Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-r5zm4 Total loading time: 0 Render date: 2024-07-02T08:03:44.451Z Has data issue: false hasContentIssue false

52 - Discovery and Development of Drugs Targeting Tumor Invasion and Metastasis

from THERAPIES

Published online by Cambridge University Press:  05 June 2012

Rob J. Jones
Affiliation:
Cancer Research UK Beatson Laboratories, United Kingdom
Tim P. Green
Affiliation:
CSL Limited, Australia
Paul Elvin
Affiliation:
AstraZeneca, United Kingdom
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

THE ERA OF MOLECULAR TARGETED THERAPY

Efforts to discover and develop novel molecular targeted cancer treatments that commenced in many pharmaceutical companies in the early 1990s have begun to make significant contributions to patient survival. Small-molecule and antibody therapeutics against targets such as EGFR (e.g. cetuximab, erlotinib, gefitinib), erbB2 (trastuzumab, lapatinib), VEGFR (bevacizumab, sorafenib, sunitinib, pazopanib, vandetanib), Abl (imatinib, nilotinib), and mTOR (sirolimus, temsirolimus) have delivered clinical efficacy in certain disease settings. The emergence of these therapies has led to an era of treatments increasingly targeted to selected patient populations, and this has implications for the development of future novel treatments [1–3]. As outcomes improve, increasingly demanding goals are set for new drugs to achieve, both preclinically and in clinical trials.

The majority of small-molecule and antibody agents to date have focused attention on proliferation, survival, and angiogenic pathways. Although there are examples of drugs targeting c-met, TGFβ src, and FAK that have progressed to clinical trials (Table 52.1), there has been relatively little commercial activity specifically targeted against the molecules and pathways contributing to the invasive phenotype and the process of metastasis. Whereas key cell proliferation and survival pathways may ultimately have an impact on metastasis [4–6], it is also clear, from much that has been discussed elsewhere in this volume, that there are targets and pathways not currently exploited, and that disease progression could be modified by modulating the underlying mechanisms contributing to tumor invasion and metastasis.

Type
Chapter
Information
Cancer Metastasis
Biologic Basis and Therapeutics
, pp. 600 - 611
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

