全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...
Sarcoma  2011 

The Molecular Pathogenesis of Osteosarcoma: A Review

DOI: 10.1155/2011/959248

Full-Text   Cite this paper   Add to My Lib

Abstract:

Osteosarcoma is the most common primary malignancy of bone. It arises in bone during periods of rapid growth and primarily affects adolescents and young adults. The 5-year survival rate for osteosarcoma is 60%–70%, with no significant improvements in prognosis since the advent of multiagent chemotherapy. Diagnosis, staging, and surgical management of osteosarcoma remain focused on our anatomical understanding of the disease. As our knowledge of the molecular pathogenesis of osteosarcoma expands, potential therapeutic targets are being identified. A comprehensive understanding of these mechanisms is essential if we are to improve the prognosis of patients with osteosarcoma through tumour-targeted therapies. This paper will outline the pathogenic mechanisms of osteosarcoma oncogenesis and progression and will discuss some of the more frontline translational studies performed to date in search of novel, safer, and more targeted drugs for disease management. 1. Introduction Osteosarcoma is a relatively uncommon cancer although it is the most common primary malignancy to arise from bone. While incidence is low, osteosarcoma predominately affects adolescents and young adults, and if untreated it is fatal. Despite modern treatment protocols that combine chemotherapy, surgery, and sometimes radiotherapy, the 5-year survival rate for patients diagnosed with osteosarcoma remains at 60%–70% [1]. Current treatments for osteosarcoma are associated with significant morbidity, and a period of rehabilitation may be required following surgery for osteosarcoma. Hence, there is a real need to optimise current treatment strategies and to develop novel approaches for treating osteosarcoma. Traditionally, our understanding of osteosarcoma has been largely anatomical. Osteosarcoma arises most commonly in the metaphyseal region of long bones, within the medullary cavity, and penetrates the cortex of the bone to involve the surrounding soft tissues. A pseudocapsule forms around the penetrating tumour [2]. Histologically, osteosarcoma is characterised as a highly cellular tumour composed of pleomorphic spindle-shaped cells capable of producing an osteoid matrix. Current standards for staging and surgical resection rely on this anatomical knowledge [3]. However, recent developments in molecular biology have provided insight into the molecular pathogenesis of osteosarcoma. Through the identification of tumour pathways and specific mediators of osteosarcoma progression, novel approaches for targeting osteosarcoma are being developed. This paper will review our current understanding

References

[1]  S. S. Bielack, B. Kempf-Bielack, G. Delling et al., “Prognostic factors in high-grade osteosarcoma of the extremities or trunk: an analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols,” Journal of Clinical Oncology, vol. 20, no. 3, pp. 776–790, 2002.
[2]  V. J. Vigorita, Orthopaedic Pathology, Lippincott, Williams & Wilkins, Philadelphia, Pa, USA, 2008.
[3]  S. T. Canale and J. H. Beaty, Campbell's Operative Orthopaedics, Mosby Elsvier, 2008.
[4]  S. J. Cotterill, C. M. Wright, M. S. Pearce, and A. W. Craft, “Stature of young people with malignant bone tumors,” Pediatric Blood and Cancer, vol. 42, no. 1, pp. 59–63, 2004.
[5]  K. H. Gelberg, E. F. Fitzgerald, S. A. Hwang, and R. Dubrow, “Growth and development and other risk factors for osteosarcoma in children and young adults,” International Journal of Epidemiology, vol. 26, no. 2, pp. 272–278, 1997.
[6]  N. Marina, M. Gebhardt, L. Teot, and R. Gorlick, “Biology and therapeutic advances for pediatric osteosarcoma,” Oncologist, vol. 9, no. 4, pp. 422–441, 2004.
[7]  A. Longhi, A. Pasini, A. Cicognani et al., “Height as a risk factor for osteosarcoma,” Journal of Pediatric Hematology/Oncology, vol. 27, no. 6, pp. 314–318, 2005.
[8]  A. P. Polednak, “Bone cancer among female radium dial workers. Latency periods and incidence rates by time after exposure: brief communication,” Journal of the National Cancer Institute, vol. 60, no. 1, pp. 77–82, 1978.
[9]  P. Picci, “Osteosarcoma (osteogenic sarcoma),” Orphanet Journal of Rare Diseases, vol. 2, no. 1, article 6, 2007.
