Osteosarcoma is the most frequent malignant primary bone tumor characterized by a high potency to form lung metastases which is the main cause of death. Unfortunately, the conventional chemotherapy is not fully effective on osteosarcoma metastases. The progression of a primary tumor to metastasis requires multiple processes, which are neovascularization, proliferation, invasion, survival in the bloodstream, apoptosis resistance, arrest at a distant organ, and outgrowth in secondary sites. Consequently, recent studies have revealed new insights into the molecular mechanisms of metastasis development. The understanding of the mechanism of molecular alterations can provide the identification of novel therapeutic targets and/or prognostic markers for osteosarcoma treatment to improve the clinical outcome. 1. Introduction Osteosarcoma (OS) most often occurs, during childhood and adolescence, in the metaphysis of long bones, including large growth plates with high proliferation activity and bone turnover [1]. Historically, patients with primary OS have been treated with resection surgery alone, resulting in poor prognosis. Clinical outcome of localized OS has improved with neoadjuvant chemotherapies, based on methotrexate, cisplatin, doxorubicin, and ifosfamide treatments. The 5-year survival has indeed increased to around 60%. However, the 5-year survival of patients with OS metastasis still remains about 30% [2–7]. OS metastases appear most frequently in the lung [8] and are the main cause of death for patients with OS, because micrometastases are undetectable at initial diagnosis [9, 10]. Taken together, OS patients with metastases present further worse clinical results than those without metastases. Thus, more effective treatments and/or a more personalized therapy (i.e., treatments according to specific genes or protein profile expressions) are needed for patients with OS associated with pulmonary metastases. The establishment of cancer metastasis involves several complex steps: intravasation, survival in the circulation, arrest at a distant organ, extravasation, and growth in secondary sites (Figure 1). Molecular alterations of these steps have been practically analyzed. The understanding of metastasis mechanism might allow us to find new molecular targets for improvement of the patients’ survival. This paper describes the molecular factors associated with OS development and summarizes the main molecular alterations involved in this bone disease, especially in metastatic OS, which strongly contribute to the development of novel therapeutic approaches.
References
[1]
P. A. Meyers and R. Gorlick, “Osteosarcoma,” Pediatric Clinics of North America, vol. 44, no. 4, pp. 973–989, 1997.
[2]
G. Rosen, B. Caparros, and A. G. Huvos, “Preoperative chemotherapy for osteogenic sarcoma: selection of postoperative adjuvant chemotherapy based on the response of the primary tumor to preoperative chemotherapy,” Cancer, vol. 49, no. 6, pp. 1221–1230, 1982.
[3]
G. Bacci, A. Longhi, M. Cesari, M. Versari, and F. Bertoni, “Influence of local recurrence on survival in patients with extremity osteosarcoma treated with neoadjuvant chemotherapy: the experience of a single institution with 44 patients,” Cancer, vol. 106, no. 12, pp. 2701–2706, 2006.
[4]
O. S. Bruland and A. Pihl, “On the current management of osteosarcoma. A critical evaluation and a proposal for a modified treatment strategy,” European Journal of Cancer, vol. 33, no. 11, pp. 1725–1731, 1997.
[5]
A. Longhi, C. Errani, M. De Paolis, M. Mercuri, and G. Bacci, “Primary bone osteosarcoma in the pediatric age: state of the art,” Cancer Treatment Reviews, vol. 32, no. 6, pp. 423–436, 2006.
[6]
D. Heymann and F. Rédini, “Bone sarcomas: pathogenesis and new therapeutic approaches,” IBMS, vol. 8, no. 9, pp. 402–414, 2011.
[7]
D. Heymann, Ed., Bone cancer: progression and therapeutic approaches, Academic Press, 2010.
[8]
G. Bacci, S. Ferrari, A. Longhi et al., “Pattern of relapse in patients with osteosarcoma of the extremities treated with neoadjuvant chemotherapy,” European Journal of Cancer, vol. 37, no. 1, pp. 32–38, 2001.
[9]
D. Dunn and L. P. Dehner, “Metastatic osteosarcoma to lung: a clinicopathologic study of surgical biopsies and resections,” Cancer, vol. 40, no. 6, pp. 3054–3064, 1977.
[10]
W. F. Enneking, S. S. Spanier, and M. A. Goodman, “A system for the surgical staging of musculoskeletal sarcoma,” Clinical Orthopaedics and Related Research, vol. 153, pp. 106–120, 1980.
[11]
J. Folkman, “The role of angiogenesis in tumor growth,” Seminars in Cancer Biology, vol. 3, no. 2, pp. 65–71, 1992.
[12]
W. D. Thompson, K. J. Shiach, and R. A. Fraser, “Tumours acquire their vasculature by vessel incorporation, not vessel ingrowth,” Journal of Pathology, vol. 151, no. 4, pp. 323–332, 1987.
[13]
D. R. Senger, S. J. Galli, A. M. Dvorak, C. A. Perruzzi, V. Susan Harvey, and H. F. Dvorak, “Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid,” Science, vol. 219, no. 4587, pp. 983–985, 1983.
[14]
D. W. Leung, G. Cachianes, W. J. Kuang, D. V. Goeddel, and N. Ferrara, “Vascular endothelial growth factor is a secreted angiogenic mitogen,” Science, vol. 246, no. 4935, pp. 1306–1309, 1989.
[15]
S. A. Eccles and D. R. Welch, “Metastasis: recent discoveries and novel treatment strategies,” The Lancet, vol. 369, no. 9574, pp. 1742–1757, 2007.
[16]
P. S. Steeg, “Tumor metastasis: mechanistic insights and clinical challenges,” Nature Medicine, vol. 12, no. 8, pp. 895–904, 2006.
[17]
R. N. Kaplan, B. Psaila, and D. Lyden, “Bone marrow cells in the “pre-metastatic niche”: within bone and beyond,” Cancer and Metastasis Reviews, vol. 25, no. 4, pp. 521–529, 2006.
[18]
K. Dan?, P. A. Andreasen, J. Gr?ndahl-Hansen, P. Kristensen, L. S. Nielsen, and L. Skriver, “Plasminogen activators, tissue degradation, and cancer,” Advances in Cancer Research, vol. 44, pp. 139–266, 1985.