Harris, L et al. (2007) American Society of Clinical Oncology 2007 update of recommendations for the use of tumour markers in breast cancer. J Clin Oncol. 25: 5287.CrossRefGoogle Scholar
Müller, MC et al. (2009) Dasatinib treatment of chronic-phase myeloid leukemia: analysis of responses according to pre-existing BCR-ABL mutations. Blood. 3: 4944.CrossRefGoogle Scholar
Fong, PC et al. (2009) Inhibition of poly (ADP-ribose) polymerase in tumours from BRCA mutation carriers. New Eng J Med. 361: 123.CrossRefGoogle ScholarPubMed
Spencer, KSR et al. (2000) ErbB2 is necessary for induction of carcinoma cell invasion by erbB family receptor tyrosine kinases. J Cell Biol. 148: 385.CrossRefGoogle ScholarPubMed
Lamouille, S, Derynck, R (2007) Cell size and invasion in TGF-β–induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway. J Cell Biol. 178: 437.CrossRefGoogle ScholarPubMed
Shulka, S et al. (2007) Activation of PI3K-Akt signaling pathway promotes prostate cancer cell invasion. Int J Cancer. 121: 1424.Google Scholar
Leaf, C (2004) Why we're losing the war on cancer (and how to win it). Fortune. Mar 29: 76.Google Scholar
Spratt, JS et al. (1986) Geometry, growth rates, and duration of cancer and carcinoma in situ of the breast before detection by screening. Cancer Res. 46; 970.Google ScholarPubMed
Knurst, E (2002) Regulation of cell shape and polarity by cell-cell adhesion. Mol Membrane Biol. 19: 113.CrossRefGoogle Scholar
Wang, F et al. (2002) Phenotypic reversion or death of cancer cells by altering signaling pathways in three-dimensional contexts. J Natl Cancer Inst. 94: 1494.CrossRefGoogle ScholarPubMed
Paszek, MJ et al. (2005) Tensional homeostasis and the malignant phenotype. Cancer Cell. 8: 241.CrossRefGoogle ScholarPubMed
Onder, TT et al. (2008) Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res. 68: 3645.CrossRefGoogle ScholarPubMed
Gradval, K et al. (2007) A switch from E-cadherin to N-cadherin expression indicates epithelial to mesenchymal transition and is of strong and independent importance for the progress of prostate cancer. Clin Cancer Res. 13: 7003.Google Scholar
Aarsen, et al. (2008) Antibody-mediated blockade of integrin alpha v beta 6 inhibits tumour progression in vivo by a transforming growth factor-beta-regulated mechanism. Cancer Res. 68: 561.CrossRefGoogle ScholarPubMed
Sawada, K et al. (2008) Loss of E-cadherin promotes ovarian cancer metastasis via alpha-5 integrin, which is a therapeutic target. Cancer Res. 68: 2329.CrossRefGoogle ScholarPubMed
Giannelli, G et al. (2004) Laminin-5 offsets the efficacy of gefitinib (‘Iressa’) in hepatocellular carcinoma cells. Br J Cancer. 91: 1964.CrossRefGoogle Scholar
Morris, VL et al. (1994) Mammary carcinoma cell lines of high and low metastatic potential differ not in extravasation but in subsequent migration and growth. Cin Exp Metastasis. 12: 357.CrossRefGoogle ScholarPubMed
Hegerfeldt, Y et al. (2002) Collective cell movement in primary melanoma explants: Plasticity of cell-cell interaction, β-integrin function and migration strategies. Cancer Res. 62: 2125.Google Scholar
Sahai, E et al. (2005) Simultaneous imaging of GFP, CFP and collagen in tumors in vivo using multiphoton microscopy. BMC Biotechnol. 5: 1472.CrossRefGoogle ScholarPubMed
Goswami, S et al. (2004) Breast cancer cells isolated by chemotaxis from primary tumours show increased survival and resistance to chemotherapy. Cancer Res. 64: 7664.CrossRefGoogle ScholarPubMed
Wang, W et al. (2007) Coordinated regulation of pathways for enhanced motility and chemotaxis is conserved in rat and mouse mammary tumours. Cancer Res. 67: 3505.CrossRefGoogle Scholar
Wolf, K et al. (2003) Compensation mechanism in tumour cell migration: mesenchymal-amoeboid transition after blocking of pericellular proteolysis. J Cell Biol. 160: 267.CrossRefGoogle ScholarPubMed
