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...

Application of Proteomics to Soft Tissue Sarcomas

DOI: 10.1155/2012/876401

Full-Text   Cite this paper   Add to My Lib

Abstract:

Soft tissue sarcomas are rare and account for less than 1% of all malignant cancers. Other than development of intensive therapies, the clinical outcome of patients with soft tissue sarcoma remains very poor, particularly when diagnosed at a late stage. Unique mutations have been associated with certain soft tissue sarcomas, but their etiologies remain unknown. The proteome is a functional translation of a genome, which directly regulates the malignant features of tumors. Thus, proteomics is a promising approach for investigating soft tissue sarcomas. Various proteomic approaches and clinical materials have been used to address clinical and biological issues, including biomarker development, molecular target identification, and study of disease mechanisms. Several cancer-associated proteins have been identified using conventional technologies such as 2D-PAGE, mass spectrometry, and array technology. The functional backgrounds of proteins identified were assessed extensively using in vitro experiments, thus supporting expression analysis. These observations demonstrate the applicability of proteomics to soft tissue sarcoma studies. However, the sample size in each study was insufficient to allow conclusive results. Given the low frequency of soft tissue sarcomas, multi-institutional collaborations are required to validate the results of proteomic approaches. 1. General Background of Soft Tissue Sarcomas Soft tissue sarcomas are defined as malignancies that originate from nonepithelial extraskeletal tissues, excluding the reticuloendothelial system, glia, and supporting tissue of parenchymal organs [1].Thus, soft tissue sarcomas may occur anywhere in the body including fibrous tissues, adipose tissues, muscles, vessels, and peripheral nerves. Thus, soft tissue sarcomas are a highly heterogeneous tumor group that is classified histogenetically. Clinically, soft tissue sarcomas range from curable tumors to those causing death via metastasis and recurrence. Thus, histological classification is accompanied by grading and staging information, which depends on clinical and pathological observations, to assess the degree of malignancy, predict prognosis, and evaluate possible therapies. Genetic alterations such as recurrent chromosomal translocations and genetic deregulations have been reported in soft tissue sarcomas. For example, chimeric genes have been reported in Ewing sarcoma, alveolar rhabdomyosarcoma (RMS), desmoplastic small round cell tumor, clear cell sarcoma (CCS), myxoid/round cell liposarcoma, myxoid chondrosarcoma, synovial sarcoma, alveolar soft

References

[1]  S. W. Weiss and J. R. Goldblum, Soft Tissue Tumors, Mosby, 2007.
[2]  G. D. Demetri, M. Von Mehren, C. D. Blanke et al., “Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors,” New England Journal of Medicine, vol. 347, no. 7, pp. 472–480, 2002.
[3]  R. Siegel, D. Naishadham, and A. Jemal, “Cancer statistics, 2012,” CA Cancer Journal for Clinicians, vol. 62, no. 1, pp. 10–29, 2012.
[4]  J. M. Coindre, I. Hostein, G. Maire et al., “Inflammatory malignant fibrous histiocytomas and dedifferentiated liposarcomas: histological review, genomic profile, and MDM2 and CDK4 status favour a single entity,” Journal of Pathology, vol. 203, no. 3, pp. 822–830, 2004.
[5]  A. Idbaih, J. M. Coindre, J. Derré et al., “Myxoid malignant fibrous histiocytoma and pleomorphic liposarcoma share very similar genomic imbalances,” Laboratory Investigation, vol. 85, no. 2, pp. 176–181, 2005.
[6]  T. O. Nielsen, R. B. West, S. C. Linn et al., “Molecular characterisation of soft tissue tumours: a gene expression study,” The Lancet, vol. 359, no. 9314, pp. 1301–1307, 2002.
[7]  J. Terry, T. Saito, S. Subramanian et al., “TLE1 as a diagnostic immunohistochemical marker for synovial sarcoma emerging from gene expression profiling studies,” American Journal of Surgical Pathology, vol. 31, no. 2, pp. 240–246, 2007.
[8]  M. Wachtel, T. Runge, I. Leuschner et al., “Subtype and prognostic classification of rhabdomyosarcoma by immunohistochemistry,” Journal of Clinical Oncology, vol. 24, no. 5, pp. 816–822, 2006.
[9]  R. B. West, C. L. Corless, X. Chen et al., “The novel marker, DOG1, is expressed ubiquitously in gastrointestinal stromal tumors irrespective of KIT or PDGFRA mutation status,” American Journal of Pathology, vol. 165, no. 1, pp. 107–113, 2004.
[10]  R. B. West, J. Harvell, S. C. Linn et al., “Apo D in soft tissue tumors: a novel marker for dermatofibrosarcoma protuberans,” American Journal of Surgical Pathology, vol. 28, no. 8, pp. 1063–1069, 2004.
[11]  M. Gry, R. Rimini, S. Str?mberg et al., “Correlations between RNA and protein expression profiles in 23 human cell lines,” BMC Genomics, vol. 10, article 365, 2009.
[12]  G. Chen, T. G. Gharib, C. C. Huang et al., “Discordant protein and mRNA expression in lung adenocarcinomas,” Molecular & Cellular Proteomics, vol. 1, no. 4, pp. 304–313, 2002.