[19]
S. Georges, C. Ruiz Velasco, V. Trichet, Y. Fortun, D. Heymann, and M. Padrines, “Proteases and bone remodelling,” Cytokine and Growth Factor Reviews, vol. 20, no. 1, pp. 29–41, 2009.
[20]
G. Bergers, R. Brekken, G. McMahon et al., “Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis,” Nature Cell Biology, vol. 2, no. 10, pp. 737–744, 2000.
[21]
R. V. Iozzo, J. J. Zoeller, and A. Nystr?m, “Basement membrane proteoglycans: modulators par excellence of cancer growth and angiogenesis,” Molecules and Cells, vol. 27, no. 5, pp. 503–513, 2009.
[22]
Y. H. Lee, T. Tokunaga, Y. Oshika et al., “Cell-retained isoforms of vascular endothelial growth factor (VEGF) are correlated with poor prognosis in osteosarcoma,” European Journal of Cancer, vol. 35, no. 7, pp. 1089–1093, 1999.
[23]
M. Kaya, T. Wada, T. Akatsuka et al., “Vascular endothelial growth factor expression in untreated osteosarcoma is predictive of pulmonary metastasis and poor prognosis,” Clinical Cancer Research, vol. 6, no. 2, pp. 572–577, 2000.
[24]
A. Franchi, L. Arganini, G. Baroni et al., “Expression of transforming growth factor β isoforms in osteosarcoma variants: association of TGFβ1 with high-grade osteosarcomas,” Journal of Pathology, vol. 185, no. 3, pp. 284–289, 1998.
[25]
K. Scotlandi, P. Picci, and H. Kovar, “Targeted therapies in bone sarcomas,” Current Cancer Drug Targets, vol. 9, no. 7, pp. 843–853, 2009.
[26]
A. Abdeen, A. J. Chou, J. H. Healey et al., “Correlation between clinical outcome and growth factor pathway expression in osteogenic sarcoma,” Cancer, vol. 115, no. 22, pp. 5243–5250, 2009.
[27]
E. Mantadakis, G. Kim, J. Reisch et al., “Lack of prognostic significance of intratumoral angiogenesis in nonmetastatic osteosarcoma,” Journal of Pediatric Hematology/Oncology, vol. 23, no. 5, pp. 286–289, 2001.
[28]
D. Mikulic, I. Ili?, M. ?epuli? et al., “Tumor angiogenesis and outcome in osteosarcoma,” Pediatric Hematology and Oncology, vol. 21, no. 7, pp. 611–619, 2004.
[29]
M. Kreuter, R. Bieker, S. S. Bielaek et al., “Prognostic relevance of increased angiogenesis in osteosarcoma,” Clinical Cancer Research, vol. 10, no. 24, pp. 8531–8537, 2004.
[30]
M. S. O'Reilly, L. Holmgren, Y. Shing et al., “Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma,” Cell, vol. 79, no. 2, pp. 315–328, 1994.
[31]
M. S. O'Reilly, T. Boehm, Y. Shing et al., “Endostatin: an endogenous inhibitor of angiogenesis and tumor growth,” Cell, vol. 88, no. 2, pp. 277–285, 1997.
[32]
M. Dhanabal, R. Volk, R. Ramchandran, M. Simons, and V. P. Sukhatme, “Cloning, expression, and in vitro activity of human endostatin,” Biochemical and Biophysical Research Communications, vol. 258, no. 2, pp. 345–352, 1999.
[33]
P. Blezinger, J. Wang, M. Gondo et al., “Systemic inhibition of tumor growth and tumor metastases by intramuscular administration of the endostatin gene,” Nature Biotechnology, vol. 17, no. 4, pp. 343–348, 1999.
[34]
S. S. Yoon, H. Eto, C. M. Lin et al., “Mouse endostatin inhibits the formation of lung and liver metastases,” Cancer Research, vol. 59, no. 24, pp. 6251–6256, 1999.
[35]
Y. Yokoyama, M. Dhanabal, A. W. Griffioen, V. P. Sukhatme, and S. Ramakrishnan, “Synergy between angiostatin and endostatin: inhibition of ovarian cancer growth,” Cancer Research, vol. 60, no. 8, pp. 2190–2196, 2000.
[36]
O. Kisker, C. M. Becker, D. Prox et al., “Continuous administration of endostatin by intraperitoneally implanted osmotic pump improves the efficacy and potency of therapy in a mouse xenograft tumor model,” Cancer Research, vol. 61, no. 20, pp. 7669–7674, 2001.
[37]
W. Shi, C. Teschendorf, N. Muzyczka, and D. W. Siemann, “Adeno-associated virus-mediated gene transfer of endostatin inhibits angiogenesis and tumor growth in vivo,” Cancer Gene Therapy, vol. 9, no. 6, pp. 513–521, 2002.
[38]
A. L. Feldman, H. R. Alexander, S. M. Hewitt et al., “Effect of retroviral endostatin gene transfer on subcutaneous and intraperitoneal growth of murine tumors,” Journal of the National Cancer Institute, vol. 93, no. 13, pp. 1014–1020, 2001.
[39]
C. Mundhenke, J. P. Thomas, G. Wilding et al., “Tissue examination to monitor antiangiogenic therapy: a phase I clinical trial with endostatin,” Clinical Cancer Research, vol. 7, no. 11, pp. 3366–3374, 2001.
[40]
J. P. Eder Jr., J. G. Supko, J. W. Clark et al., “Phase I clinical trial of recombinant human endostatin administered as a short intravenous infusion repeated daily,” Journal of Clinical Oncology, vol. 20, no. 18, pp. 3772–3784, 2002.
[41]
R. S. Herbst, K. R. Hess, H. T. Tran et al., “Phase I study of recombinant human endostatin in patients with advanced solid tumors,” Journal of Clinical Oncology, vol. 20, no. 18, pp. 3792–3803, 2002.
[42]
J. P. Thomas, R. Z. Arzoomanian, D. Alberti et al., “Phase I pharmacokinetic and pharmacodynamic study of recombinant human endostatin in patients with advanced solid tumors,” Journal of Clinical Oncology, vol. 21, no. 2, pp. 223–231, 2003.