[10]  A. Longhi, E. Barbieri, N. Fabbri et al., “Radiation-induced osteosarcoma arising 20 years after the treatment of Ewing's sarcoma,” Tumori, vol. 89, no. 5, pp. 569–572, 2003.
[11]  A. C. Paulino and B. Z. Fowler, “Secondary neoplasms after radiotherapy for a childhood solid tumor,” Pediatric Hematology and Oncology, vol. 22, no. 2, pp. 89–101, 2005.
[12]  A. S. Rani and S. Kumar, “Transformation of non-tumorigenic osteoblast-like human osteosarcoma cells by hexavalent chromates: alteration of morphology, induction of anchorage-independence and proteolytic function,” Carcinogenesis, vol. 13, no. 11, pp. 2021–2027, 1992.
[13]  F. R. Dutra and E. J. Largent, “Osteosarcoma induced by beryllium oxide,” American Journal of Pathology, vol. 26, no. 2, pp. 197–209, 1950.
[14]  A. Mazabraud, “Experimental production of bone sarcomas in the rabbit by a single local injection of beryllium,” Bulletin du Cancer, vol. 62, no. 1, pp. 49–58, 1975.
[15]  M. L. Tan, P. F. M. Choong, and C. R. Dass, “Osteosarcoma: conventional treatment vs. gene therapy,” Cancer Biology and Therapy, vol. 8, no. 2, pp. 106–117, 2009.
[16]  S. M. Mendoza, T. Konishi, and C. W. Miller, “Integration of SV40 in human osteosarcoma DNA,” Oncogene, vol. 17, no. 19, pp. 2457–2462, 1998.
[17]  E. A. Engels, “Cancer risk associated with receipt of vaccines contaminated with simian virus 40: epidemiologic research,” Expert Review of Vaccines, vol. 4, no. 2, pp. 197–206, 2005.
[18]  F. López-Ríos, P. B. Illei, V. Rusch, and M. Ladanyi, “Evidence against a role for SV40 infection in human mesotheliomas and high risk of false-positive PCR results owing to presence of SV40 sequences in common laboratory plasmids,” The Lancet, vol. 364, no. 9440, pp. 1157–1166, 2004.
[19]  J. J. Manfredi, J. Dong, W. J. Liu et al., “Evidence against a role for SV40 in human mesothelioma,” Cancer Research, vol. 65, no. 7, pp. 2602–2609, 2005.
[20]  A. Greenspan, G. Jundt, and W. Remagen, Differential Diagnosis in Orthopaedic Oncology, Lippincott Williams & Wilkins, Philadelphia, Pa, USA, 2007.
[21]  J. German, L. P. Crippa, and D. Bloom, “Bloom's syndrome. III. Analysis of the chromosome aberration characteristic of this disorder,” Chromosoma, vol. 48, no. 4, pp. 361–366, 1974.
[22]  K. Fukuchi, G. M. Martin, and R. J. Monnat Jr., “Mutator phenotype of Werner syndrome is characterized by extensive deletions,” Proceedings of the National Academy of Sciences of the United States of America, vol. 86, no. 15, pp. 5893–5897, 1989.
[23]  J. Smida, D. Baumhoer, M. Rosemann, et al., “Genomic alterations and allelic imbalances are strong prognostic predictors in osteosarcoma,” Clinical Cancer Research, vol. 16, no. 16, pp. 4256–4267, 2010.
[24]  H. T. Ta, C. R. Dass, P. F. M. Choong, and D. E. Dunstan, “Osteosarcoma treatment: state of the art,” Cancer and Metastasis Reviews, vol. 28, no. 1-2, pp. 247–263, 2009.
[25]  B. Alberts, A. Johnson, J. Lewis, et al., “Molecular biology of the cell,” Garland Science, 2008.
[26]  J. G. Teodoro, S. K. Evans, and M. R. Green, “Inhibition of tumor angiogenesis by p53: a new role for the guardian of the genome,” Journal of Molecular Medicine, vol. 85, no. 11, pp. 1175–1186, 2007.
[27]  E. I. Hauben, J. Arends, J. P. Vandenbroucke, C. J. van Asperen, E. van Marck, and P. C. W. Hogendoorn, “Multiple primary malignancies in osteosarcoma patients. Incidence and predictive value of osteosarcoma subtype for cancer syndromes related with osteosarcoma,” European Journal of Human Genetics, vol. 11, no. 8, pp. 611–618, 2003.