[13]  B. Schwanhüusser, D. Busse, N. Li et al., “Global quantification of mammalian gene expression control,” Nature, vol. 473, no. 7347, pp. 337–342, 2011.
[14]  J. D. Tipton, J. C. Tran, A. D. Catherman, D. R. Ahlf, K. R. Durbin, and N. L. Kelleher, “Analysis of intact protein isoforms by mass spectrometry,” Journal of Biological Chemistry, vol. 286, no. 29, pp. 25451–25458, 2011.
[15]  T. Hasegawa, S. Yamamoto, T. Nojima et al., “Validity and reproducibility of histologic diagnosis and grading for adult soft-tissue sarcomas,” Human Pathology, vol. 33, no. 1, pp. 111–115, 2002.
[16]  Y. Suehara, T. Kondo, K. Fujii et al., “Proteomic signatures corresponding to histological classification and grading of soft-tissue sarcomas,” Proteomics, vol. 6, no. 15, pp. 4402–4409, 2006.
[17]  W. O. Russell, J. Cohen, and F. Enzinger, “A clinical and pathological staging system for soft tissue sarcomas,” Cancer, vol. 40, no. 4, pp. 1562–1570, 1977.
[18]  H. Hashimoto and M. Enjoji, “Liposarcoma. A clinicopathologic subtyping of 52 cases,” Acta Pathologica Japonica, vol. 32, no. 6, pp. 933–948, 1982.
[19]  D. Cervi, T. T. Yip, N. Bhattacharya et al., “Platelet-associated PF-4 as a biomarker of early tumor growth,” Blood, vol. 111, no. 3, pp. 1201–1207, 2008.
[20]  S. Taylor and J. Folkman, “Protamine is an inhibitor of angiogenesis,” Nature, vol. 297, no. 5864, pp. 307–312, 1982.
[21]  T. E. Maione, G. S. Gray, J. Petro et al., “Inhibition of angiogenesis by recombinant human platelet factor-4 and related peptides,” Science, vol. 247, no. 4938, pp. 77–79, 1990.
[22]  H. Joensuu and L. G. Kindblom, “Gastrointestinal stromal tumors—a review,” Acta Orthopaedica Scandinavica, Supplement, vol. 75, no. 311, pp. 62–71, 2004.
[23]  M. Miettinen, W. El-Rifai, L. Sobin, and J. Lasota, “Evaluation of malignancy and prognosis of gastrointestinal stromal tumors: a review,” Human Pathology, vol. 33, no. 5, pp. 478–483, 2002.
[24]  J. J. Rumessen, S. Peters, and L. Thuneberg, “Light- and electron microscopical studies of interstitial cells of Cajal and muscle cells at the submucosal border of human colon,” Laboratory Investigation, vol. 68, no. 4, pp. 481–495, 1993.
[25]  J. H. Zhong, L. Ma, L. Q. Li, H. M. Ru, and Y. N. Zhao, “Adjuvant imatinib for gastrointestinal stromal tumors: the current situation and problems,” Scandinavian Journal of Gastroenterology, vol. 46, no. 6, pp. 645–651, 2011.
[26]  R. P. DeMatteo, K. V. Ballman, C. R. Antonescu et al., “Adjuvant imatinib mesylate after resection of localised, primary gastrointestinal stromal tumour: a randomised, double-blind, placebo-controlled trial,” The Lancet, vol. 373, no. 9669, pp. 1097–1104, 2009.
[27]  J. Y. Blay, M. Von Mehren, and M. E. Blackstein, “Perspective on updated treatment guidelines for patients with gastrointestinal stromal tumors,” Cancer, vol. 116, no. 22, pp. 5126–5137, 2010.
[28]  Y. Suehara, T. Kondo, K. Seki et al., “Pfetin as a prognostic biomarker of gastrointestinal stromal tumors revealed by proteomics,” Clinical Cancer Research, vol. 14, no. 6, pp. 1707–1717, 2008.
[29]  B. L. Resendes, S. F. Kuo, N. G. Robertson et al., “Isolation from cochlea of a novel human intronless gene with predominant fetal expression,” Journal of the Association for Research in Otolaryngology, vol. 5, no. 2, pp. 185–202, 2004.
[30]  J. Schwenk, M. Metz, G. Zolles et al., “Native GABAB receptors are heteromultimers with a family of auxiliary subunits,” Nature, vol. 465, no. 7295, pp. 231–235, 2010.
[31]  K. Kikuta, M. Gotoh, T. Kanda et al., “Pfetin as a prognostic biomarker in gastrointestinal stromal tumor: novel monoclonal antibody and external validation study in multiple clinical facilities,” Japanese Journal of Clinical Oncology, vol. 40, no. 1, Article ID hyp125, pp. 60–72, 2009.
[32]  D. Kubota, H. Orita, A. Yoshida et al., “Pfetin as a prognostic biomarker for gastrointestinal stromal tumor: validation study in multiple clinical facilities,” Japanese Journal of Clinical Oncology, vol. 41, no. 10, Article ID hyr121, pp. 1194–1202, 2011.