[43]
F. Deschaseaux, L. Sensébé, and D. Heymann, “Mechanisms of bone repair and regeneration,” Trends in Molecular Medicine, vol. 15, no. 9, pp. 417–429, 2009.
[44]
K. Bjornland, K. Flatmark, S. Pettersen, A. O. Aaasen, ?. Fodstad, and G. M. M?landsmo, “Matrix metalloproteinases participate in osteosarcoma invasion,” Journal of Surgical Research, vol. 127, no. 2, pp. 151–156, 2005.
[45]
M. Kansara and D. M. Thomas, “Molecular pathogenesis of osteosarcoma,” DNA and Cell Biology, vol. 26, no. 1, pp. 1–18, 2007.
[46]
D. G. Fan, J. Y. Dai, J. Tang et al., “Silencing of calpain expression reduces the metastatic potential of human osteosarcoma cells,” Cell Biology International, vol. 33, no. 12, pp. 1263–1267, 2009.
[47]
C. M. Díaz-Montero, J. N. Wygant, and B. W. McIntyre, “PI3-K/Akt-mediated anoikis resistance of human osteosarcoma cells requires Src activation,” European Journal of Cancer, vol. 42, no. 10, pp. 1491–1500, 2006.
[48]
P. Hingorani, W. Zhang, R. Gorlick, and E. A. Kolb, “Inhibition of Src phosphorylation alters metastatic potential of osteosarcoma in vitro but not in vivo,” Clinical Cancer Research, vol. 15, no. 10, pp. 3416–3422, 2009.
[49]
L. C. Kim, L. Song, and E. B. Haura, “Src kinases as therapeutic targets for cancer,” Nature Reviews Clinical Oncology, vol. 6, no. 10, pp. 587–595, 2009.
[50]
E. M. Rubin, Y. Guo, K. Tu, J. Xie, X. Zi, and B. H. Hoang, “Wnt inhibitory factor 1 decreases tumorigenesis and metastasis in osteosarcoma,” Molecular Cancer Therapeutics, vol. 9, no. 3, pp. 731–741, 2010.
[51]
Y. Guo, X. Zi, Z. Koontz et al., “Blocking Wnt/LRP5 signaling by a soluble receptor modulates the epithelial to mesenchymal transition and suppresses met and metalloproteinases in osteosarcoma Saos-2 cells,” Journal of Orthopaedic Research, vol. 25, no. 7, pp. 964–971, 2007.
[52]
F. Engin, T. Bertin, O. Ma et al., “Notch signaling contributes to the pathogenesis of human osteosarcomas,” Human Molecular Genetics, vol. 18, no. 8, pp. 1464–1470, 2009.
[53]
D. P. M. Hughes, “How the NOTCH pathway contributes to the ability of osteosarcoma cells to metastasize,” Cancer Treatment and Research, vol. 152, pp. 479–496, 2009.
[54]
M. Tanaka, T. Setoguchi, M. Hirotsu et al., “Inhibition of Notch pathway prevents osteosarcoma growth by cell cycle regulation,” British Journal of Cancer, vol. 100, no. 12, pp. 1957–1965, 2009.
[55]
P. Zhang, Y. Yang, P. A. Zweidler-McKay, and D. P. M. Hughes, “Critical role of notch signaling in osteosarcoma invasion and metastasis,” Clinical Cancer Research, vol. 14, no. 10, pp. 2962–2969, 2008.
[56]
Y. Soini, J. Satta, M. M??tt?, and H. Autio-Harmainen, “Expression of MMP2, MMP9, MT1-MMP, TIMP1, and TIMP2 mRNA in valvular lesions of the heart,” The Journal of Pathology, vol. 194, no. 2, pp. 225–231, 2001.
[57]
Y. Kato, T. Yamashita, and M. Ishikawa, “Relationship between expression of matrix metalloproteinase-2 and matrix metalloproteinase-9 and invasion ability of cervical cancer cells,” Oncology reports, vol. 9, no. 3, pp. 565–569, 2002.
[58]
P. P. H. Lee, J. J. Hwang, G. Murphy, and M. M. Ip, “Functional significance of MMP-9 in tumor necrosis factor-induced proliferation and branching morphogenesis of mammary epithelial cells,” Endocrinology, vol. 141, no. 10, pp. 3764–3773, 2000.
[59]
K. Soreide, E. A. Janssen, H. K?mer, and J. P. A. Baak, “Trypsin in colorectal cancer: molecular biological mechanisms of proliferation, invasion, and metastasis,” Journal of Pathology, vol. 209, no. 2, pp. 147–156, 2006.
[60]
Y. Y. Cheng, L. Huang, K. M. Lee, K. Li, and S. M. Kumta, “Alendronate regulates cell invasion and MMP-2 secretion in human osteosarcoma cell lines,” Pediatric Blood and Cancer, vol. 42, no. 5, pp. 410–415, 2004.
[61]
P. Heikkila, O. Teronen, M. Y. Hirn et al., “Inhibition of matrix metalloproteinase-14 in osteosarcoma cells by clodronate,” Journal of Surgical Research, vol. 111, no. 1, pp. 45–52, 2003.
[62]
Z. F. Xin, Y. K. Kim, and S. T. Jung, “Risedronate inhibits human osteosarcoma cell invasion,” Journal of Experimental and Clinical Cancer Research, vol. 28, no. 1, article 105, 2009.
[63]
D. Heymann, B. Ory, F. Gouin, J. R. Green, and F. Rédini, “Bisphosphonates: new therapeutic agents for the treatment of bone tumors,” Trends in Molecular Medicine, vol. 10, no. 7, pp. 337–343, 2004.
[64]
G. Moriceau, B. Ory, B. Gobin et al., “Therapeutic approach of primary bone tumours by bisphosphonates,” Current Pharmaceutical Design, vol. 16, no. 27, pp. 2981–2987, 2010.
[65]
H. J. Cho, T. S. Lee, J. B. Park et al., “Disulfiram suppresses invasive ability of osteosarcoma cells via the inhibition of MMP-2 and MMP-9 expression,” Journal of Biochemistry and Molecular Biology, vol. 40, no. 6, pp. 1069–1076, 2007.