[28]  J. F. McIntyre, B. Smith-Sorensen, S. H. Friend et al., “Germline mutations of the p53 tumor suppressor gene in children with osteosarcoma,” Journal of Clinical Oncology, vol. 12, no. 5, pp. 925–930, 1994.
[29]  N. Chandar, B. Billig, J. McMaster, and J. Novak, “Inactivation of p53 gene in human and murine osteosarcoma cells,” British Journal of Cancer, vol. 65, no. 2, pp. 208–214, 1992.
[30]  C. W. Miller, A. Aslo, A. Won, M. Tan, B. Lampkin, and H. P. Koeffler, “Alterations of the p53, Rb and MDM2 genes in osteosarcoma,” Journal of Cancer Research and Clinical Oncology, vol. 122, no. 9, pp. 559–565, 1996.
[31]  H. Ganjavi, M. Gee, A. Narendran et al., “Adenovirus-mediated p53 gene therapy in osteosarcoma cell lines: sensitization to cisplatin and doxorubicin,” Cancer Gene Therapy, vol. 13, no. 4, pp. 415–419, 2006.
[32]  X. Hu, A. X. Yu, B. W. Qi, et al., “The expression and significance of IDH1 and p53 in osteosarcom,” Journal of Experimental & Clinical Cancer Research, vol. 29, p. 43, 2010.
[33]  H. R. Park, W. Won Jung, F. Bertoni et al., “Molecular analysis of p53, MDM2 and H-ras genes in low-grade central osteosarcoma,” Pathology Research and Practice, vol. 200, no. 6, pp. 439–445, 2004.
[34]  F. Lonardo, T. Ueda, A. G. Huvos, J. Healey, and M. Ladanyi, “p53 and MDM2 alterations in osteosarcomas: correlation with clinicopathologic features and proliferative rate,” Cancer, vol. 79, no. 8, pp. 1541–1547, 1997.
[35]  Y. B. Park, H. S. Kim, J. H. Oh, and S. H. Lee, “The co-expression of p53 protein and P-glycoprotein is correlated to a poor prognosis in osteosarcoma,” International Orthopaedics, vol. 24, no. 6, pp. 307–310, 2001.
[36]  A. Longhi, “Osteosarcoma in blood relatives,” Oncology Reports, vol. 8, no. 1, pp. 131–136, 2001.
[37]  M. Serena Benassi, L. Molendini, G. Gamberi et al., “Alteration of pRb/p16/cdk4 regulation in human osteosarcoma,” International Journal of Cancer, vol. 84, no. 5, pp. 489–493, 1999.
[38]  S. Heinsohn, U. Evermann, U. Zur Stadt, S. Bielack, and H. Kabisch, “Determination of the prognostic value of loss of heterozygosity at the retinoblastoma gene in osteosarcoma,” International Journal of Oncology, vol. 30, no. 5, pp. 1205–1214, 2007.
[39]  B. I. Wadayama, J. Toguchida, T. Shimizu et al., “Mutation spectrum of the retinoblastoma gene in osteosarcomas,” Cancer Research, vol. 54, no. 11, pp. 3042–3048, 1994.
[40]  O. Feugeas, N. Guriec, A. Babin-Boilletot et al., “Loss of heterozygosity of the RB gene is a poor prognostic factor in patients with osteosarcoma,” Journal of Clinical Oncology, vol. 14, no. 2, pp. 467–472, 1996.
[41]  K. Iida, T. Nobori, A. Matsumine et al., “Effect of retinoblastoma tumor suppressor gene expression on chemosensitivity of human osteosarcoma cell lines,” Oncology Reports, vol. 10, no. 6, pp. 1961–1965, 2003.
[42]  J. X. Wu, P. M. Carpenter, C. Gresens et al., “The proto-oncogene c-fos is over-expressed in the majority of human osteosarcomas,” Oncogene, vol. 5, no. 7, pp. 989–1000, 1990.
[43]  A. Franchi, A. Calzolari, and G. Zampi, “Immunohistochemical detection of c-fos and c-jun expression in osseous and cartilaginous tumours of the skeleton,” Virchows Archiv, vol. 432, no. 6, pp. 515–519, 1998.
[44]  G. Gamberi, M. S. Benassi, T. Bohling et al., “C-myc and c-fos in human osteosarcoma: prognostic value of mRNA and protein expression,” Oncology, vol. 55, no. 6, pp. 556–563, 1998.
