researchers were of the mindset that the oncogenic phenotype was always dominant. In other words, a mutation need happen on only a single allele for a normal cell to be transformed into a tumor cell. However, not all disease incidence data seemed to fit neatly into this hypothesis. By studying retinoblastoma case histories, a “two-hit” hypothesis emerged, predicting that for at least some cancers, two mutations must occur (one on each allele) to successfully transform a cell [1]. Indeed, the retinoblastoma gene, or Rb, would become known as the first described tumor suppressor. We now know that the “two hits” need not come in the form of distinct somatic mutations but may be the result of any combination of germinal and/or somatic mutations, mitotic recombinations, gene conversions, and functional inactivation of genes owing to promoter hypermethylation.

Type
Chapter
Information
Cancer Metastasis
Biologic Basis and Therapeutics
, pp. 64 - 76
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

Knudson, AG. (1971) Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci USA. 68: 820–3.CrossRefGoogle ScholarPubMed
Steeg, PS, Bevilacqua, G, Pozzatti, R, Liotta, , Sobel, ME (1988) Altered expression of NM23, a gene associated with low tumor metastatic potential, during adenovirus 2 Ela inhibition of experimental metastasis. Cancer Res. 48: 6550–4.Google ScholarPubMed
Steeg, PS, Bevilacqua, G, Kopper, L et al. (1988) Evidence for a novel gene associated with low tumor metastatic potential. J Natl Cancer Inst. 80: 200–4.CrossRefGoogle ScholarPubMed
Leone, A, Flatow, U, King, CR et al. (1991) Reduced tumor incidence, metastatic potential, and cytokine responsiveness of nm23-transfected melanoma cells. Cell. 65: 25–35.CrossRefGoogle ScholarPubMed
Leone, A, Flatow, U, VanHoutte, K, Steeg, PS (1993) Transfection of human nm23-H1 into the human MDA-MB-435 breast carcinoma cell line: effects on tumor metastatic potential, colonization and enzymatic activity. Oncogene. 8: 2325–33.Google ScholarPubMed
McNeill, CA, Brown, RL (1980) Genetic manipulation by means of microcell-mediated transfer of normal human chromosomes into recipient mouse cells. Proc Natl Acad Sci USA. 77: 5394–8.CrossRefGoogle ScholarPubMed
Welch, DR (1997) Technical considerations for studying cancer metastasis in vivo. Clin Exp Metastasis. 15: 272–306.CrossRefGoogle ScholarPubMed
Steeg, PS (2003) Metastasis suppressors alter the signal transduction of cancer cells. Nat Rev Cancer. 3: 55–63.CrossRefGoogle ScholarPubMed
Steeg, PS, Bevilacqua, G, Kopper, L et al. (1988) Evidence for a novel gene associated with low tumor metastatic potential. J Natl Cancer Inst. 80: 200–4.CrossRefGoogle ScholarPubMed
Steeg, P (2003) Metastasis suppressors alter the signal transduction of cancer cells. Nature Cancer Rev. 3: 55–63.CrossRefGoogle ScholarPubMed
Rinker-Schaeffer, CW, O'Keefe, JP, Welch, DR, Theodorescu, D (2006) Metastasis suppressor proteins: discovery, molecular mechanisms, and clinical application. Clin Cancer Res. 12: 3882–9.CrossRefGoogle ScholarPubMed
Parhar, RS, Shi, Y, Zou, M, Farid, NR, Ernst, P, al-Sedairy, ST (1995) Effects of cytokine-mediated modulation of nm23 expression on the invasion and metastatic behavior of B16F10 melanoma cells. Int J Cancer. 60: 204–10.CrossRefGoogle ScholarPubMed