[33]  T. Kondo and S. Hirohashi, “Application of highly sensitive fluorescent dyes (CyDye DIGE Fluor saturation dyes) to laser microdissection and two-dimensional difference gel electrophoresis (2D-DIGE) for cancer proteomics,” Nature protocols, vol. 1, no. 6, pp. 2940–2956, 2007.
[34]  K. Kikuta, D. Kubota, T. Saito et al., “Clinical proteomics identified ATP-dependent RNA helicase DDX39 as a novel biomarker to predict poor prognosis of patients with gastrointestinal stromal tumor,” Journal of Proteomics, vol. 75, no. 4, pp. 1089–1098, 2012.
[35]  T. Sugiura, K. Sakurai, and Y. Nagano, “Intracellular characterization of DDX39, a novel growth-associated RNA helicase,” Experimental Cell Research, vol. 313, no. 4, pp. 782–790, 2007.
[36]  T. Sugiura, Y. Nagano, and Y. Noguchi, “DDX39, upregulated in lung squamous cell cancer, displays RNA helicase activities and promotes cancer cell growth,” Cancer Biology and Therapy, vol. 6, no. 6, pp. 957–964, 2007.
[37]  H. H. Yoo and I. K. Chung, “Requirement of DDX39 DEAD box RNA helicase for genome integrity and telomere protection,” Aging Cell, vol. 10, no. 4, pp. 557–571, 2011.
[38]  T. S. Emory, L. H. Sobin, L. Lukes, D. H. Lee, and T. J. O'Leary, “Prognosis of gastrointestinal smooth-muscle (stromal) tumors: dependence on anatomic site,” American Journal of Surgical Pathology, vol. 23, no. 1, pp. 82–87, 1999.
[39]  C. D. M. Fletcher, J. J. Berman, C. Corless et al., “Diagnosis of gastrointestinal stromal tumors: a consensus approach,” Human Pathology, vol. 33, no. 5, pp. 459–465, 2002.
[40]  Y. Suehara, K. Kikuta, R. Nakayama et al., “Anatomic site-specific proteomic signatures of gastrointestinal stromal tumors,” Proteomics, vol. 3, no. 5, pp. 584–596, 2009.
[41]  H. Hashimoto, Y. Daimaru, M. Tsuneyoshi, and M. Enjoji, “Leiomyosarcoma of the external soft tissues. A clinicopathologic, immunohistochemical, and electron microscopic study,” Cancer, vol. 57, no. 10, pp. 2077–2088, 1986.
[42]  J. Yang, J. A. Eddy, Y. Pan et al., “Integrated proteomics and genomics analysis reveals a novel mesenchymal to epithelial reverting transition in leiomyosarcoma through regulation of slug,” Molecular and Cellular Proteomics, vol. 9, no. 11, pp. 2405–2413, 2010.
[43]  S. C. Abraham, “Distinguishing gastrointestinal stromal tumors from their mimics: an update,” Advances in Anatomic Pathology, vol. 14, no. 3, pp. 178–188, 2007.
[44]  A. Wells, C. Yates, and C. R. Shepard, “E-cadherin as an indicator of mesenchymal to epithelial reverting transitions during the metastatic seeding of disseminated carcinomas,” Clinical and Experimental Metastasis, vol. 25, no. 6, pp. 621–628, 2008.
[45]  S. Dhingra, M. E. Rodriguez, Q. Shen et al., “Constitutive activation with overexpression of the mTORC2-phospholipase D1 pathway in uterine leiomyosarcoma and STUMP: morphoproteomic analysis with therapeutic implications,” International Journal of Clinical and Experimental Pathology, vol. 4, no. 2, pp. 134–146, 2011.
[46]  B. Edris, I. Espinosa, T. Mühlenberg et al., “ROR2 is a novel prognostic biomarker and a potential therapeutic target in leiomyosarcoma and gastrointestinal stromal tumour,” Journal of Pathology, vol. 227, no. 2, pp. 223–233, 2012.
[47]  A. H. Beck, C. H. Lee, D. M. Witten et al., “Discovery of molecular subtypes in leiomyosarcoma through integrative molecular profiling,” Oncogene, vol. 29, no. 6, pp. 845–854, 2010.
[48]  R. Dagher and L. Helman, “Rhabdomyosarcoma: an overview,” Oncologist, vol. 4, no. 1, pp. 34–44, 1999.
[49]  G. Merlino and L. J. Helman, “Rhabdomyosarcoma—working out the pathways,” Oncogene, vol. 18, no. 38, pp. 5340–5348, 1999.
[50]  S. L. Spunt, L. M. Smith, F. B. Ruymann et al., “Cyclophosphamide dose intensification during induction therapy for intermediate-risk pediatric rhabdomyosarcoma is feasible but does not improve outcome: a report from the soft tissue sarcoma committee of the children's oncology group,” Clinical Cancer Research, vol. 10, no. 18 I, pp. 6072–6079, 2004.
[51]  W. M. Crist, J. R. Anderson, J. L. Meza et al., “Intergroup Rhabdomyosarcoma Study-IV: results for patients with nonmetastatic disease,” Journal of Clinical Oncology, vol. 19, no. 12, pp. 3091–3102, 2001.