[45]  Z. Q. Wang, J. Liang, K. Schellander, E. F. Wagner, and A. E. Grigoriadis, “c-fos-induced osteosarcoma formation in transgenic mice: cooperativity with c-jun and the role of endogenous c-fos,” Cancer Research, vol. 55, no. 24, pp. 6244–6251, 1995.
[46]  V. D. Leaner, J. F. Chick, H. Donninger et al., “Inhibition of AP-1 transcriptional activity blocks the migration, invasion, and experimental metastasis of murine osteosarcoma,” American Journal of Pathology, vol. 174, no. 1, pp. 265–275, 2009.
[47]  M. L. Tan, P. F. M. Choong, and C. R. Dass, “Direct anti-metastatic efficacy by the DNA enzyme Dz13 and downregulated MMP-2, MMP-9 and MT1-MMP in tumours,” Cancer Cell International, vol. 10, article 9, 2010.
[48]  T. Shimizu, T. Ishikawa, E. Sugihara, et al., “c-MYC overexpression with loss of Ink4a/Arf transforms bone marrow stromal cells into osteosarcoma accompanied by loss of adipogenesis,” Oncogene, vol. 29, no. 42, pp. 5687–5699, 2010.
[49]  C. M. Hattinger, G. Stoico, F. Michelacci et al., “Mechanisms of gene amplification and evidence of coamplification in drug-resistant human osteosarcoma cell lines,” Genes Chromosomes and Cancer, vol. 48, no. 4, pp. 289–309, 2009.
[50]  I. Scionti, F. Michelacci, M. Pasello et al., “Clinical impact of the methotrexate resistance-associated genes C-MYC and dihydrofolate reductase (DHFR) in high-grade osteosarcoma,” Annals of Oncology, vol. 19, no. 8, pp. 1500–1508, 2008.
[51]  X. K. Xie, D. S. Yang, Z. M. Ye, and H. M. Tao, “Enhancement effect of adenovirus-mediated antisense c-myc and caffeine on the cytotoxicity of cisplatin in osteosarcoma cell lines,” Chemotherapy, vol. 55, no. 6, pp. 433–440, 2009.
[52]  C. Arvanitis, P. K. Bendapudi, J. R. Tseng, S. S. Gambhir, and D. W. Felsher, “18F and 18FDG PET imaging of osteosarcoma to non-invasively monitor in situ changes in cellular proliferation and bone differentiation upon MYC inactivation,” Cancer Biology and Therapy, vol. 7, no. 12, pp. 1947–1951, 2008.
[53]  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.
[54]  F. Navid, J. J. Letterio, C. L. Yeung, M. Pegtel, and L. J. Helman, “Autocrine transforming growth factor-β growth pathway in murine osteosarcoma cell lines associated with inability to affect phosphorylation of retinoblastoma protein,” Sarcoma, vol. 4, no. 3, pp. 93–102, 2000.
[55]  Y. S. Hu, Y. Pan, W. H. Li, Y. Zhang, J. Li, and B. A. Ma, “Int7G24A variant of transforming growth factor-beta receptor 1 is associated with osteosarcoma susceptibility in a Chinese population,” Medical Oncology. In Press.
[56]  Y. S. Hu, Y. Pan, W. H. Li, Y. Zhang, J. Li, and B. A. Ma, “Association between TGFBR1*6A and osteosarcoma: a Chinese case-control study,” BMC Cancer, vol. 10, article 169, 2010.
[57]  B. Rikhof, S. De Jong, A. J. H. Suurmeijer, C. Meijer, and W. T. A. van der Graaf, “The insulin-like growth factor system and sarcomas,” Journal of Pathology, vol. 217, no. 4, pp. 469–482, 2009.
[58]  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.
[59]  J. Dong, S. J. Demarest, A. Sereno, et al., “Combination of two insulin-like growth factor-I receptor inhibitory antibodies targeting distinct epitopes leads to an enhanced antitumor response,” Molecular Cancer Therapeutics, vol. 9, no. 9, pp. 2593–2604, 2010.
[60]  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.
[61]  L. F. Lau and S. C. T. Lam, “The CCN family of angiogenic regulators: the integrin connection,” Experimental Cell Research, vol. 248, no. 1, pp. 44–57, 1999.
[62]  T. Nishida, T. Nakanishi, M. Asano, T. Shimo, and M. Takigawa, “Effects of CTGF/Hcs 24, a hypertrophic chondrocyte-specific gene product, on the proliferation and differentiation of osteoblastic cells in vitro,” Journal of Cellular Physiology, vol. 184, no. 2, pp. 197–206, 2000.