[66]
C. E. Macsai, B. K. Foster, and C. J. Xian, “Roles of Wnt signalling in bone growth, remodelling, skeletal disorders and fracture repair,” Journal of Cellular Physiology, vol. 215, no. 3, pp. 578–587, 2008.
[67]
D. A. Glass, P. Bialek, J. D. Ahn et al., “Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation,” Developmental Cell, vol. 8, no. 5, pp. 751–764, 2005.
[68]
Y. Kawano and R. Kypta, “Secreted antagonists of the Wnt signalling pathway,” Journal of Cell Science, vol. 116, no. 13, pp. 2627–2634, 2003.
[69]
R. Baron and G. Rawadi, “Minireview: targeting the Wnt/β-catenin pathway to regulate bone formation in the adult skeleton,” Endocrinology, vol. 148, no. 6, pp. 2635–2643, 2007.
[70]
E. Canalis, A. Giustina, and J. P. Bilezikian, “Mechanisms of anabolic therapies for osteoporosis,” The New England Journal of Medicine, vol. 357, no. 9, pp. 850–916, 2007.
[71]
J. C. Hsieh, L. Kodjabachian, M. L. Rebbert et al., “A new secreted protein that binds to Wnt proteins and inhibits their activites,” Nature, vol. 398, no. 6726, pp. 431–436, 1999.
[72]
O. Tetsu and F. McCormick, “β-catenin regulates expression of cyclin D1 in colon carcinoma cells,” Nature, vol. 398, no. 6726, pp. 422–426, 1999.
[73]
T. C. He, A. B. Sparks, C. Rago et al., “Identification of c-MYC as a target of the APC pathway,” Science, vol. 281, no. 5382, pp. 1509–1512, 1998.
[74]
H. C. Crawford, B. M. Fingleton, L. A. Rudolph-Owen et al., “The metalloproteinase matrilysin is a target of β-catenin transactivation in intestinal tumors,” Oncogene, vol. 18, no. 18, pp. 2883–2891, 1999.
[75]
P. J. Kim, J. Plescia, H. Clevers, E. R. Fearon, and D. C. Altieri, “Survivin and molecular pathogenesis of colorectal cancer,” The Lancet, vol. 362, no. 9379, pp. 205–209, 2003.
[76]
R. C. Haydon, A. Deyrup, A. Ishikawa et al., “Cytoplasmic and/or nuclear accumulation of the β-catenin protein is a frequent event in human osteosarcoma,” International Journal of Cancer, vol. 102, no. 4, pp. 338–342, 2002.
[77]
K. Iwaya, H. Ogawa, M. Kuroda, M. Izumi, T. Ishida, and K. Mukai, “Cytoplasmic and/or nuclear staining of beta-catenin is associated with lung metastasis,” Clinical and Experimental Metastasis, vol. 20, no. 6, pp. 525–529, 2003.
[78]
K. Iwao, Y. Miyoshi, G. Nawa, H. Yoshikawa, T. Ochi, and Y. Nakamura, “Frequent β-catenin abnormalities in bone and soft-tissue tumors,” Japanese Journal of Cancer Research, vol. 90, no. 2, pp. 205–209, 1999.
[79]
B. H. Hoang, T. Kubo, J. H. Healey et al., “Expression of LDL receptor-related protein 5 (LRP5) as a novel marker for disease progression in high-grade osteosarcoma,” International Journal of Cancer, vol. 109, no. 1, pp. 106–111, 2004.
[80]
B. H. Hoang, T. Kubo, J. H. Healey et al., “Dickkopf 3 inhibits invasion and motility of Saos-2 osteosarcoma cells by modulating the Wnt-β-catenin pathway,” Cancer Research, vol. 64, no. 8, pp. 2734–2739, 2004.
[81]
P. Polakis, “Wnt signaling and cancer,” Genes and Development, vol. 14, no. 15, pp. 1837–1851, 2000.
[82]
P. C. Leow, Q. Tian, Z. Y. Ong, Z. Yang, and P. L. R. Ee, “Antitumor activity of natural compounds, curcumin and PKF118-310, as Wnt/β-catenin antagonists against human osteosarcoma cells,” Investigational New Drugs, vol. 28, no. 6, pp. 766–782, 2010.
[83]
J. Mazieres, B. He, L. You et al., “Wnt inhibitory factor-1 is silenced by promoter hypermethylation in human lung cancer,” Cancer Research, vol. 64, no. 14, pp. 4717–4720, 2004.
[84]
Y. C. Lin, L. You, Z. Xu et al., “Wnt signaling activation and WIF-1 silencing in nasopharyngeal cancer cell lines,” Biochemical and Biophysical Research Communications, vol. 341, no. 2, pp. 635–640, 2006.
[85]
L. Ai, Q. Tao, S. Zhong et al., “Inactivation of Wnt inhibitory factor-1 (WIF1) expression by epigenetic silencing is a common event in breast cancer,” Carcinogenesis, vol. 27, no. 7, pp. 1341–1348, 2006.
[86]
S. Yamashita, Y. Tsujino, K. Moriguchi, M. Tatematsu, and T. Ushijima, “Chemical genomic screening for methylation-silenced genes in gastric cancer cell lines using 5-aza- -deoxycytidine treatment and oligonucleotide microarray,” Cancer Science, vol. 97, no. 1, pp. 64–71, 2006.
[87]
S. Artavanis-Tsakonas, M. D. Rand, and R. J. Lake, “Notch signaling: cell fate control and signal integration in development,” Science, vol. 284, no. 5415, pp. 770–776, 1999.
[88]
R. Kopan and M. X. Ilagan, “The canonical notch signaling pathway: unfolding the activation mechanism,” Cell, vol. 137, no. 2, pp. 216–233, 2009.
[89]
T. Iso, L. Kedes, and Y. Hamamori, “HES and HERP families: multiple effectors of the Notch signaling pathway,” Journal of Cellular Physiology, vol. 194, no. 3, pp. 237–255, 2003.
[90]
M. C. Maa, T. H. Leu, D. J. Mccarley, R. C. Schatzman, and S. J. Parsons, “Potentiation of epidermal growth factor receptor-mediated oncogenesis by c-Src: implications for the etiology of multiple human cancers,” Proceedings of the National Academy of Sciences of the United States of America, vol. 92, no. 15, pp. 6981–6985, 1995.