[52]  J. G. Pressey, C. S. Pressey, G. Robinson et al., “2D-difference gel electrophoretic proteomic analysis of a cell culture model of alveolar rhabdomyosarcoma,” Journal of Proteome Research, vol. 10, no. 2, pp. 624–636, 2011.
[53]  E. F. Petricoin, V. Espina, R. P. Araujo et al., “Phosphoprotein pathway mapping: Akt/mammalian target of rapamycin activation is negatively associated with childhood rhabdomyosarcoma survival,” Cancer Research, vol. 67, no. 7, pp. 3431–3440, 2007.
[54]  C. A. Clifford, A. J. Mackin, and C. J. Henry, “Treatment of canine hemangiosarcoma: 2000 and beyond,” Journal of Veterinary Internal Medicine, vol. 14, no. 5, pp. 479–485, 2000.
[55]  A. N. Smith, “Hemangiosarcoma in dogs and cats,” Veterinary Clinics of North America, vol. 33, no. 3, pp. 533–552, 2003.
[56]  P. C. Schultheiss, “A retrospective study of visceral and nonvisceral hemangiosarcoma and hemangiomas in domestic animals,” Journal of Veterinary Diagnostic Investigation, vol. 16, no. 6, pp. 522–526, 2004.
[57]  G. M. Kirby, A. Mackay, A. Grant et al., “Concentration of lipocalin region of collagen XXVII Alpha 1 in the serum of dogs with hemangiosarcoma,” Journal of Veterinary Internal Medicine, vol. 25, no. 3, pp. 497–503, 2011.
[58]  R. J. Young, N. J. Brown, M. W. Reed, D. Hughes, and P. J. Woll, “Angiosarcoma,” The Lancet Oncology, vol. 11, no. 10, pp. 983–991, 2010.
[59]  L. Lynen, M. Zolfo, V. Huyst et al., “Management of Kaposi's sarcoma in resource-limited settings in the era of HAART,” AIDS Reviews, vol. 7, no. 1, pp. 13–21, 2005.
[60]  R. A. Schwartz, “Kaposi's sarcoma: an update,” Journal of Surgical Oncology, vol. 87, no. 3, pp. 146–151, 2004.
[61]  T. Kang, F. C. Ye, S. J. Gao, and L. D. Wang, “Angiogenesis, Kaposi's sarcoma and Kaposi's sarcoma-associated herpesvirus,” Virologica Sinica, vol. 23, no. 6, pp. 449–458, 2008.
[62]  F. X. Zhu, J. M. Chong, L. Wu, and Y. Yuan, “Virion proteins of Kaposi's sarcoma-associated herpesvirus,” Journal of Virology, vol. 79, no. 2, pp. 800–811, 2005.
[63]  J. E. Jong, J. Park, S. Kim, and T. Seo, “Kaposi's sarcoma-associated herpesvirus viral protein kinase interacts with RNA helicase a and regulates host gene expression,” Journal of Microbiology, vol. 48, no. 2, pp. 206–212, 2010.
[64]  R. Kaul, S. C. Verma, and E. S. Robertson, “Protein complexes associated with the Kaposi's sarcoma-associated herpesvirus-encoded LANA,” Virology, vol. 364, no. 2, pp. 317–329, 2007.
[65]  E. Bartee, A. McCormack, and K. Früh, “Quantitative membrane proteomics reveals new cellular targets of viral immune modulators,” PLoS pathogens, vol. 2, no. 10, p. e107, 2006.
[66]  S. E. Ong, B. Blagoev, I. Kratchmarova et al., “Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics,” Molecular & Cellular Proteomics, vol. 1, no. 5, pp. 376–386, 2002.
[67]  P. Uetz, Y. A. Dong, C. Zeretzke et al., “Herpesviral protein networks and their interaction with the human proteome,” Science, vol. 311, no. 5758, pp. 239–242, 2006.
[68]  R. Rozen, N. Sathish, Y. Li, and Y. Yuan, “Virion-wide protein interactions of Kaposi's sarcoma-associated herpesvirus,” Journal of Virology, vol. 82, no. 10, pp. 4742–4750, 2008.
[69]  D. Zheng, J. Wan, Y. G. Cho et al., “Comparison of humoral immune responses to epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus using a viral proteome microarray,” Journal of Infectious Diseases, vol. 204, no. 11, pp. 1683–1691, 2011.
[70]  A. N. Husain, T. V. Colby, N. G. Ordó?ez et al., “Guidelines for pathologic diagnosis of malignant mesothelioma: a consensus statement from the International Mesothelioma Interest Group,” Archives of Pathology and Laboratory Medicine, vol. 133, no. 8, pp. 1317–1331, 2009.
[71]  D. Martino and H. I. Pass, “Integration of multimodality approaches in the management of malignant pleural mesothelioma,” Clinical Lung Cancer, vol. 5, no. 5, pp. 290–298, 2004.
[72]  P. Ellis, A. M. Davies, W. K. Evans, A. E. Haynes, and N. S. Lloyd, “The use of chemotherapy in patients with advanced malignant pleural mesothelioma: a systematic review and practice guideline,” Journal of Thoracic Oncology, vol. 1, no. 6, pp. 591–601, 2006.