[63]  B. Perbal, M. Zuntini, D. Zambelli et al., “Prognostic value of CCN3 in osteosarcoma,” Clinical Cancer Research, vol. 14, no. 3, pp. 701–709, 2008.
[64]  C. P. Rodda, M. Kubota, J. A. Heath et al., “Evidence for a novel parathyroid hormone-related protein in fetal lamb parathyroid glands and sheep placenta: comparisons with a similar protein implicated in humoral hypercalcaemia of malignancy,” Journal of Endocrinology, vol. 117, no. 2, pp. 261–271, 1988.
[65]  R. Yang, B. H. Hoang, T. Kubo et al., “Over-expression of parathyroid hormone type 1 receptor confers an aggressive phenotype in osteosarcoma,” International Journal of Cancer, vol. 121, no. 5, pp. 943–954, 2007.
[66]  S. Gagiannis, M. Müller, S. Uhlemann et al., “Parathyroid hormone-related protein confers chemoresistance by blocking apoptosis signaling via death receptors and mitochondria,” International Journal of Cancer, vol. 125, no. 7, pp. 1551–1557, 2009.
[67]  A. Berdiaki, G. A. Datsis, D. Nikitovic et al., “Parathyroid hormone (PTH) peptides through the regulation of hyaluronan metabolism affect osteosarcoma cell migration,” IUBMB Life, vol. 62, no. 5, pp. 377–386, 2010.
[68]  G. M. F. Pasquini, R. A. M. Davey, P. W. M. Ho et al., “Local secretion of parathyroid hormone-related protein by an osteoblastic osteosarcoma (UMR 106-01) cell line results in growth inhibition,” Bone, vol. 31, no. 5, pp. 598–605, 2002.
[69]  M. L. Broadhead, C. R. Dass, and P. F. M. Choong, “Cancer cell apoptotic pathways mediated by PEDF: prospects for therapy,” Trends in Molecular Medicine, vol. 15, no. 10, pp. 461–467, 2009.
[70]  Y. Jan, M. Matter, J. T. Pai et al., “A mitochondrial protein, Bit1, mediates apoptosis regulated by integrins and Groucho/TLE corepressors,” Cell, vol. 116, no. 5, pp. 751–762, 2004.
[71]  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.
[72]  K. M. Nicholson and N. G. Anderson, “The protein kinase B/Akt signalling pathway in human malignancy,” Cellular Signalling, vol. 14, no. 5, pp. 381–395, 2002.
[73]  S. J. Coniglio, T. S. Jou, and M. Symons, “Rac1 protects epithelial cells against anoikis,” Journal of Biological Chemistry, vol. 276, no. 30, pp. 28113–28120, 2001.
[74]  R. Ley, K. E. Ewings, K. Hadfield, E. Howes, K. Balmanno, and S. J. Cook, “Extracellular signal-regulated kinases 1/2 are serum-stimulated “Bim kinases” that bind to the BH3-only protein bim causing its phosphorylation and turnover,” Journal of Biological Chemistry, vol. 279, no. 10, pp. 8837–8847, 2004.
[75]  D. J. Hicklin and L. M. Ellis, “Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis,” Journal of Clinical Oncology, vol. 23, no. 5, pp. 1011–1027, 2005.
[76]  H. F. Dvorak, “Angiogenesis: update 2005,” Journal of Thrombosis and Haemostasis, vol. 3, no. 8, pp. 1835–1842, 2005.
[77]  M. Shibuya and L. Claesson-Welsh, “Signal transduction by VEGF receptors in regulation of angiogenesis and lymphangiogenesis,” Experimental Cell Research, vol. 312, no. 5, pp. 549–560, 2006.
[78]  J. A. Nagy, A. M. Dvorak, and H. F. Dvorak, “VEGF-A and the induction of pathological angiogenesis,” Annual Review of Pathology, vol. 2, pp. 251–275, 2007.
[79]  T. Matsumoto and H. Mugishima, “Signal transduction via vascular endothelial growth factor (VEGF) receptors and their roles in atherogenesis,” Journal of Atherosclerosis and Thrombosis, vol. 13, no. 3, pp. 130–135, 2006.
[80]  D. Liao and R. S. Johnson, “Hypoxia: a key regulator of angiogenesis in cancer,” Cancer and Metastasis Reviews, vol. 26, no. 2, pp. 281–290, 2007.