[91]
S. Mori, L. Ronnstrand, K. Yokote et al., “Identification of two juxtamembrane autophosphorylation sites in the PGDF β-receptor; Involvement in the interaction with Src family tyrosine kinases,” The EMBO Journal, vol. 12, no. 6, pp. 2257–2264, 1993.
[92]
M. P. Playford and M. D. Schaller, “The interplay between Src and integrins in normal and tumor biology,” Oncogene, vol. 23, no. 48, pp. 7928–7946, 2004.
[93]
M. Talpaz, N. P. Shah, H. Kantarjian et al., “Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias,” The New England Journal of Medicine, vol. 354, no. 24, pp. 2531–2541, 2006.
[94]
F. M. Johnson, B. Saigal, M. Talpaz, and N. J. Donato, “Dasatinib (BMS-354825) tyrosine kinase inhibitor suppresses invasion and induces cell cycle arrest and apoptosis of head and neck squamous cell carcinoma and non-small cell lung cancer cells,” Clinical Cancer Research, vol. 11, no. 19, part 1, pp. 6924–6932, 2005.
[95]
M. M. Schittenhelm, S. Shiraga, A. Schroeder et al., “Dasatinib (BMS-354825), a dual SRC/ABL kinase inhibitor, inhibits the kinase activity of wild-type, juxtamembrane, and activation loop mutant KIT isoforms associated with human malignancies,” Cancer Research, vol. 66, no. 1, pp. 473–481, 2006.
[96]
A. C. Shor, E. A. Keschman, F. Y. Lee et al., “Dasatinib inhibits migration and invasion in diverse human sarcoma cell lines and induces apoptosis in bone sarcoma cells dependent on Src kinase for survival,” Cancer Research, vol. 67, no. 6, pp. 2800–2808, 2007.
[97]
L. B. Owen-Schaub, K. L. Van Golen, L. L. Hill, and J. E. Price, “Fas and Fas ligand interactions suppress melanoma lung metastasis,” Journal of Experimental Medicine, vol. 188, no. 9, pp. 1717–1723, 1998.
[98]
T. J. Sayers, A. D. Brooks, J. K. Lee et al., “Molecular mechanisms of immune-mediated lysis of murine renal cancer: differential contributions of perforin-dependent versus fas-mediated pathways in lysis by NK and T cells,” Journal of Immunology, vol. 161, no. 8, pp. 3957–3965, 1998.
[99]
J. K. Lee, T. J. Sayers, A. D. Brooks et al., “IFN-γ-dependent delay of in vivo tumor progression by Fas overexpression on murine renal cancer cells,” Journal of Immunology, vol. 164, no. 1, pp. 231–239, 2000.
[100]
N. Gordon, C. A. Arndt, D. S. Hawkins et al., “Fas expression in lung metastasis from osteosarcoma patients,” Journal of Pediatric Hematology/Oncology, vol. 27, no. 11, pp. 611–615, 2005.
[101]
L. L. Worth, E. A. Lafleur, S. F. Jia, and E. S. Kleinerman, “Fas expression inversely correlates with metastatic potential in osteosarcoma cells,” Oncology Reports, vol. 9, no. 4, pp. 823–827, 2002.
[102]
M. E. Peter, R. C. Budd, J. Desbarats et al., “The CD95 receptor: apoptosis revisited,” Cell, vol. 129, no. 3, pp. 447–450, 2007.
[103]
L. Chen, S. M. Park, A. V. Tumanov et al., “CD95 promotes tumour growth,” Nature, vol. 465, no. 7297, pp. 492–496, 2010.
[104]
A. Strasser, P. J. Jost, and S. Nagata, “The many roles of FAS receptor signaling in the immune system,” Immunity, vol. 30, no. 2, pp. 180–192, 2009.
[105]
F. H. Igney and P. H. Krammer, “Death and anti-death: tumour resistance to apoptosis,” Nature Reviews Cancer, vol. 2, no. 4, pp. 277–288, 2002.
[106]
R. Gorlick, P. Anderson, I. Andrulis et al., “Biology of ghildhood osteogenic sarcoma and potential targets for therapeutic development: meeting summary,” Clinical Cancer Research, vol. 9, no. 15, pp. 5442–5453, 2003.
[107]
N. V. Koshkina, C. Khanna, A. Mendoza, H. Guan, L. DeLauter, and E. S. Kleinerman, “Fas-negative osteosarcoma tumor cells are selected during metastasis to the lungs: the role of the fas pathwayin the metastatic process of osteosarcoma,” Molecular Cancer Research, vol. 5, no. 10, pp. 991–999, 2007.
[108]
E. A. Lafleur, S. F. Jia, L. L. Worth, Z. Zhou, L. B. Owen-Schaub, and E. S. Kleinerman, “Interleukin (IL)-12 and IL-12 gene transfer up-regulate Fas expression in human osteosarcoma and breast cancer cells,” Cancer Research, vol. 61, no. 10, pp. 4066–4071, 2001.
[109]
E. A. Lafleur, N. V. Koshkina, J. Stewart et al., “Increased Fas expression reduces the metastatic potential of human osteosarcoma cells,” Clinical Cancer Research, vol. 10, no. 23, pp. 8114–8119, 2004.
[110]
Z. Zhou, E. A. Lafleur, N. V. Koshkina, L. L. Worth, M. S. Lester, and E. S. Kleinerman, “Interleukin-12 up-regulates Fas expression in human osteosarcoma and Ewing's sarcoma cells by enhancing its promoter activity,” Molecular Cancer Research, vol. 3, no. 12, pp. 685–691, 2005.
[111]
X. Duan, S. F. Jia, N. Koshkina, and E. S. Kleinerman, “Intranasal interleukin-12 gene therapy enhanced the activity of ifosfamide against osteosarcoma lung metastases,” Cancer, vol. 106, no. 6, pp. 1382–1388, 2006.
[112]
K. Mori, K. Ando, and D. Heymann, “Liposomal muramyl tripeptide phosphatidyl ethanolamine: a safe and effective agent against osteosarcoma pulmonary metastases,” Expert Review of Anticancer Therapy, vol. 8, no. 2, pp. 151–159, 2008.
