DOI QR코드

DOI QR Code

Tumor-Suppression Mechanisms of Protein Tyrosine Phosphatase O and Clinical Applications

  • Kang, Man-Man (The Center of Radiation Oncology, the 82th Hospital of People's Liberation Army of China) ;
  • Shan, Shun-Lin (The Center of Radiation Oncology, the 82th Hospital of People's Liberation Army of China) ;
  • Wen, Xu-Yang (The Center of Radiation Oncology, the 82th Hospital of People's Liberation Army of China) ;
  • Shan, Hu-Sheng (The Center of Radiation Oncology, the 82th Hospital of People's Liberation Army of China) ;
  • Wang, Zheng-Jun (The Center of Radiation Oncology, the 82th Hospital of People's Liberation Army of China)
  • Published : 2015.10.06

Abstract

Tyrosine phosphorylation plays an important role in regulating human physiological and pathological processes. Functional stabilization of tyrosine phosphorylation largely contributes to the balanced, coordinated regulation of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs). Research has revealed PTPs play an important suppressive role in carcinogenesis and progression by reversing oncoprotein functions. Receptor-type protein tyrosine phosphatase O (PTPRO) as one member of the PTPs family has also been identified to have some roles in tumor development. Some reports have shown PTPRO over-expression in tumors can not only inhibit the frequency of tumor cell division and induce tumor cell death, but also suppress migration. However, the tumor-suppression mechanisms are very complex and understanding is incomplete, which in some degree blocks the further development of PTPRO. Hence, in order to resolve this problem, we here have summarized research findings to draw meaningful conclusions. We found tumor-suppression mechanisms of PTPRO to be diverse, such as controlling G0/G1 of the tumor cell proliferation cycle, inhibiting substrate phosphorylation, down-regulating transcription activators and other activities. In clinical anticancer efforts, expression level of PTPRO in tumors can not only serve as a biomarker to monitor the prognosis of patients, but act as an epigenetic biomarker for noninvasive diagnosis. In addition, the re-activation of PTPRO in tumor tissues, not only can induce tumor volume reduction, but also enhance the susceptibility to chemotherapy drugs. So, we can propose that these research findings of PTPRO will not only support new study ideas and directions for other tumor-suppressors, importantly, but also supply a theoretical basis for researching new molecular targeting agents in the future.