[73]  J. M. Hillegass, A. Shukla, M. B. MacPherson, J. P. Bond, C. Steele, and B. T. Mossman, “Utilization of gene profiling and proteomics to determine mineral pathogenicity in a human mesothelial cell line (LP9/TERT-1),” Journal of Toxicology and Environmental Health A, vol. 73, no. 5-6, pp. 423–436, 2010.
[74]  F. Roberts, C. M. Harper, I. Downie, and R. A. Burnett, “Immunohistochemical analysis still has a limited role in the diagnosis of malignant mesothelioma: a study of thirteen antibodies,” American Journal of Clinical Pathology, vol. 116, no. 2, pp. 253–262, 2001.
[75]  F. Roberts, A. E. McCall, and R. A. Burnett, “Malignant mesothelioma: a comparison of biopsy and postmortem material by light microscopy and immunohistochemistry,” Journal of Clinical Pathology, vol. 54, no. 10, pp. 766–770, 2001.
[76]  M. Hosako, T. Muto, Y. Nakamura et al., “Proteomic study of malignant pleural mesothelioma by laser microdissection and two-dimensional difference gel electrophoresis identified cathepsin D as a novel candidate for a differential diagnosis biomarker,” Journal of Proteomics, vol. 75, no. 3, pp. 833–844, 2012.
[77]  A. Ziegler, F. Cerciello, C. Bigosch et al., “Proteomic surfaceome analysis of mesothelioma,” Lung Cancer, vol. 75, no. 2, pp. 189–196, 2011.
[78]  J. P. J. J. Hegmans, J. D. Veltman, E. T. Fung et al., “Protein profiling of pleural effusions to identify malignant pleural mesothelioma using SELDI-TOF MS,” Technology in Cancer Research and Treatment, vol. 8, no. 5, pp. 323–332, 2009.
[79]  J. P. J. J. Hegmans, A. Hemmes, H. Hammad, L. Boon, H. C. Hoogsteden, and B. N. Lambrecht, “Mesothelioma environment comprises cytokines and T-regulatory cells that suppress immune responses,” European Respiratory Journal, vol. 27, no. 6, pp. 1086–1095, 2006.
[80]  F. Pagès, J. Galon, M. C. Dieu-Nosjean, E. Tartour, C. Sautès-Fridman, and W. H. Fridman, “Immune infiltration in human tumors: a prognostic factor that should not be ignored,” Oncogene, vol. 29, no. 8, pp. 1093–1102, 2010.
[81]  C. Théry, L. Zitvogel, and S. Amigorena, “Exosomes: composition, biogenesis and function,” Nature Reviews Immunology, vol. 2, no. 8, pp. 569–579, 2002.
[82]  J. Wolfers, A. Lozier, G. Raposo et al., “Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming,” Nature Medicine, vol. 7, no. 3, pp. 297–303, 2001.
[83]  M. P. Bard, J. P. Hegmans, A. Hemmes et al., “Proteomic analysis of exosomes isolated from human malignant pleural effusions,” American Journal of Respiratory Cell and Molecular Biology, vol. 31, no. 1, pp. 114–121, 2004.
[84]  J. P. J. J. Hegmans, M. P. L. Bard, A. Hemmes et al., “Proteomic Analysis of Exosomes Secreted by Human Mesothelioma Cells,” American Journal of Pathology, vol. 164, no. 5, pp. 1807–1815, 2004.
[85]  D. T. Archimandriti, Y. A. Dalavanga, R. Cianti et al., “Proteome analysis of bronchoalveolar lavage in individuals from metsovo, nonoccupationally exposed to asbestos,” Journal of Proteome Research, vol. 8, no. 2, pp. 860–869, 2009.
[86]  S. H. Constantopoulos, J. A. Goudevenos, and N. Saratzis, “Metsovo lung: pleural calcification and restrictive lung function in northwestern Greece. Environmental exposure to mineral fiber as etiology,” Environmental Research, vol. 38, no. 2, pp. 319–331, 1985.
[87]  A. M. Langer, R. P. Nolan, S. H. Constantopoulos, and H. M. Moutsopoulos, “Association of metsovo lung and pleural mesothelioma with exposure to tremolite-containing whitewash,” The Lancet, vol. 1, no. 8539, pp. 965–967, 1987.
[88]  C. W. Menges, Y. Chen, B. T. Mossman, J. Chernoff, A. T. Yeung, and J. R. Testa, “A phosphotyrosine proteomic screen identifies multiple tyrosine kinase signaling pathways aberrantly activated in malignant mesothelioma,” Genes and Cancer, vol. 1, no. 5, pp. 493–505, 2010.
[89]  L. L. Garland, C. Rankin, D. R. Gandara et al., “Phase II study of erlotinib in patients with malignant pleural mesothelioma: a Southwest Oncology Group study,” Journal of Clinical Oncology, vol. 25, no. 17, pp. 2406–2413, 2007.
[90]  Y. Kuramitsu, H. Miyamoto, T. Tanaka et al., “Proteomic differential display analysis identified upregulated astrocytic phosphoprotein PEA-15 in human malignant pleural mesothelioma cell lines,” Proteomics, vol. 9, no. 22, pp. 5078–5089, 2009.