Lee, HY, Lee, H (1999) Inhibitory activity of nm23-H1 on invasion and colonization of human prostate carcinoma cells is not mediated by its NDP kinase activity. Cancer Lett. 145: 93–9.CrossRefGoogle Scholar
Tagashira, H, Hamazaki, K, Tanaka, N, Gao, C, Namba, M (1998) Reduced metastatic potential and c-myc overexpression of colon adenocarcinoma cells (Colon 26 line) transfected with nm23-R2/rat nucleoside diphosphate kinase alpha isoform. Int J Mol Med. 2: 65–8.Google ScholarPubMed
Bhujwalla, ZM, Aboagye, EO, Gillies, RJ, Chacko, VP, Mendola, CE, Backer, JM (1999) Nm23-transfected MDA-MB-435 human breast carcinoma cells form tumors with altered phospholipid metabolism and pH: a 31P nuclear magnetic resonance study in vivo and in vitro. Magn Reson Med. 41: 897–903.3.0.CO;2-T>CrossRefGoogle ScholarPubMed
Miyazaki, H, Fukuda, M, Ishijima, Y et al. (1999) Overexpression of Nm23-H2/NDP kinase B in a human oral squamous cell carcinoma cell line results in reduced metastasis, differentiated phenotype in the metastatic site, and growth factor-independent proliferative activity in culture. Clin Cancer Res. 5: 4301–7.Google Scholar
Boissan, M, Wendum, D, Arnaud-Dabernat, S, et al. (2005) Increased lung metastasis in transgenic NM23-Null/SV40 mice with hepatocellular carcinoma. J Natl Cancer Inst. 97: 836–45.CrossRefGoogle ScholarPubMed
Lacombe, ML, Milon, L, Munier, A, Mehus, JG, Lambeth, (2000) The human Nm23/nucleoside diphosphate kinases. J Bioenerg Biomembr. 32: 247–58.CrossRefGoogle ScholarPubMed
McDermott, WG, Boissan, M, Lacombe, ML, Steeg, PS, Horak, CE (2008) Nm23-H1 homologs suppress tumor cell motility and anchorage independent growth. Clin Exp Metastasis. 25: 131–8.CrossRefGoogle ScholarPubMed
Hartsough, MT, Morrison, DK, Salerno, M et al. (2002) Nm23-H1 metastasis suppressor phosphorylation of kinase suppressor of Ras via a histidine protein kinase pathway. J Biol Chem. 277: 32389–99.CrossRefGoogle Scholar
Aguirre-Ghiso, JA, Estrada, Y, Liu, D, Ossowski, L (2003) ERK(MAPK) activity as a determinant of tumor growth and dormancy; regulation by p38(SAPK). Cancer Res. 63: 1684–95.Google ScholarPubMed
Aguirre-Ghiso, JA, Ossowski, L, Rosenbaum, SK (2004) Green fluorescent protein tagging of extracellular signal-regulated kinase and p38 pathways reveals novel dynamics of pathway activation during primary and metastatic growth. Cancer Res. 64: 7336–45.CrossRefGoogle ScholarPubMed
Ranganathan, AC, Adam, AP, Aguirre-Ghiso, JA (2006) Opposing roles of mitogenic and stress signaling pathways in the induction of cancer dormancy. Cell Cycle. 5: 1799–807.CrossRefGoogle ScholarPubMed
Steeg, PS, Horak, CE, Miller, KD (2008) Clinical-translational approaches to the Nm23-H1 metastasis suppressor. Clin Cancer Res. 14: 5006–12.CrossRefGoogle ScholarPubMed
Horak, CE, Lee, JH, Elkahloun, AG et al. (2007) Nm23-H1 suppresses tumor cell motility by down-regulating the lysophosphatidic acid receptor EDG2. Cancer Res. 67: 7238–46.CrossRefGoogle ScholarPubMed