[113]
S. Lebel-Binay, A. Berger, F. Zinzindohoué et al., “Interleukin-18: biological properties and clinical implications,” European Cytokine Network, vol. 11, no. 1, pp. 15–25, 2000.
[114]
J. Golab, “Interleukin 18—Interferon γ inducing factor—a novel player in tumour immunotherapy?” Cytokine, vol. 12, no. 4, pp. 332–338, 2000.
[115]
R. Cao, J. Farnebo, M. Kurimoto, and Y. Cao, “Interleukin-18 acts as an angiogenesis and tumor suppressor,” The FASEB Journal, vol. 13, no. 15, pp. 2195–2202, 1999.
[116]
T. Ohtsuki, M. J. Micallef, K. Kohno, T. Tanimoto, M. Ikeda, and M. Kurimoto, “Interleukin 18 enhances Fas ligand expression and induces apoptosis in Fas-expressing human myelomonocytic KG-1 cells,” Anticancer Research, vol. 17, no. 5, pp. 3253–3258, 1997.
[117]
Y. Nakamura, N. Yamada, H. Ohyama et al., “Effect of interleukin-18 on metastasis of mouse osteosarcoma cells,” Cancer Immunology, Immunotherapy, vol. 55, no. 9, pp. 1151–1158, 2006.
[118]
N. Yamada, M. Hata, H. Ohyama et al., “Immunotherapy with interleukin-18 in combination with preoperative chemotherapy with ifosfamide effectively inhibits postoperative progression of pulmonary metastases in a mouse osteosarcoma model,” Tumor Biology, vol. 30, no. 4, pp. 176–184, 2009.
[119]
N. Boudreau, C. J. Sympson, Z. Werb, and M. J. Bissell, “Suppression of ICE and apoptosis in mammary epithelial cells by extracellular matrix,” Science, vol. 267, no. 5199, pp. 891–893, 1995.
[120]
J. Grossmann, K. Walther, M. Artinger, S. Kiessling, and J. Sch?lmerich, “Apoptotic signaling during initiation of detachment-induced apoptosis ("anoikis") of primary human intestinal epithelial cells,” Cell Growth and Differentiation, vol. 12, no. 3, pp. 147–155, 2001.
[121]
S. M. Frisch and R. A. Screaton, “Anoikis mechanisms,” Current Opinion in Cell Biology, vol. 13, no. 5, pp. 555–562, 2001.
[122]
C. M. Diaz-Montero and B. W. McIntyre, “Acquisition of anoikis resistance in human osteosarcoma cells,” European Journal of Cancer, vol. 39, no. 16, pp. 2395–2402, 2003.
[123]
L. A. Liotta and E. Kohn, “Anoikis: cancer and the homeless cell,” Nature, vol. 430, no. 7003, pp. 973–974, 2004.
[124]
P. Mehlen and A. Puisieux, “Metastasis: a question of life or death,” Nature Reviews Cancer, vol. 6, no. 6, pp. 449–458, 2006.
[125]
L. Trusolino, A. Bertotti, and P. M. Comoglio, “A signaling adapter function for α6β4 integrin in the control of HGF-dependent invasive growth,” Cell, vol. 107, no. 5, pp. 643–654, 2001.
[126]
S. N. Nikolopoulos, P. Blaikie, T. Yoshioka, W. Guo, and F. G. Giancotti, “Integrin β4 signaling promotes tumor angiogenesis,” Cancer Cell, vol. 6, no. 5, pp. 471–483, 2004.
[127]
W. Guo, Y. Pylayeva, A. Pepe et al., “β4 integrin amplifies ErbB2 signaling to promote mammary tumorigenesis,” Cell, vol. 126, no. 3, pp. 489–502, 2006.
[128]
X. Wan, S. Y. Kim, L. M. Guenther et al., “Beta4 integrin promotes osteosarcoma metastasis and interacts with ezrin,” Oncogene, vol. 28, no. 38, pp. 3401–3411, 2009.
[129]
P. Chiarugi and E. Giannoni, “Anoikis: a necessary death program for anchorage-dependent cells,” Biochemical Pharmacology, vol. 76, no. 11, pp. 1352–1364, 2008.
[130]
S. M. Janes and F. M. Watt, “Switch from αvβ5 to αvβ6 integrin expression protects squamous cell carcinomas from anoikis,” Journal of Cell Biology, vol. 166, no. 3, pp. 419–431, 2004.
[131]
R. V. Stan, “Structure of caveolae,” Biochimica et Biophysica Acta, vol. 1746, no. 3, pp. 334–348, 2005.
[132]
K. G. Rothberg, J. E. Heuser, W. C. Donzell, Y. S. Ying, J. R. Glenney, and R. G. W. Anderson, “Caveolin, a protein component of caveolae membrane coats,” Cell, vol. 68, no. 4, pp. 673–682, 1992.
[133]
K. R. Solomon, T. E. Danciu, L. D. Adolphson, L. E. Hecht, and P. V. Hauschka, “Caveolin-enriched membrane signaling complexes in human and murine osteoblasts,” Journal of Bone and Mineral Research, vol. 15, no. 12, pp. 2380–2390, 2000.
[134]
L. Cantiani, M. C. Manara, C. Zucchini et al., “Caveolin-1 reduces osteosarcoma metastases by inhibiting c-Src activity and met signaling,” Cancer Research, vol. 67, no. 16, pp. 7675–7685, 2007.
[135]
A. B. Al-Mehdi, K. Tozawa, A. B. Fisher, L. Shientag, A. Lee, and R. J. Muschel, “Intravascular origin of metastasis from the proliferation of endothelium-attached tumor cells: a new model for metastasis,” Nature Medicine, vol. 6, no. 1, pp. 100–102, 2000.
[136]
G. M. Jeffree, C. H. Price, and H. A. Sissons, “The metastatic patterns of osteosarcoma,” British Journal of Cancer, vol. 32, no. 1, pp. 87–107, 1975.
[137]
F. Lin, S.-E. Zheng, Z. Shen et al., “Relationships between levels of CXCR4 and VEGF and blood-borne metastasis and survival in patients with osteosarcoma,” Medical Oncology, vol. 28, no. 2, pp. 649–653, 2011.