Comoglio, PM, Trusolino, L (2002) Invasive growth: from development to metastasis. J Clin Invest 109: 857.CrossRefGoogle Scholar
Spencer, KSR et al. (2000) ErbB2 is necessary for induction of carcinoma cell invasion by erbB family receptor tyrosine kinases. J Cell Biol. 148: 385.CrossRefGoogle ScholarPubMed
Massagué, J (2008) TGFb in cancer. Cell. 134: 215.CrossRefGoogle Scholar
Li, Y et al. (2009) Axl as a potential therapeutic target in cancer: role of Axl in tumour growth, metastasis and angiogenesis. Oncogene. 28: 3442.CrossRefGoogle ScholarPubMed
Itoh, K et al. (1999) An essential part for Rho-associated kinase in the transcellular invasion of tumour cells. Nature Med. 5: 221.CrossRefGoogle Scholar
Nakajima, M et al. (2003) Wf-536 prevents tumour metastasis by inhibiting both tumour motility and angiogenic actions. Eur J Pharmacol. 459: 113.CrossRefGoogle ScholarPubMed
Roberts, WG et al. (2008) Antitumour activity and pharmacology of a selective focal adhesion kinase inhibitor, PF-562,271. Cancer Res. 68: 1935.CrossRefGoogle Scholar
Liu, TJ et al. (2007) Inhibition of both focal adhesion kinase and insulin-like growth factor-I receptor kinase suppresses glioma proliferation in vitro and in vivo. Mol Cancer Ther. 6: 1357.CrossRefGoogle ScholarPubMed
Green, TP et al. (2009) Preclinical anticancer activity of the potent, oral src inhibitor AZD0530. Mol Oncol. 3: 248.CrossRefGoogle ScholarPubMed
Golas, JM et al. (2005) SKI-606, a src/abl inhibitor with in vivo activity in colon tumour xenograft models. Cancer Res. 65: 5358.CrossRefGoogle Scholar
Lombardo, LJ et al. (2004) Discovery of N-(2-chloro-6-methyl-phenyl)-2-(6(4-(2-hydroxyethyl)-piperazin-1-yl)-2-methylpyrimidin-4-ylamino) thiazole-5-carboxamide (BMS-354825) a dual Src/Abl kinase inhibitor with potent antitumour activity in preclinical assays. J Med Chem. 47: 6658.CrossRefGoogle Scholar
Müller, A et al. (2001) Involvement of chemokine receptors in breast cancer metastasis. Nature. 410: 50.CrossRefGoogle ScholarPubMed
Michigami, T et al. (2001) Receptor activator of nuclear factor kappaB ligand (RANKL) is a key molecule of osteoclast formation for bone metastasis in a newly developed model of human neuroblastoma. Cancer Res. 61: 1637.Google Scholar
Granchi, S et al. (2001) Endothelin-1 production by prostate cancer cell lines is up-regulated by factors involved in cancer progression and down-regulated by androgens. Prostate. 49: 267.CrossRefGoogle ScholarPubMed
Albini, A et al. (1987) A rapid in vitro assay for quantitating the invasive potential of tumour cells. Cancer Res. 47: 3239.Google Scholar
Carragher, NO (2009) Profiling distinct mechanisms of tumour invasion for drug discovery: imaging adhesion, signalling and matrix turnover. Clin Exp Metastasis. 26: 381.CrossRefGoogle ScholarPubMed
Fidler, IJ (1990) Critical factors in the biology of human cancer metastasis: Twenty-eighth GHA Clowes memorial award lecture. Cancer Res. 50:6130.Google Scholar
Havens, AM et al. (2008) An in vivo mouse model for human prostate cancer metastasis. Neoplasia. 10: 371.CrossRefGoogle Scholar
Marangoni, E et al. (2007) A new model of patient tumour-derived breast cancer xenografts for pre-clinical assays. Clin Cancer Res. 13: 3989.CrossRefGoogle Scholar
Fu, XY et al. (1991) Models of human metastatic colon cancer in nude mice orthotopically constructed by using histologically intact patient specimens. Proc Natl Acad Sci USA. 88: 9345.CrossRefGoogle ScholarPubMed
Dangles-Marie, V et al. (2007) Establishment of human colon cancer cell lines from fresh tumours versus xenografts: Comparison of success rate and cell line features. Cancer Res. 67: 398.CrossRefGoogle ScholarPubMed