Horak, CE, Mendoza, A, Vega-Valle, E et al. (2007) Nm23-H1 suppresses metastasis by inhibiting expression of the lysophosphatidic acid receptor EDG2. Cancer Res. 15;67(24):11751–9.CrossRefGoogle Scholar
Ouatas, T, Halverson, D, Steeg, PS (2003) Dexamethasone and medroxyprogesterone acetate elevate Nm23-H1 metastasis suppressor gene expression in metastatic human breast carcinoma cells: new uses for old compounds. Clin Cancer Res. 9: 3763–72.Google ScholarPubMed
Ohtaki, T, Shintani, Y, Honda, S et al. (2001) Metastasis suppressor gene KiSS-1 encodes peptide ligand of a G-protein-coupled receptor. Nature. 411: 613–7.CrossRefGoogle ScholarPubMed
Kotani, M, Detheux, M, Vandenbogaerde, A et al. (2001) The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54. J Biol Chem. 276: 34631–6.CrossRefGoogle ScholarPubMed
Castellano, JM, Roa, J, Luque, RM et al. (2008) KiSS-1/kisspeptins and the metabolic control of reproduction: Physiologic roles and putative physiopathological implications. Peptides. 30(1): 139–45.CrossRefGoogle ScholarPubMed
Nash, KT, Phadke, PA, Navenot, JM et al. (2007) Requirement of KISS1 secretion for multiple organ metastasis suppression and maintenance of tumor dormancy. J Natl Cancer Inst. 99: 309–21.CrossRefGoogle ScholarPubMed
Ichikawa, T, Ichikawa, Y, Isaacs, JT (1991) Genetic factors and metastatic potential of prostatic cancer. Cancer Surv. 11: 35–42.Google ScholarPubMed
Takahashi, M, Sugiura, T, Abe, M, Ishii, K, Shirasuna, K (2007) Regulation of c-Met signaling by the tetraspanin KAI-1/CD82 affects cancer cell migration. Int J Cancer. 121: 1919–29.CrossRefGoogle ScholarPubMed
Bandyopadhyay, S, Zhan, R, Chaudhuri, A et al. (2006) Interaction of KAI1 on tumor cells with DARC on vascular endothelium leads to metastasis suppression. Nat Med. 12: 933–8.CrossRefGoogle ScholarPubMed
Prince, S, Carreira, S, Vance, KW, Abrahams, A, Goding, CR. (2004) Tbx2 directly represses the expression of the p21(WAF1) cyclin-dependent kinase inhibitor. Cancer Res. 64: 1669–74.CrossRefGoogle ScholarPubMed
Tsai, YC, Mendoza, A, Mariano, JM et al. (2007) The ubiquitin ligase gp78 promotes sarcoma metastasis by targeting KAI1 for degradation. Nat Med. 13: 1504–9.CrossRefGoogle ScholarPubMed
Sridhar, SC, Miranti, CK (2006) Tetraspanin KAI1/CD82 suppresses invasion by inhibiting integrin-dependent crosstalk with c-Met receptor and Src kinases. Oncogene. 25: 2367–78.CrossRefGoogle ScholarPubMed
Chekmareva, MA, Hollowell, CM, Smith, RC, Davis, EM, LeBeau, MM, Rinker-Schaeffer, CW (1997) Localization of prostate cancer metastasis-suppressor activity on human chromosome 17. Prostate. 33: 271–80.3.0.CO;2-K>CrossRefGoogle ScholarPubMed
Vander Griend, DJ, Kocherginsky, M, Hickson, JA, Stadler, WM, Lin, A, Rinker-Schaeffer, CW (2005) Suppression of metastatic colonization by the context-dependent activation of the c-Jun NH2-terminal kinase kinases JNKK1/MKK4 and MKK7. Cancer Res. 65: 10984–91.CrossRefGoogle ScholarPubMed
Yamada, SD, Hickson, JA, Hrobowski, Y et al. (2002) Mitogen-activated protein kinase kinase 4 (MKK4) acts as a metastasis suppressor gene in human ovarian carcinoma. Cancer Res. 62: 6717–23.Google ScholarPubMed
Hickson, JA, Huo, D, Vander Griend, DJ, Lin, A, Rinker-Schaeffer, CW, Yamada, SD (2006) The p38 kinases MKK4 and MKK6 suppress metastatic colonization in human ovarian carcinoma. Cancer Res. 66: 2264–70.CrossRefGoogle ScholarPubMed