[138]
C. Laverdiere, B. H. Hoang, R. Yang et al., “Messenger RNA expression levels of CXCR4 correlate with metastatic behavior and outcome in patients with osteosarcoma,” Clinical Cancer Research, vol. 11, no. 7, pp. 2561–2567, 2005.
[139]
C. Y. Huang, C. Y. Lee, M. Y. Chen et al., “Stromal cell-derived factor-1/CXCR4 enhanced motility of human osteosarcoma cells involves MEK1/2, ERK and NF-κB-dependent pathways,” Journal of Cellular Physiology, vol. 221, no. 1, pp. 204–212, 2009.
[140]
P. M. Murphy, “Chemokines and the molecular basis of cancer metastasis,” The New England Journal of Medicine, vol. 345, no. 11, pp. 833–835, 2001.
[141]
A. Müller, B. Homey, H. Soto et al., “Involvement of chemokine receptors in breast cancer metastasis,” Nature, vol. 410, no. 6824, pp. 50–56, 2001.
[142]
T. Murakami, W. Maki, A. R. Cardones et al., “Expression of CXC chemokine receptor-4 enhances the pulmonary metastatic potential of murine B16 melanoma cells,” Cancer Research, vol. 62, no. 24, pp. 7328–7334, 2002.
[143]
C. J. Scotton, J. L. Wilson, K. Scott et al., “Multiple actions of the chemokine CXCL12 on epithelial tumor cells in human ovarian cancer,” Cancer Research, vol. 62, no. 20, pp. 5930–5938, 2002.
[144]
E. Pradelli, B. Karimdjee-Soilihi, J. F. Michiels et al., “Antagonism of chemokine receptor CXCR3 inhibits osteosarcoma metastasis to lungs,” International Journal of Cancer, vol. 125, no. 11, pp. 2586–2594, 2009.
[145]
F. de Nigris, R. Rossiello, C. Schiano et al., “Deletion of Yin Yang 1 protein in osteosarcoma cells on cell invasion and CXCR4/angiogenesis and metastasis,” Cancer Research, vol. 68, no. 6, pp. 1797–1808, 2008.
[146]
M. M. Robledo, R. A. Bartolomé, N. Longo et al., “Expression of functional chemokine receptors CXCR3 and CXCR4 on human melanoma cells,” Journal of Biological Chemistry, vol. 276, no. 48, pp. 45098–45105, 2001.
[147]
D. Jones, R. J. Benjamin, A. Shahsafaei, and D. M. Dorfman, “The chemokine receptor CXCR3 is expressed in a subset of B-cell lymphomas and is a marker of B-cell chronic lymphocytic leukemia,” Blood, vol. 95, no. 2, pp. 627–632, 2000.
[148]
L. Goldberg-Bittman, E. Neumark, O. Sagi-Assif et al., “The expression of the chemokine receptor CXCR3 and its ligand, CXCL10, in human breast adenocarcinoma cell lines,” Immunology Letters, vol. 92, no. 1-2, pp. 171–178, 2004.
[149]
P. Mangeat, C. Roy, and M. Martin, “ERM proteins in cell adhesion and membrane dynamics,” Trends in Cell Biology, vol. 9, no. 5, pp. 187–192, 1999.
[150]
M. Hirao, N. Sato, T. Kondo et al., “Regulation mechanism of ERM (ezrin/radixin/moesin) protein/plasma membrane association: possible involvement of phosphatidylinositol turnover and rho-dependent signaling pathway,” Journal of Cell Biology, vol. 135, no. 1, pp. 37–51, 1996.
[151]
A. Gautreau, P. Poullet, D. Louvard, and M. Arpin, “Ezrin, a plasma membrane-microfilament linker, signals cell survival through the phosphatidylinositol 3-kinase/Akt pathway,” Proceedings of the National Academy of Sciences of the United States of America, vol. 96, no. 13, pp. 7300–7305, 1999.
[152]
K. W. Hunter, “Ezrin, a key component in tumor metastasis,” Trends in Molecular Medicine, vol. 10, no. 5, pp. 201–204, 2004.
[153]
S. Ilmonen, A. Vaheri, S. Asko-Seljavaara, and O. Carpen, “Ezrin in primary cutaneous melanoma,” Modern Pathology, vol. 18, no. 4, pp. 503–510, 2005.
[154]
W. H. Weng, J. ?hlén, K. ?str?m, W. O. Lui, and C. Larsson, “Prognostic impact of immunohistochemical expression of ezrin in highly malignant soft tissue sarcomas,” Clinical Cancer Research, vol. 11, no. 17, pp. 6198–6204, 2005.
[155]
C. Khanna, X. Wan, S. Bose et al., “The membrane-cytoskeleton linker ezrin is necessary for osteosarcoma metastasis,” Nature Medicine, vol. 10, no. 2, pp. 182–186, 2004.
[156]
L. Ren, S. H. Hong, J. Cassavaugh et al., “The actin-cytoskeleton linker protein ezrin is regulated during osteosarcoma metastasis by PKC,” Oncogene, vol. 28, no. 6, pp. 792–802, 2009.
[157]
C. Khanna, J. Khan, P. Nguyen et al., “Metastasis-associated differences in gene expression in a murine model of osteosarcoma,” Cancer Research, vol. 61, no. 9, pp. 3750–3759, 2001.
[158]
S. Ikeda, H. Sumii, K. Akiyama et al., “Amplification of both c-myc and c-raf-1 oncogenes in a human osteosarcoma,” Japanese Journal of Cancer Research, vol. 80, no. 1, pp. 6–9, 1989.
[159]
S. M. Wilhelm, C. Carter, L. Tang et al., “BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis,” Cancer Research, vol. 64, no. 19, pp. 7099–7109, 2004.
[160]
Y. Pignochino, G. Grignani, G. Cavalloni et al., “Sorafenib blocks tumour growth, angiogenesis and metastatic potential in preclinical models of osteosarcoma through a mechanism potentially involving the inhibition of ERK1/2, MCL-1 and ezrin pathways,” Molecular Cancer, vol. 8, article 118, 2009.