Furukawa, T et al. (1993) Differential chemosensitivity of local and metastatic human gastric cancer after orthotopic transplantation of histologically intact tumor tissue in nude mice. Int J Cancer. 54: 397.CrossRefGoogle ScholarPubMed
Man, S et al. (2007) On the development o models in mice of advanced visceral metastatic disease for anti-cancer drug testing. Cancer Metastasis Rev. 26: 737.CrossRefGoogle Scholar
Gingrich, JR et al. (1996) Metastatic prostate cancer in a transgenic mouse. Cancer Res. 56: 4096.Google Scholar
Sharpless, NE, Depinho, RA (2006) The mighty mouse: genetically engineered mouse models in cancer drug development. Nat Rev Drug Discovery. 5: 741.CrossRefGoogle ScholarPubMed
Chishima, T et al. (1997) Cancer invasion and micrometastasis visualised in live tissue by green fluorescent protein expression. Cancer Res. 57: 2042.Google Scholar
MacDonald, C et al. (1992) Intravital videomicroscopy of the chorioallantoic microcirculation: A model system for studying metastasis. Microvasc Res. 44: 185.CrossRefGoogle ScholarPubMed
Kedrin, D et al. (2008) Intravital imaging of metastatic behavior through a mammary imaging window. Nat Methods. 5:1019.CrossRefGoogle ScholarPubMed
Alexander, S et al. (2008) Dynamic imaging of cancer growth and invasion: a modified skin-fold chamber model. Histochem Cell Biol. 130: 1147.CrossRefGoogle ScholarPubMed
Jenkins, et al. (2003) Bioluminescent imaging (BLI) to improve and refine traditional murine models of tumor growth and metastasis. Clin Exp Metastasis. 20: 733.CrossRefGoogle ScholarPubMed
Therasse, P et al. (2000) New guidelines to evaluate the response to treatment in solid tumours. J Natl Cancer Inst. 92: 205.CrossRefGoogle Scholar
Eisenhauer, EA et al. (2009) New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur J Cancer. 45: 228.CrossRefGoogle Scholar
Sabeh, F et al. (2004) Tumor cell traffic through the extracellular matrix is controlled by the membrane-anchored collagenase MT1-MMP. J Cell Biol. 167: 769.CrossRefGoogle ScholarPubMed
Maquoi, E et al. (1999) Paradoxical stimulation of matrix metalloproteinase-9 expression in HT1080 cells by a broad-spectrum hydroxamate-based matrix metalloproteinase inhibitor. Ann NY Acad Sci. 878: 744.CrossRefGoogle ScholarPubMed
Krüger, A et al. (2001) Hydroxamate-type matrix metalloproteinase inhibitor Batimastat promotes liver metastasis. Cancer Res. 61: 1272.Google ScholarPubMed
McCrawley, LJ et al. (2004) A protective role for matrix metalloproteinase-3 in squamous cell carcinoma. Cancer Res. 64: 6965.CrossRefGoogle Scholar
Overall, CM, Kleifeld, O (2006) Validating matrix metalloproteinases as drug targets and anti-targets for cancer therapy. Nat Rev Cancer. 6: 227.CrossRefGoogle ScholarPubMed
Leff, RL et al. (2003) Molecular changes in human osteoarthritic cartilage after 3 weeks of oral administration of BAY 12–9566, a matrix metalloproteinase inhibitor. J Rheumatol. 30: 544.Google ScholarPubMed
Erlichman, C et al. (2001) Phase I study of the matrix metalloproteinase inhibitor, BAY 12–9566. Ann Oncol. 12: 389.CrossRefGoogle ScholarPubMed
Duivenvoorden, WC et al. (2001) Quantification of matrix metalloproteinase activity in plasma of patients enrolled in a BAY 12–9566 phase I study. Int J Cancer. 91: 857.3.0.CO;2-M>CrossRefGoogle Scholar
Levin, VA et al. (2006) Randomized, double-blind, placebo-controlled trial of marimastat in glioblastoma multiforme patients following surgery and irradiation. J Neurooncol. 78: 295.CrossRefGoogle ScholarPubMed
Rigas, JR et al. (2003) Randomized placebo-controlled trials of the matrix metalloproteinase inhibitor (MMPI), BAY12–9566 as aduvant therapy for patients with small cell and non-small cell lung cancer. ASCO Annual Meeting 2003. Proc Am Soc Clin Oncol. 22: Abstract 2525.Google Scholar