Chang, L, Karin, M (2001) Mammalian MAP kinase signalling cascades. Nature. 410: 37–40.CrossRefGoogle ScholarPubMed
Whitmarsh, AJ, Davis, RJ (2007) Role of mitogen-activated protein kinase kinase 4 in cancer. Oncogene. 26: 3172–84.CrossRefGoogle Scholar
Teng, DH, Perry, WL, Hogan, JK et al. (1997) Human mitogen-activated protein kinase kinase 4 as a candidate tumor suppressor. Cancer Res. 57: 4177–82.Google ScholarPubMed
Su, GH, Song, JJ, Repasky, EA, Schutte, M, Kern, SE (2002) Mutation rate of MAP2K4/MKK4 in breast carcinoma. Human Mutation. 19: 81.CrossRefGoogle ScholarPubMed
Su, GH, Hilgers, W, Shekher, MC et al. (1998) Alterations in pancreatic, biliary, and breast carcinomas support MKK4 as a genetically targeted tumor suppressor gene. Cancer Res. 58: 2339–42.Google ScholarPubMed
Xin, W, Yun, KJ, Ricci, F et al. (2004) MAP2K4/MKK4 expression in pancreatic cancer: genetic validation of immunohistochemistry and relationship to disease course. Clin Cancer Res. 10: 8516–20.CrossRefGoogle ScholarPubMed
Cunningham, SC, Kamangar, F, Kim, MP et al. (2006) MKK4 status predicts survival after resection of gastric adenocarcinoma. Arch Surg. 141: 1095–9; discussion 1100.CrossRefGoogle ScholarPubMed
Stark, AM, Tongers, K, Maass, N, Mehdorn, HM, Held-Feindt, J (2005) Reduced metastasis-suppressor gene mRNA-expression in breast cancer brain metastases. J Cancer Res Clin Oncol. 131: 191–8.CrossRefGoogle ScholarPubMed
Kim, HL, Vander Griend, DJ, Yang, X et al. (2001) Mitogen-activated protein kinase kinase 4 metastasis suppressor gene expression is inversely related to histological pattern in advancing human prostatic cancers. Cancer Res. 61: 2833–7.Google ScholarPubMed
Kennedy, NJ, Davis, RJ (2003) Role of JNK in tumor development. Cell Cycle. 2: 199–201.Google ScholarPubMed
Wang, L, Pan, Y, Dai, JL (2004) Evidence of MKK4 pro-oncogenic activity in breast and pancreatic tumors. Oncogene. 7: 7.Google Scholar
Cunningham, SC, Gallmeier, E, Hucl, T et al. (2006) Targeted deletion of MKK4 in cancer cells: a detrimental phenotype manifests as decreased experimental metastasis and suggests a counterweight to the evolution of tumor-suppressor loss. Cancer Res. 66: 5560–4.CrossRefGoogle ScholarPubMed
Lotan, TL, Lyon, M, Huo, D et al. (2007) Up-regulation of MKK4, MKK6 and MKK7 during prostate cancer progression: an important role for SAPK signalling in prostatic neoplasia. J Pathol. 212: 386–94.CrossRefGoogle ScholarPubMed
Lotan, T, Hickson, J, Souris, J et al. (2008) JNKK1/MKK4 mediated inhibition of SKOV3ip.1 ovarian cancer metastasis involves growth arrest and p21 upregulation. Cancer Res. 1;68(7):2166–75.CrossRefGoogle Scholar
Seraj, MJ, Samant, RS, Verderame, MF, Welch, DR (2000) Functional evidence for a novel human breast carcinoma metastasis suppressor, BRMS1, encoded at chromosome 11q13. Cancer Res. 60: 2764–9.Google ScholarPubMed
Shevde, , Samant, RS, Goldberg, SF et al. (2002) Suppression of human melanoma metastasis by the metastasis suppressor gene, BRMS1. Exp Cell Res. 273: 229–39.CrossRefGoogle ScholarPubMed