[81]  P. Carmeliet, “VEGF as a key mediator of angiogenesis in cancer,” Oncology, vol. 69, no. 3, pp. 4–10, 2005.
[82]  J. Tran, J. Rak, C. Sheehan et al., “Marked induction of the IAP family antiapoptotic proteins survivin and XIAP by VEGF in vascular endothelial cells,” Biochemical and Biophysical Research Communications, vol. 264, no. 3, pp. 781–788, 1999.
[83]  G. D. Yancopoulos, S. Davis, N. W. Gale, J. S. Rudge, S. J. Wiegand, and J. Holash, “Vascular-specific growth factors and blood vessel formation,” Nature, vol. 407, no. 6801, pp. 242–248, 2000.
[84]  I. B. Lobov, R. A. Renard, N. Papadopoulos et al., “Delta-like ligand 4 (DII4) is induced by VEGF as a negative regulator of angiogenic sprouting,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 9, pp. 3219–3224, 2007.
[85]  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.
[86]  H. Hara, T. Akisue, T. Fujimoto et al., “Expression of VEGF and its receptors and angiogenesis in bone and soft tissue tumors,” Anticancer Research, vol. 26, no. 6B, pp. 4307–4311, 2006.
[87]  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.
[88]  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.
[89]  B. Ren, K. O. Yee, J. Lawler, and R. Khosravi-Far, “Regulation of tumor angiogenesis by thrombospondin-1,” Biochimica et Biophysica Acta, vol. 1765, no. 2, pp. 178–188, 2006.
[90]  J. Cai, C. Parr, G. Watkins, W. G. Jiang, and M. Boulton, “Decreased pigment epithelium-derived factor expression in human breast cancer progression,” Clinical Cancer Research, vol. 12, no. 11, part 1, pp. 3510–3517, 2006.
[91]  J. C. M. Clark, D. M. Thomas, P. F. M. Choong, and C. R. Dass, “RECK—a newly discovered inhibitor of metastasis with prognostic significance in multiple forms of cancer,” Cancer and Metastasis Reviews, vol. 26, no. 3-4, pp. 675–683, 2007.
[92]  G. M. Y. Quan, J. Ojaimi, Y. Li, V. Kartsogiannis, H. Zhou, and P. F. M. Choong, “Localization of pigment epithelium-derived factor in growing mouse bone,” Calcified Tissue International, vol. 76, no. 2, pp. 146–153, 2005.
[93]  M. A. Moses, D. Wiederschain, I. Wu et al., “Troponin I is present in human cartilage and inhibits angiogenesis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 96, no. 6, pp. 2645–2650, 1999.
[94]  E. T. H. Ek, C. R. Dass, K. G. Contreras, and P. F. M. Choong, “Pigment epithelium-derived factor overexpression inhibits orthotopic osteosarcoma growth, angiogenesis and metastasis,” Cancer Gene Therapy, vol. 14, no. 7, pp. 616–626, 2007.
[95]  E. T. H. Ek, C. R. Dass, K. G. Contreras, and P. F. M. Choong, “Inhibition of orthotopic osteosarcoma growth and metastasis by multitargeted antitumor activities of pigment epithelium-derived factor,” Clinical and Experimental Metastasis, vol. 24, no. 2, pp. 93–106, 2007.
[96]  B. H. Luo, C. V. Carman, and T. A. Springer, “Structural basis of integrin regulation and signaling,” Annual Review of Immunology, vol. 25, pp. 619–647, 2007.
[97]  A. L. Berrier and K. M. Yamada, “Cell-matrix adhesion,” Journal of Cellular Physiology, vol. 213, no. 3, pp. 565–573, 2007.
[98]  A. S. Nimnual, L. J. Taylor, and D. Bar-Sagi, “Redox-dependent downregulation of Rho by Rac,” Nature Cell Biology, vol. 5, no. 3, pp. 236–241, 2003.
[99]  O. Fromigué, Z. Hamidouche, and P. J. Marie, “Blockade of the RhoA-JNK-c-Jun-MMP2 cascade by atorvastatin reduces osteosarcoma cell invasion,” Journal of Biological Chemistry, vol. 283, no. 45, pp. 30549–30556, 2008.
[100]  C. M. Lo, H. B. Wang, M. Dembo, and Y. L. Wang, “Cell movement is guided by the rigidity of the substrate,” Biophysical Journal, vol. 79, no. 1, pp. 144–152, 2000.