Bergers, G et al. (1999) Effects of angiogenesis inhibitors on multistage carcinogenesis in mice. Science. 284: 808.CrossRefGoogle ScholarPubMed
Kola, ILandis, J (2004) Can the pharmaceutical industry reduce attrition rates?Nat Rev Drug Discovery. 3: 711.CrossRefGoogle ScholarPubMed
Hodgson, DR et al. (2009) Biomarkes in oncology drug development. Mol Oncol. 3: 24.CrossRefGoogle Scholar
Petit, V et al. (2000) Phosphorylation of tyrosine residues 31 and 118 on paxillin regulates cell migration through an association with CRK in NBTII cells. J Cell Biol. 148: 957.CrossRefGoogle Scholar
Wang, HB et al. (2001) Focal adhesion kinase is involved in mechanosensing during fibroblast migration. Proc Natl Acad Sci USA. 98: 11295.CrossRefGoogle ScholarPubMed
Croft, DR et al. (2004) Conditional ROCK activation in vivo induces tumour cell dissemination and angiogenesis. Cancer Res. 64: 8994.CrossRefGoogle ScholarPubMed
Jones, RJ et al. (2009) Src inhibitors in early breast cancer: a methodology, feasibility and variability study. Breast Cancer Res Treat. 114: 211.CrossRefGoogle ScholarPubMed
Ma, PC et al. (2007) Downstream signalling and specific inhibition of c-MET/HGF pathway in small cell lung cancer: implications for tumour invasion. Br J Cancer. 97: 368.CrossRefGoogle ScholarPubMed
Parr, C et al. (2001) The HGF/SF-induced phosphorylation of paxillin, matrix adhesion, and invasion of prostate cancer cells were suppressed by NK4, an HGF/SF variant. Biochem Biophys Res Commun. 285: 1330.CrossRefGoogle ScholarPubMed
Lepage, M et al. (2007) Noninvasive detection of matrix metalloproteinase activity in vivo using a novel magnetic resonance imaging contrast agent with a solubility switch. Mol Imaging. 6: 393.CrossRefGoogle ScholarPubMed
Nemirovskiy, OV et al. (2007) Discovery and development of a type II collagen neopeptide (TIINE) biomarker for matrix metalloproteinase activity: from in vitro to in vivo. Anal Biochem. 361: 93.CrossRefGoogle Scholar
Riisbro, R et al. (2002) Prognostic significance of soluble urokinase plasminogen activator receptor in serum and cytosol of tumour tissue from patients with primary breast cancer. Clin Cancer Res 8: 1132.Google ScholarPubMed
Stylli, SS et al. (2008) Invadopodia: At the cutting edge of tumour invasion. J Clin Neurosci. 15: 725.CrossRefGoogle ScholarPubMed
Seals, DF et al. (2005) The adaptor protein TKs5/Fish is required for podosome formation and function, and for the protease-driven invasion of cancer cells. Cancer Cell. 7: 155.CrossRefGoogle ScholarPubMed
Blouw, B et al. (2008) A role for the podosome/invadopodia scaffold protein Tks5 in tumour growth in vivo. Eur J Cell Biol. 87: 555.CrossRefGoogle Scholar
Brockhorn, M et al. (2007) Active versus Passive mechanisms in metastasis: do cancer cells crawl into vessels, or are they pushed? Lancet Oncol. 8: 444.CrossRefGoogle Scholar
Hayes, DF et al. (2006) Circulating tumour cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin Cancer Res. 12: 4218.CrossRefGoogle ScholarPubMed
Garnero, P et al. (2000) Markers of bone turnover for the management of patients with bone metastases from prostate cancer. Br J Cancer. 82: 858.CrossRefGoogle ScholarPubMed
Finlay, IG et al. (1982) Incidence and detection of occult hepatic metastases in colorectal carcinoma. Br Med J. 284: 803.CrossRefGoogle ScholarPubMed
Manfredi, S et al. (2006) Epidemiology and management of liver metastases from colorectal cancer. Ann Surg. 244: 254.CrossRefGoogle ScholarPubMed
Avritscher, EB et al. (2006) Clinical model of lifetime cost of treating bladder cancer and associated complications. Urology. 68: 549.CrossRefGoogle ScholarPubMed
Chang, SS et al. (2005) Society of urologic oncology position statement: Redefining the management of hormone-refractory prostate carcinoma. Cancer. 103: 11.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
×