Seraj, MJ, Harding, MA, Gildea, JJ, Welch, DR, Theodorescu, D (2000) The relationship of BRMS1 and RhoGDI2 gene expression to metastatic potential in lineage related human bladder cancer cell lines. Clin Exp Metastasis. 18: 519–25.CrossRefGoogle ScholarPubMed
Saunders, MM, Seraj, MJ, Li, Z et al. Breast cancer metastatic potential correlates with a breakdown in homospecific and heterospecific gap junctional intercellular communication. Cancer Res 2001) 61: 1765–7.Google ScholarPubMed
Hedley, BD, Vaidya, KS, Phadke, P et al. (2008) BRMS1 suppresses breast cancer metastasis in multiple experimental models of metastasis by reducing solitary cell survival and inhibiting growth initiation. Clin Exp Metastasis. 25(7):727–40.CrossRefGoogle ScholarPubMed
DeWald, DB, Torabinejad, J, Samant, RS et al. (2005) Metastasis suppression by breast cancer metastasis suppressor 1 involves reduction of phosphoinositide signaling in MDA-MB-435 breast carcinoma cells. Cancer Res. 65: 713–7.Google ScholarPubMed
Zhang, S, Lin, QD, Di, W (2006) Suppression of human ovarian carcinoma metastasis by the metastasis-suppressor gene, BRMS1. Int J Gynecol Cancer. 16: 522–31.CrossRefGoogle ScholarPubMed
Vaidya, KS, Harihar, S, Phadke, PA et al. (2008) Breast cancer metastasis suppressor-1 differentially modulates growth factor signaling. J Biol Chem. 283: 28354–60.CrossRefGoogle ScholarPubMed
Hurst, DR, Xie, Y, Vaidya, KS et al. (2008) Alterations of BRMS1-ARID4A interaction modify gene expression but still suppress metastasis in human breast cancer cells. J Biol Chem. 283: 7438–44.CrossRefGoogle ScholarPubMed
Gildea, JJ, Seraj, MJ, Oxford, G et al. (2002) RhoGDI2 is an invasion and metastasis suppressor gene in human cancer. Cancer Res. 62: 6418–23.Google ScholarPubMed
Titus, B, Frierson, HF., Conaway, M et al. (2005) Endothelin axis is a target of the lung metastasis suppressor gene RhoGDI2. Cancer Res. 65: 7320–7.CrossRefGoogle ScholarPubMed
Chiappori, AA, Haura, E, Rodriguez, FA et al. (2008) Phase I/II study of atrasentan, an endothelin A receptor antagonist, in combination with paclitaxel and carboplatin as first-line therapy in advanced non-small cell lung cancer. Clin Cancer Res. 14: 1464–9.CrossRefGoogle ScholarPubMed
Hagan, S, Al-Mulla, F, Mallon, E et al. (2005) Reduction of Raf-1 kinase inhibitor protein expression correlates with breast cancer metastasis. Clin Cancer Res. 11: 7392–7.CrossRefGoogle ScholarPubMed
Fu, Z, Smith, PC, Zhang, L et al. (2003) Effects of raf kinase inhibitor protein expression on suppression of prostate cancer metastasis. J Natl Cancer Inst. 95: 878–89.CrossRefGoogle ScholarPubMed
Keller, ET (2004) Metastasis suppressor genes: a role for raf kinase inhibitor protein (RKIP). Anticancer Drugs. 15: 663–9.CrossRefGoogle ScholarPubMed
Chatterjee, D, Bai, Y, Wang, Z et al. (2004) RKIP sensitizes prostate and breast cancer cells to drug-induced apoptosis. J Biol Chem. 279: 17515–23.CrossRefGoogle ScholarPubMed
Zlobec, I, Baker, K, Minoo, P, Jass, JR, Terracciano, L, Lugli, A (2008) Node-negative colorectal cancer at high risk of distant metastasis identified by combined analysis of lymph node status, vascular invasion, and Raf-1 kinase inhibitor protein expression. Clin Cancer Res. 14: 143–8.CrossRefGoogle ScholarPubMed