Keywords

References

  1. Andres A, Joanna S, Nunzio B, et al (2004). Protein tyrosine phosphatases in the human genome. Cell, 117, 699-711. https://doi.org/10.1016/j.cell.2004.05.018
  2. Andersen JN, Jansen PG., Echwald, S.M., et al (2004). Agenomic perspective on protein tyrosine phosphatases: gene structure, pseudogenes, and genetic disease linkage. FASEB J, 18, 8-13. https://doi.org/10.1096/fj.02-1212rev
  3. Andersen JN, Mortensen, OH, Peters GH, et al (2001). Structural and evolutionary relationships among protein tyrosine phosphatase domains. Mol Cell Biol, 21, 7117-36. https://doi.org/10.1128/MCB.21.21.7117-7136.2001
  4. Arena S, Benvenuti S, Bardelli (2005). Genetic analysis of the kinome and phosphatome in cancer. Cell Mol Life Sci, 62, 2092-9. https://doi.org/10.1007/s00018-005-5205-1
  5. Adams RH, Wilkinson GA, Weiss C, Diella F, et al (1999). Roles of ephrinB ligands and EphB receptors in cardiovascular development: demarcation of arterial/venous domains, vascular morphogenesis, and sprouting angiogenesis. Genes Dev, 3, 295-306.
  6. Andres A-C, Reid HH, Zurcher G, Blaschke RJ, et al (1994). Expression of two novel eph-related receptor protein tyrosine kinases in mammary gland development and carcinogenesis. Oncogene, 9, 1461-7.
  7. Alvarez JV, Greulich H, Sellers WR, et al (2006). Signal transducer and activator of transcription 3 is required for the oncogenic effects of non-small-cell lung cancer-associated mutations of the epidermal growth factor receptor. Cancer Res, 66, 3162-8. https://doi.org/10.1158/0008-5472.CAN-05-3757
  8. Bhise SB, Nalawade AD, Wadhawa H (2004). Role of potein tyrosine kinase inhibitors in cancer therapeutics. Indian J Biochem Biophys, 41, 273-80.
  9. Bradford JW, Baldwin AS (2014). Ikk/nuclear factor-kappaB and oncogensis: roles in tumor-initiating cells and in the tumor microenvironment. Adv Cancer Res, 121, 125-45. https://doi.org/10.1016/B978-0-12-800249-0.00003-2
  10. Bursavich MG, Parker DP, Willardsen JA, et al (2010). 2-Anilino-4aryl-1,3-thiazole inhibitors of valosin-containing protein(VCP or p97). Bioorg Med Chem Lett, 20, 1677-9. https://doi.org/10.1016/j.bmcl.2010.01.058
  11. Brukner KKR (1998). Signaling by Eph receptors and their ephrin ligands. Curr Opin Neurobiol, 8, 375-82. https://doi.org/10.1016/S0959-4388(98)80064-0
  12. Boccaccio C, Ando M, Tamagnone L, et al (1998). Induction of epithelial tubules by growth factor HGF depends on the STAT pathway. Nature, 391, 285-8. https://doi.org/10.1038/34657
  13. Bournazou E, Bromberg J (2013). Targeting the tumor microenvironment JAK-STAT3 signaling. JAKSTAT, 2, 23828.
  14. Chen LF, Juszcynski P, Takeyama K, et al (2006). Protein tyrosine phosphatase receptor-type O truncated (PTPROt) regulates SYK phosphorylation proximal B-cell-receptor signaling, and cellular proliferation. Blood, 108, 3428-33. https://doi.org/10.1182/blood-2006-03-013821
  15. Coopman PJ, Do MT, Barth M, et al (2000). The Syk tyrosine kinase suppresses malignant growth of human breast cancer cells. Nature, 406, 742-7. https://doi.org/10.1038/35021086
  16. Campbell KS (1999). Signal transduction from the B cell antigen-receptor. Curr Opin Immunol, 11, 256-64. https://doi.org/10.1016/S0952-7915(99)80042-9
  17. Chen Z, Han ZC (2008). STAT3: a critical transcription activator in angiogenesis. Med Res Rev, 28, 185-200. https://doi.org/10.1002/med.20101
  18. D’Arcy P, Maruwge W, Wolahan B, et al (2014). Oncogenic functions of the cancertestis antigen SSX on the proliferation, survival, and singaling pathways of cancer cells. PloS One, 9, 95136. https://doi.org/10.1371/journal.pone.0095136
  19. Dana BS, Sonja S, Monica P, et al (2004). Eph Receptor Tyrosine Kinases in Tumor and Tumor Microenvironment. Current Pharmaceutical Design, 10, 3431-42. https://doi.org/10.2174/1381612043383160
  20. Davy A, Robbins SM (2000). EphrinA5 modultes cell adhesion and morphology in an integrin-dependent manner. EMBO J, 19, 5346-406.
  21. David V, Dana M BS, Jin C (2008). Eph receptors in breast cancer: roles in tumor promotion and tumor suppression. Breast Cancer Res, 10, 217-24. https://doi.org/10.1186/bcr2207
  22. Dudka AA, Sweet SM, Heath JK (2010). Signal transducers and activators of transcription-3 binding to the fibroblast growth factor receptor is activated by receptor amplification. Cancer Res, 70, 3391-401. https://doi.org/10.1158/0008-5472.CAN-09-3033
  23. Douglas Hanahan, Robert A (2011). Weinberg. hallmarks of cancer: the next generation. Cell, 144, 646-674. https://doi.org/10.1016/j.cell.2011.02.013
  24. De Raeve H, Van Marck E, Van Camp B, et al (2004). Angiogenesis and therole of bone marrow endothelial cells in haematological malignancies. Histol Histopathol, 19, 935-50.
  25. Easty D, Hill S, Hsu M, et al (1999). Up-regulation of ephrin-A1 during melanoma progression. Int J Cancer, 84, 494-501. https://doi.org/10.1002/(SICI)1097-0215(19991022)84:5<494::AID-IJC8>3.0.CO;2-O
  26. Fabbro D, Ruetz S, Buchdunger E, et al (2002). Protein kinases as a targets for anticancer agents: from inhibitors to useful drugs, Pharmacol Ther, 93, 79-98. https://doi.org/10.1016/S0163-7258(02)00179-1
  27. Fu C, Turck CW, Kurosaki T, et al (1998). BLNK: a central linker protein in B cell activation. Immunity, 9, 93-103. https://doi.org/10.1016/S1074-7613(00)80591-9