[91]  W. Amin, A. V. Parwani, L. Schmandt et al., “National Mesothelioma Virtual Bank: a standard based biospecimen and clinical data resource to enhance translational research,” BMC Cancer, vol. 8, article 236, 2008.
[92]  S. K. Mohanty, A. T. Mistry, W. Amin et al., “The development and deployment of Common Data Elements for tissue banks for translational research in cancer—an emerging standard based approach for the Mesothelioma Virtual Tissue Bank,” BMC Cancer, vol. 8, article 91, 2008.
[93]  D. C. Adamson, T. J. Cummings, and A. H. Friedman, “Malignant peripheral nerve sheath tumor of the spine after radiation therapy for Hodgkin's lymphoma,” Clinical Neuropathology, vol. 23, no. 5, pp. 245–255, 2004.
[94]  A. N. D'Agostino, E. H. Soule, and R. H. Miller, “Sarcomas of the peripheral nerves and somatic soft tissues associated with multiple neurofibromatosis (Von Recklinghausen's Disease),” Cancer, vol. 16, pp. 1015–1027, 1963.
[95]  A. A. King, M. R. Debaun, V. M. Riccardi, and D. H. Gutmann, “Malignant peripheral nerve sheath tumors in neurofibromatosis 1,” American Journal of Medical Genetics, vol. 93, no. 5, pp. 388–392, 2000.
[96]  B. S. Ducatman, B. W. Scheithauer, and D. G. Piepgras, “Malignant peripheral nerve sheath tumors. A clinicopathologic study of 120 cases,” Cancer, vol. 57, no. 10, pp. 2006–2021, 1986.
[97]  E. Legius, D. A. Marchuk, F. S. Collins, and T. W. Glover, “Somatic deletion of the neurofibromatosis type 1 gene in a neurofibrosarcoma supports a tumour suppressor gene hypothesis,” Nature Genetics, vol. 3, no. 2, pp. 122–126, 1993.
[98]  B. Dasgupta, Y. Yi, D. Y. Chen, J. D. Weber, and D. H. Gutmann, “Proteomic analysis reveals hyperactivation of the mammalian target of rapamycin pathway in neurofibromatosis 1-associated human and mouse brain tumors,” Cancer Research, vol. 65, no. 7, pp. 2755–2760, 2005.
[99]  S. Patrakitkomjorn, D. Kobayashi, T. Morikawa et al., “Neurofibromatosis type 1 (NF1) tumor suppressor, neurofibromin, regulates the neuronal differentiation of PC12 cells via its associating protein, CRMP-2,” Journal of Biological Chemistry, vol. 283, no. 14, pp. 9399–9413, 2008.
[100]  G. M. Brodeur, “Neuroblastoma: biological insights into a clinical enigma,” Nature Reviews Cancer, vol. 3, no. 3, pp. 203–216, 2003.
[101]  Q. R. Chen, Y. K. Song, L. R. Yu et al., “Global genomic and proteomic analysis identifies biological pathways related to high-risk neuroblastoma,” Journal of Proteome Research, vol. 9, no. 1, pp. 373–382, 2010.
[102]  F. Yu, X. Zhu, C. Feng et al., “Proteomics-based identification of spontaneous regression-associated proteins in neuroblastoma,” Journal of Pediatric Surgery, vol. 46, no. 10, pp. 1948–1955, 2011.
[103]  T. Simon, B. Hero, D. H. Hunneman, and F. Berthold, “Tumour markers are poor predictors for relapse or progression in neuroblastoma,” European Journal of Cancer, vol. 39, no. 13, pp. 1899–1903, 2003.
[104]  W. M. Hsu, H. Lee, H. F. Juan et al., “Identification of GRP75 as an independent favorable prognostic marker of neuroblastoma by a proteomics analysis,” Clinical Cancer Research, vol. 14, no. 19, pp. 6237–6245, 2008.
[105]  C. Zanini, F. Pulerà, F. Carta et al., “Proteomic identification of heat shock protein 27 as a differentiation and prognostic marker in neuroblastoma but not in Ewing's sarcoma,” Virchows Archiv, vol. 452, no. 2, pp. 157–167, 2008.
[106]  Q. Y. He, R. Zhu, Y. Ren, P. K. H. Tam, and J. F. Chiu, “Serological protein profiling of neuroblastoma by ProteinChip SELDI-TOF technology,” Journal of Cellular Biochemistry, vol. 95, no. 1, pp. 165–172, 2005.
[107]  J. A. Sandoval, D. J. Hoelz, H. A. Woodruff et al., “Novel peptides secreted from human neuroblastoma: useful clinical tools?” Journal of Pediatric Surgery, vol. 41, no. 1, pp. 245–251, 2006.
[108]  J. A. Sandoval, L. E. Dobrolecki, J. Huang, J. L. Grosfeld, R. J. Hickey, and L. H. Malkas, “Neuroblastoma detection using serum proteomic profiling: a novel mining technique for cancer?” Journal of Pediatric Surgery, vol. 41, no. 4, pp. 639–646, 2006.