Granovsky, AE, Rosner, MR (2008) Raf kinase inhibitory protein: a signal transduction modulator and metastasis suppressor. Cell Res. 18: 452–7.CrossRefGoogle ScholarPubMed
Iizumi, M, Bandyopadhyay, S, Watabe, K. Interaction of Duffy antigen receptor for chemokines and Kai1: a critical step in metastasis suppression. Cancer Res. 67: 1411–14.CrossRef
Eves, EM, Shapiro, P, Naik, K, Klein, UR, Trakul, N, Rosner, MR (2006) Raf kinase inhibitory protein regulates aurora B kinase and the spindle checkpoint. Mol Cell 23: 561–74.CrossRefGoogle ScholarPubMed
Fu, Z, Smith, PC, Zhang, L, Rubin, MA, Dunn, RL, Yao, Z, Keller, ET (2003) Effects of raf kinase inhibitor protein expression on suppression of prostate cancer metastasis. J Natl Cancer Inst 95: 878–89.CrossRefGoogle ScholarPubMed
Schuierer, MM, Bataille, F, Hagan, S, Kolch, W, Bosserhoff, AK (2004) Reduction in Raf kinase inhibitor protein expression is associated with increased Ras-extracellular signal-regulated kinase signaling in melanoma cell lines. Cancer Res 64: 5186–92.CrossRefGoogle ScholarPubMed
Rodrigues, S, Wever, O, Bruyneel, E, Rooney, RJ, Gespach, C (2007) Opposing roles of netrin-1 and the dependence receptor DCC in cancer cell invasion, tumor growth and metastasis. Oncogene 26: 5615–25.CrossRefGoogle Scholar
Stupack, DG, Teitz, T, Potter, MD, Mikolon, D, Houghton, PJ, Kidd, VJ, Lahti, JM, Cheresh, DA (2006) Potentiation of neuroblastoma metastasis by loss of caspase-8. Nature 439: 95–99.CrossRefGoogle ScholarPubMed
Fujita, H, Okada, F, Hamada, J, Hosokawa, M, Moriuchi, T, Koya, RC, Kuzumaki, N (2001) Gelsolin functions as a metastasis suppressor in B16-BL6 mouse melanoma cells and requirement of the carboxyl-terminus for its effect. Int J Cancer 93: 773–80.CrossRefGoogle ScholarPubMed
Kippenberger, S, Loitsch, S, Thaci, D, Muller, J, Guschel, M, Kaufmann, R, Bernd, A (2006) Restoration of E-cadherin sensitizes human melanoma cells for apoptosis. Melanoma Res 16: 393–403.CrossRefGoogle ScholarPubMed
Michl, P, Barth, C, Buchholz, M, Lerch, MM, Rolke, M, Holzmann, KH, Menke, A, Fensterer, H, Giehl, K, Lohr, M, et al. (2003) Claudin-4 expression decreases invasiveness and metastatic potential of pancreatic cancer. Cancer Res 63: 6265–71.Google ScholarPubMed
Gao, AC, Lou, W, Dong, JT, Isaacs, JT (1997) CD44 is a metastasis suppressor gene for prostatic cancer located on human chromosome 11p13. Cancer Res 57: 846–9.Google ScholarPubMed
Gao, AC, Lou, W, Sleeman, JP, Isaacs, JT (1998) Metastasis suppression by the standard CD44 isoform does not require the binding of prostate cancer cells to hyaluronate. Cancer Res 58: 2350–2.Google Scholar
Sato, S, Miyauchi, M, Kato, M, Kitajima, S, Kitagawa, S, Hiraoka, M, Kudo, Y, Ogawa, I, Takata, T (2004) Upregulated CD44v9 expression inhibits the invasion of oral squamous cell carcinoma cells. Pathobiology 71: 171–5.CrossRefGoogle ScholarPubMed
Sato, S, Miyauchi, M, Ogawa, I, Kudo, Y, Kitagawa, S, Hiraoka, M, Takata, T (2003) Inhibition of CD44v9 upregulates the invasion ability of oral squamous cell carcinoma cells. Oral Oncol 39: 27–30.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
×