[161]
V. Hesse, G. Jahreis, H. Schambach et al., “Insulin-like growth factor I correlations to changes of the hormonal status in puberty and age,” Experimental and Clinical Endocrinology, vol. 102, no. 4, pp. 289–298, 1994.
[162]
N. Kawai, S. Kanzaki, S. Takano-Watou et al., “Serum free insulin-like growth factor I (IGF-I), total IGF-I, and IGF- binding protein-3 concentrations in normal children and children with growth hormone deficiency,” Journal of Clinical Endocrinology and Metabolism, vol. 84, no. 1, pp. 82–89, 1999.
[163]
N. Herzlieb, B. W. Gallaher, A. Berthold, R. Hille, and W. Kiess, “Insulin-like growth factor-I inhibits the progression of human U-2 OS osteosarcoma cells towards programmed cell death through interaction with the IGF-I receptor,” Cellular and Molecular Biology, vol. 46, no. 1, pp. 71–77, 2000.
[164]
C. C. Kappel, M. C. Velez-Yanguas, S. Hirschfeld, and L. J. Helman, “Human osteosarcoma cell lines are dependent on insulin-like growth factor I for in vitro growth,” Cancer Research, vol. 54, no. 10, pp. 2803–2807, 1994.
[165]
M. Pollak, A. W. Sem, M. Richard, E. Tetenes, and R. Bell, “Inhibition of metastatic behavior of murine osteosarcoma by hypophysectomy,” Journal of the National Cancer Institute, vol. 84, no. 12, pp. 966–971, 1992.
[166]
P. J. Mansky, D. J. Liewehr, S. M. Steinberg et al., “Treatment of metastatic osteosarcoma with the somatostatin analog oncoLar: significant reduction of insulin-like growth factor-1 serum levels,” Journal of Pediatric Hematology/Oncology, vol. 24, no. 6, pp. 440–446, 2002.
[167]
C. Khanna, J. Prehn, D. Hayden et al., “A randomized controlled trial of octreotide pamoate long-acting release and carboplatin versus carboplatin alone in dogs with naturally occurring osteosarcoma: evaluation of insulin-like growth factor suppression and chemotherapy,” Clinical Cancer Research, vol. 8, no. 7, pp. 2406–2412, 2002.
[168]
Y. H. Wang, Z. X. Wang, Y. Qiu et al., “Lentivirus-mediated RNAi knockdown of insulin-like growth factor-1 receptor inhibits growth, reduces invasion, and enhances radiosensitivity in human osteosarcoma cells,” Molecular and Cellular Biochemistry, vol. 327, no. 1-2, pp. 257–266, 2009.
[169]
Y. H. Wang, J. Xiong, S. F. Wang et al., “Lentivirus-mediated shRNA targeting insulin-like growth factor-1 receptor (IGF-1R) enhances chemosensitivity of osteosarcoma cells in vitro and in vivo,” Molecular and Cellular Biochemistry, vol. 341, no. 1-2, pp. 225–233, 2010.
[170]
E. A. Kolb, R. Gorlick, P. J. Houghton et al., “Initial testing (stage 1) of a monoclonal antibody (SCH 717454) against the IGF-1 receptor by the pediatric preclinical testing program,” Pediatric Blood and Cancer, vol. 50, no. 6, pp. 1190–1197, 2008.
[171]
E. A. Kolb, D. Kamara, W. Zhang et al., “R1507, a fully human monoclonal antibody targeting IGF-1R, is effective alone and in combination with rapamycin in inhibiting growth of osteosarcoma xenografts,” Pediatric Blood and Cancer, vol. 55, no. 1, pp. 67–75, 2010.
[172]
Y. H. Wang, X. D. Han, Y. Qiu, et al., “Increased expression of insulin-like growth factor-1 receptor is correlated with tumor metastasis and prognosis in patients with osteosarcoma,” Journal of Surgical Oncology, vol. 105, no. 3, pp. 235–243, 2012.
[173]
K. Pantel and R. H. Brakenhoff, “Dissecting the metastatic cascade,” Nature Reviews Cancer, vol. 4, no. 6, pp. 448–456, 2004.
[174]
J. A. Aguirre-Ghiso, “Models, mechanisms and clinical evidence for cancer dormancy,” Nature Reviews Cancer, vol. 7, no. 11, pp. 834–846, 2007.
[175]
G. N. Naumov, J. L. Townson, I. C. MacDonald et al., “Ineffectiveness of doxorubicin treatment on solitary dormant mammary carcinoma cells or late-developing metastases,” Breast Cancer Research and Treatment, vol. 82, no. 3, pp. 199–206, 2003.
[176]
L. Holmgren, M. S. O'Reilly, and J. Folkman, “Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression,” Nature Medicine, vol. 1, no. 2, pp. 149–153, 1995.
[177]
H. Wikman, R. Vessella, and K. Pantel, “Cancer micrometastasis and tumour dormancy,” APMIS, vol. 116, no. 7-8, pp. 754–770, 2008.
[178]
G. N. Naumov, E. Bender, D. Zurakowski et al., “A model of human tumor dormancy: an angiogenic switch from the nonangiogenic phenotype,” Journal of the National Cancer Institute, vol. 98, no. 5, pp. 316–325, 2006.
[179]
Y. Cao, M. S. O'Reilly, B. Marshall, E. Flynn, R. W. Ji, and J. Folkman, “Expression of angiostatin cDNA in a murine fibrosarcoma suppresses primary tumor growth and produces long-term dormancy of metastases,” Journal of Clinical Investigation, vol. 101, no. 5, pp. 1055–1063, 1998.
[180]
E. Favaro, A. Amadori, and S. Indraccolo, “Cellular interactions in the vascular niche: implications in the regulation of tumor dormancy,” APMIS, vol. 116, no. 7-8, pp. 648–659, 2008.
[181]
D. Barkan, J. E. Green, and A. F. Chambers, “Extracellular matrix: a gatekeeper in the transition from dormancy to metastatic growth,” European Journal of Cancer, vol. 46, no. 7, pp. 1181–1188, 2010.
[182]
C. M. Koebel, W. Vermi, J. B. Swann et al., “Adaptive immunity maintains occult cancer in an equilibrium state,” Nature, vol. 450, no. 7171, pp. 903–907, 2007.