[101]  M. J. Paszek, N. Zahir, K. R. Johnson et al., “Tensional homeostasis and the malignant phenotype,” Cancer Cell, vol. 8, no. 3, pp. 241–254, 2005.
[102]  K. W. Hunter, “Ezrin, a key component in tumor metastasis,” Trends in Molecular Medicine, vol. 10, no. 5, pp. 201–204, 2004.
[103]  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.
[104]  H. Birkedal-Hansen, W. G. I. Moore, M. K. Bodden et al., “Matrix metalloproteinases: a review,” Critical Reviews in Oral Biology and Medicine, vol. 4, no. 2, pp. 197–250, 1993.
[105]  S. Chakraborti, M. Mandal, S. Das, A. Mandal, and T. Chakraborti, “Regulation of matrix metalloproteinases. An overview,” Molecular and Cellular Biochemistry, vol. 253, no. 1-2, pp. 269–285, 2003.
[106]  J. Oh, R. Takahashi, S. Kondo et al., “The membrane-anchored MMP inhibitor RECK is a key regulator of extracellular matrix integrity and angiogenesis,” Cell, vol. 107, no. 6, pp. 789–800, 2001.
[107]  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.
[108]  M. F. Burbridge, F. Cogé, J. P. Galizzi, J. A. Boutin, D. C. West, and G. C. Tucker, “The role of the matrix metalloproteinases during in vitro vessel formation,” Angiogenesis, vol. 5, no. 3, pp. 215–226, 2002.
[109]  V. Masson, L. Rodriguez de la Ballina, C. Munaut et al., “Contribution of host MMP-2 and MMP-9 to promote tumor vascularization and invasion of malignant keratinocytes,” FASEB Journal, vol. 19, no. 2, pp. 234–236, 2005.
[110]  P. F. Choong and A. P. Nadesapillai, “Urokinase plasminogen activator system: a multifunctional role in tumor progression and metastasis,” Clinical Orthopaedics and Related Research, vol. 415, supplement, pp. S46–S58, 2003.
[111]  V. Pillay, C. R. Dass, and P. F. M. Choong, “The urokinase plasminogen activator receptor as a gene therapy target for cancer,” Trends in Biotechnology, vol. 25, no. 1, pp. 33–39, 2007.
[112]  P. F. M. Choong, M. Fern?, M. ?kermans et al., “Urokinase-plasminogen-activator levels and prognosis in 69 soft-tissue sarcomas,” International Journal of Cancer, vol. 69, no. 4, pp. 268–272, 1996.
[113]  C. R. Dass, A. P. W. Nadesapillai, D. Robin et al., “Downregulation of uPAR confirms link in growth and metastasis of osteosarcoma,” Clinical and Experimental Metastasis, vol. 22, no. 8, pp. 643–652, 2005.
[114]  T. A. Guise and J. M. Chirgwin, “Transforming growth factor-beta in osteolytic breast cancer bone metastases,” Clinical Orthopaedics and Related Research, vol. 415, supplement, pp. S32–S38, 2003.
[115]  J. M. W. Quinn, K. Itoh, N. Udagawa et al., “Transforming growth factor β affects osteoclast differentiation via direct and indirect actions,” Journal of Bone and Mineral Research, vol. 16, no. 10, pp. 1787–1794, 2001.
[116]  L. A. Kingsley, P. G. J. Fournier, J. M. Chirgwin, and T. A. Guise, “Molecular biology of bone metastasis,” Molecular Cancer Therapeutics, vol. 6, no. 10, pp. 2609–2617, 2007.
[117]  J. M. Chirgwin and T. A. Guise, “Skeletal metastases: decreasing tumor burden by targeting the bone microenvironment,” Journal of Cellular Biochemistry, vol. 102, no. 6, pp. 1333–1342, 2007.
[118]  K. Kinpara, “Osteoclast differentiation factor in human osteosarcoma cell line,” Journal of Immunoassay, vol. 21, no. 4, pp. 327–340, 2000.
[119]  L. C. Hofbauer and A. E. Heufelder, “Osteoprotegerin and its cognate ligand: a new paradigm of osteoclastogenesis,” European Journal of Endocrinology, vol. 139, no. 2, pp. 152–154, 1998.