[109]  J. A. Sandoval, K. E. Turner, D. J. Hoelz, F. J. Rescorla, R. J. Hickey, and L. H. Malkas, “Serum protein profiling to identify high-risk neuroblastoma: preclinical relevance of blood-based biomarkers,” Journal of Surgical Research, vol. 142, no. 2, pp. 268–274, 2007.
[110]  R. A. Egler, Y. Li, T. A. T. Dang et al., “An integrated proteomic approach to identifying circulating biomarkers in high-risk neuroblastoma and their potential in relapse monitoring,” Proteomics, vol. 5, no. 9-10, pp. 532–541, 2011.
[111]  A. Urban, J. Poland, S. Bernardini et al., “A proteomic investigation into etoposide chemo-resistance of neuroblastoma cell lines,” Proteomics, vol. 5, no. 3, pp. 796–804, 2005.
[112]  E. Izbicka, D. Campos, J. Marty, G. Carrizales, G. Mangold, and A. Tolcher, “Molecular determinants of differential sensitivity to docetaxel and paclitaxel in human pediatric cancer models,” Anticancer Research, vol. 26, no. 3 A, pp. 1983–1988, 2006.
[113]  S. D'Aguanno, A. D'Alessandro, L. Pieroni et al., “New insights into neuroblastoma cisplatin resistance: a comparative proteomic and meta-mining investigation,” Journal of Proteome Research, vol. 10, no. 2, pp. 416–428, 2011.
[114]  X. J. Wang, Z. Sun, N. F. Villeneuve et al., “Nrf2 enhances resistance of cancer cells to chemotherapeutic drugs, the dark side of Nrf2,” Carcinogenesis, vol. 29, no. 6, pp. 1235–1243, 2008.
[115]  V. H. Bull, K. Rajalingam, and B. Thiede, “Sorafenib-induced mitochondrial complex i inactivation and cell death in human neuroblastoma cells,” Journal of Proteome Research, vol. 11, no. 3, pp. 1609–1620, 2012.
[116]  C. Vishal, J. U. Kumar, C. V. B. Swamy et al., “Repercussion of mitochondria deformity induced by anti-Hsp90 drug 17AAG in human tumor cells,” Drug Target Insights, vol. 2011, no. 5, pp. 11–32, 2011.
[117]  K. Motani, K. Tabata, Y. Kimura et al., “Proteomic analysis of apoptosis induced by xanthoangelol, a major constituent of Angelica keiskei, in neuroblastoma,” Biological and Pharmaceutical Bulletin, vol. 31, no. 4, pp. 618–626, 2008.
[118]  T. Hayama, K. Tabata, T. Uchiyama, Y. Fujimoto, and T. Suzuki, “Ferrearin C induces apoptosis via heme oxygenase-1 (HO-1) induction in neuroblastoma,” Journal of Natural Medicines, vol. 65, no. 3-4, pp. 431–439, 2011.
[119]  F. Cimmino, D. Spano, M. Capasso et al., “Comparative proteomic expression profile in all-trans retinoic acid differentiated neuroblastoma cell line,” Journal of Proteome Research, vol. 6, no. 7, pp. 2550–2564, 2007.
[120]  J. A. Sandoval, A. C. Eppstein, D. J. Hoelz et al., “Proteomic analysis of neuroblastoma subtypes in response to mitogen-activated protein kinase inhibition: profiling multiple targets of cancer kinase signaling,” Journal of Surgical Research, vol. 134, no. 1, pp. 61–67, 2006.
[121]  Q. Fang, K. Kani, V. M. Faca et al., “Impact of protein stability, cellular localization, and abundance on proteomic detection of Tumor-Derived proteins in plasma,” PLoS ONE, vol. 6, no. 7, Article ID e23090, 2011.
[122]  N. Campostrini, J. Pascali, M. Hamdan et al., “Proteomic analysis of an orthotopic neuroblastoma xenograft animal model,” Journal of Chromatography B, vol. 808, no. 2, pp. 279–286, 2004.
[123]  K. E. Turner, H. R. Kumar, D. J. Hoelz et al., “Proteomic analysis of neuroblastoma microenvironment: effect of the host-tumor interaction on disease progression,” Journal of Surgical Research, vol. 156, no. 1, pp. 116–122, 2009.
[124]  H. R. Kumar, X. Zhong, D. J. Hoelz et al., “Three-dimensional neuroblastoma cell culture: proteomic analysis between monolayer and multicellular tumor spheroids,” Pediatric Surgery International, vol. 24, no. 11, pp. 1229–1234, 2008.
[125]  A. J. Spillane, R. A'Hern, I. R. Judson, C. Fisher, and J. M. Thomas, “Synovial sarcoma: a clinicopathologic, staging, and prognostic assessment,” Journal of Clinical Oncology, vol. 18, no. 22, pp. 3794–3803, 2000.
[126]  M. Trassard, V. Le Doussal, K. Hacène et al., “Prognostic factors in localized primary synovial sarcoma: a multicenter study of 128 adult patients,” Journal of Clinical Oncology, vol. 19, no. 2, pp. 525–534, 2001.
[127]  J. Clark, P. J. Rocques, A. J. Crew et al., “Identification of novel genes, SYT and SSX, involved in the t(X;18)(p11.2;q11.2) translocation found in human synovial sarcoma,” Nature Genetics, vol. 7, no. 4, pp. 502–508, 1994.