[120]  H. Takayanagi, “The role of NFAT in osteoclast formation,” Annals of the New York Academy of Sciences, vol. 1116, pp. 227–237, 2007.
[121]  S. A. Stoch and J. A. Wagner, “Cathepsin K inhibitors: a novel target for osteoporosis therapy,” Clinical Pharmacology and Therapeutics, vol. 83, no. 1, pp. 172–176, 2008.
[122]  C. Le Gall, A. Bellahcène, E. Bonnelye et al., “A cathepsin K inhibitor reduces breast cancer-induced osteolysis and skeletal tumor burden,” Cancer Research, vol. 67, no. 20, pp. 9894–9902, 2007.
[123]  K. Husmann, R. Muff, M. E. Bolander, G. Sarkar, W. Born, and B. Fuchs, “Cathepsins and osteosarcoma: expression analysis identifies cathepsin K as an indicator of metastasis,” Molecular Carcinogenesis, vol. 47, no. 1, pp. 66–73, 2008.
[124]  S. Tanaka, M. Amling, L. Neff et al., “c-Cbl is downstream of c-Src in a signalling pathway necessary for bone resorption,” Nature, vol. 383, no. 6600, pp. 528–531, 1996.
[125]  J. Schlessinger, “New roles for Src kinases in control of cell survival and angiogenesis,” Cell, vol. 100, no. 3, pp. 293–296, 2000.
[126]  H. Glantschnig, J. E. Fisher, G. Wesolowski, et al., “M-CSF, TNFalpha and RANK ligand promote osteoclast survival by signaling through mTOR/S6 kinase,” Cell Death Differ, vol. 10, no. 10, pp. 1165–1177, 2003.
[127]  N. Rucci, M. ?u?a, and A. Teti, “Inhibition of protein kinase c-Src as a therapeutic approach for cancer and bone metastases,” Anti-Cancer Agents in Medicinal Chemistry, vol. 8, no. 3, pp. 342–349, 2008.
[128]  T. Akiyama, C. R. Dass, and P. F. M. Choong, “Novel therapeutic strategy for osteosarcoma targeting osteoclast differentiation, bone-resorbing activity, and apoptosis pathway,” Molecular Cancer Therapeutics, vol. 7, no. 11, pp. 3461–3469, 2008.
[129]  S. Tanaka, K. Nakamura, N. Takahasi, and T. Suda, “Role of RANKL in physiological and pathological bone resorption and therapeutics targeting the RANKL-RANK signaling system,” Immunological Reviews, vol. 208, pp. 30–49, 2005.
[130]  F. Lamoureux, P. Richard, Y. Wittrant, et al., “Therapeutic relevance of osteoprotegerin gene therapy in osteosarcoma: blockade of the vicious cycle between tumor cell proliferation and bone resorption,” Cancer Research, vol. 67, no. 15, pp. 7308–7318, 2007.
[131]  G. Ottaviani and N. Jaffe, “The epidemiology of osteosarcoma,” Cancer Treatment and Research, vol. 152, pp. 3–13, 2009.
[132]  T. A. Guise, R. O'Keefe, R. L. Randall, and R. M. Terek, “Molecular biology and therapeutics in musculoskeletal oncology,” Journal of Bone and Joint Surgery. American, vol. 91, no. 3, pp. 724–732, 2009.
[133]  M. L. Broadhead, T. Akiyama, P. F. M. Choong, and C. R. Dass, “The pathophysiological role of PEDF in bone diseases,” Current Molecular Medicine, vol. 10, no. 3, pp. 296–301, 2010.
[134]  E. T. H. Ek, C. R. Dass, K. G. Contreras, and P. F. M. Choong, “Inhibition of orthotopic osteosarcoma growth and metastasis by multitargeted antitumor activities of pigment epithelium-derived factor,” Clinical and Experimental Metastasis, vol. 24, no. 2, pp. 93–106, 2007.
[135]  E. T. H. Ek, C. R. Dass, K. G. Contreras, and P. F. M. Choong, “PEDF-derived synthetic peptides exhibit antitumor activity in an orthotopic model of human osteosarcoma,” Journal of Orthopaedic Research, vol. 25, no. 12, pp. 1671–1680, 2007.
[136]  H. G. Kang, H. S. Kim, K. J. Kim et al., “RECK expression in osteosarcoma: correlation with matrix metalloproteinases activation and tumor invasiveness,” Journal of Orthopaedic Research, vol. 25, no. 5, pp. 696–702, 2007.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413