[128]  A. Kawai, J. Woodruff, J. H. Healey, M. F. Brennan, C. R. Antonescu, and M. Ladanyi, “SYT-SSX gene fusion as a determinant of morphology and prognosis in synovial sarcoma,” New England Journal of Medicine, vol. 338, no. 3, pp. 153–160, 1998.
[129]  Y. Suehara, N. Tochigi, D. Kubota et al., “Secernin-1 as a novel prognostic biomarker candidate of synovial sarcoma revealed by proteomics,” Journal of Proteomics, vol. 74, no. 6, pp. 829–842, 2011.
[130]  Y. Suehara, K. Kikuta, R. Nakayama et al., “GST-P1 as a histological biomarker of synovial sarcoma revealed by proteomics,” Proteomics, vol. 3, no. 5, pp. 623–634, 2009.
[131]  F. M. Enzinger, “Clear-cell sarcoma of tendons and aponeuroses. An analysis of 21 cases,” Cancer, vol. 18, pp. 1163–1174, 1965.
[132]  J. Zucman, O. Delattre, C. Desmaze et al., “EWS and ATF-1 gene fusion induced by t(12;22) translocation in malignant melanoma of soft parts,” Nature Genetics, vol. 4, no. 4, pp. 341–345, 1993.
[133]  E. B. Chung and F. M. Enzinger, “Malignant melanoma of soft parts. A reassessment of clear cell sarcoma,” American Journal of Surgical Pathology, vol. 7, no. 5, pp. 405–413, 1983.
[134]  H. A. Tawbi and J. M. Kirkwood, “Management of Metastatic Melanoma,” Seminars in Oncology, vol. 34, no. 6, pp. 532–545, 2007.
[135]  A. Kawai, A. Hosono, R. Nakayama et al., “Clear cell sarcoma of tendons and aponeuroses: a study of 75 patients,” Cancer, vol. 109, no. 1, pp. 109–116, 2007.
[136]  K. Dimas, C. Tsimplouli, A. K. Anagnostopoulos et al., “The proteome profile of two cell lines and their xenografts isolated from a patient with clear cell sarcoma (soft tissue melanoma),” Cancer Genomics and Proteomics, vol. 5, no. 3-4, pp. 175–238, 2008.
[137]  A. Celis, H. H. Rasmussen, P. Celis et al., “Short-term culturing of low-grade superficial bladder transitional cell carcinomas leads to changes in the expression levels of several proteins involved in key cellular activities,” Electrophoresis, vol. 20, no. 2, pp. 355–361, 1999.
[138]  S. W. Weiss and J. R. Goldblum, “Malignant soft tissue tumors of uncertain type,” in Enzinger and Weiss's Soft Tissue Tumors, pp. 1161–1220, Elsevier Mosby, Philadelphia, Pa, USA, 2008.
[139]  N. G. Ordó?ez, “Alveolar soft part sarcoma: a review and update,” Advances in Anatomic Pathology, vol. 6, no. 3, pp. 125–139, 1999.
[140]  F. H. Shipkey, P. H. Lieberman, F. W. Foote, and F. W. Stewart, “Ultrastructure of alveolar soft part sarcoma,” Cancer, vol. 17, pp. 821–830, 1964.
[141]  K. Tsuji, Y. Ishikawa, and T. Imamura, “Technique for differentiating alveolar soft part sarcoma from other tumors in paraffin-embedded tissue: comparison of immunohistochemistry for TFE3 and CD147 and of reverse transcription polymerase chain reaction for ASPSCR1-TFE3 fusion transcript,” Human Pathology, vol. 43, no. 3, p. 356, 2011.
[142]  B. M. Balgley, T. Guo, K. Zhao, X. Fang, F. A. Tavassoli, and C. S. Lee, “Evaluation of archival time on shotgun proteomics of formalin-fixed and paraffin-embedded tissues,” Journal of Proteome Research, vol. 8, no. 2, pp. 917–925, 2009.
[143]  S. Meding, U. Nitsche, B. Balluff et al., “Tumor classification of six common cancer types based on proteomic profiling by MALDI imaging,” Journal of Proteome Research, vol. 11, no. 3, pp. 1996–2003, 2012.
[144]  C.-Y. Chang, P. Picotti, R. Hüttenhain et al., “Protein significance analysis in selected reaction monitoring (SRM) measurements,” Molecular and Cellular Proteomics, vol. 11, no. 4, 2012.
[145]  T. Kondo, “Cancer proteome-expression database: genome Medicine Database of Japan Proteomics,” Expert Review of Proteomics, vol. 7, no. 1, pp. 21–27, 2010.
[146]  M. Watanabe, Y. Inoue, C. Chang et al., “For what am I participating? The need for communication after receiving consent from biobanking project participants: experience in Japan,” Journal of Human Genetics, vol. 56, no. 5, pp. 358–363, 2011.
[147]  C. F. Taylor, N. W. Paton, K. S. Lilley et al., “The minimum information about a proteomics experiment (MIAPE),” Nature Biotechnology, vol. 25, no. 8, pp. 887–893, 2007.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413