  28. Frohlich KU, Fries HW, Rudiger M, et al (1991). Yeast cell cycle protein CDC48p shows full-length homology to the mammalian protein VCP and is a member of a protein family involved in secretion, peroxisome formation, and gene expression. JCell Biol, 114, 443-53. https://doi.org/10.1083/jcb.114.3.443
  29. Flanagan JG, Vanderhaeghen P (1998). The eprhins and Eph receptors in neural development. Annu Rev Neurosci, 21, 309-45. https://doi.org/10.1146/annurev.neuro.21.1.309
  30. Gauld SB, Dal Porto JM, Cambier JC (2002). B cell antigen receptor signaling: roles in cell development and disease. Sci, 296, 1641-2. https://doi.org/10.1126/science.1071546
  31. Gammarota M, Bevilaqua LR, Dunkley PR, et al (2001). Angiotensin II promotes the phosphorylation of cyclin AMP-responsive element binding protein (CREB) at Ser 133 through an ERK1/2 dependent mechanism. J Neurochem, 79, 1122-8.
  32. Gaughn C, Liu WQ, Vidal M, et al (2000). Inhibitors of Ras signal transductio as antitumor agents. Biochem Pharmacol, 60, 1165-9. https://doi.org/10.1016/S0006-2952(00)00428-7
  33. Gasparini C, Celeghini C, Monasta L, et al (2014). NF-kB pathways in hematological malignancies. Cell Mol Life Sci, 71, 2083-102. https://doi.org/10.1007/s00018-013-1545-4
  34. Gale NW, Holland SJ, Valenzuela DM, et al (1996). Eph receptors and ligands comprise two major specificity subclasses and are reciprocally compartmentalized during embryogenesis. Neuron, 17, 9-19. https://doi.org/10.1016/S0896-6273(00)80276-7
  35. Hsu, Tasneem M, Satavisha R, et al (2013). Methylation of gene encoding the growth suppressor protein tyrosine phosphatase receptor-type O (PTPRO) in human hepatocellular carcinoma and identification of VCP as its bona fide substrate. J Cellular Biochemistry, 1002, 134-55.
  36. Hashimoto S, Iwamatsu A, Ishiai M, Okawa K, et al (1999). Identification of the SH2 domain binding protein of Bruton’s tyrosin kinase as BLNK-functional significance of Btk-SH2 domain in B-cell antigen receptor coupled calcium signaling. Blood, 94, 2357-64.
  37. Hirai H, Maru Y, Hagiwara K, et al (1987). A novel putative tyrosine kinase receptor encoded by the Eph gene. Sci, 238, 1717-20. https://doi.org/10.1126/science.2825356
  38. Himanen JP, Nikolov DB (2003). Eph receptors and ephrins. Int J Biochem Cell Biol, 35, 130-4. https://doi.org/10.1016/S1357-2725(02)00096-1
  39. Hart, I. R, Fidler, I. J (1980). Role of organ selectivity in the determination of metastatic patterns of the B16 melanoma. Cancer Res, 40, 2281-7.
  40. Izabela L, Maria M S (2011). Tyrosine phosphatases as a superfamily of tumor suppressors in colorectal cancer. Acta Biochimica Polonica, 4, 467-70.
  41. Johnson KG, Van Vactor D (2003). Receptor protein tyrosine phosphatases in nervous system development. Physiol Rev, 83, 1-24. https://doi.org/10.1152/physrev.00016.2002
  42. Jia JH, Juan X, Run QJ, You JW, et al (2013). Estrogen-sensitive PTPRO expression represses hepatocellular carcinoma progression by control of STAT3. Hepatol, 57, 678-88. https://doi.org/10.1002/hep.25980
  43. Johnson GL, Lapadat R (2002). Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases. Sci, 298, 1911-2. https://doi.org/10.1126/science.1072682
  44. Jentsch S, Rumpf S (2007). Cdc48 (p97): a "molecular gearbox" in the ubiquitin pathway? Trends Biochem Sci, 32, 6-11. https://doi.org/10.1016/j.tibs.2006.11.005
  45. Kalo MS, Pasquale EB (1999). Signal transfer by Eph receptors. Cell Tissue Res, 298, 1-9. https://doi.org/10.1007/PL00008807
  46. Kinch MS, Moore MB, Harpole DH Jr (2003). Predictive value of the EphA2 receptor tyrosine kinase in lung cancer recurrence and survival. Clin Cancer Res, 9, 613-8.
  47. Kreis S, Munz GA, Haan S, et al (2007). Cell density dependent increase of constitutive signal transducers and activators of transcription 3 activity in melanoma cells is mediated by Janus Kinases. Mol Cancer Res, 5, 1331-41. https://doi.org/10.1158/1541-7786.MCR-07-0317
  48. Laurie Stepanek, Andrew W. Stoker, Esther Stoeckli, et al (2005). Receptor tyrosine phosphatases guide vertebrate motor axons during development. Soc Neurosci, 25, 3813-23. https://doi.org/10.1523/JNEUROSCI.4531-04.2005
  49. Liu B, Kuang A (2012). Genetic alterations in MAPK and PI3K/ Akt signaling pathways and the generation, progression, diagnosis and therapy of thyroid cancer. Sheng Wu Yi Xue Gong Cheng Xue Za Zhi, 29, 1221-5.
  50. Long XH, Zhang ZH, Liu ZL, et al (2013). Inhibiting valosincontaining protein suppresses osteosarcoma cell metastasis via AKT/nuclear factor of kappa B singaling pathway in vitro. Indian J Pathol Microbiol, 56, 190-5. https://doi.org/10.4103/0377-4929.120358
  51. Layka AA, Iria V, Loredana V, et al (2014). The tyrosine phosphatase PTPRO sensitizes colon cancer cells to anti- EGFR therapy through activation of SRC-mediated EGFR signaling. Oncotarget, 5, 10070-83. https://doi.org/10.18632/oncotarget.2458
  52. Miller CR, Oliver KE, Farley JH (2014). MEK1/2 inhibitors in the treatment of gynecologic maligancies. Gynecol Oncol, 133, 128-37.
  53. Motiwala T, Majumder S, Kutsy H, et al (2007). Methylation and silencing of protein tyrosine phosphatase receptor type O in chronic lymphocytic leukemia. Clin Cancer Res, 13, 3174-81. https://doi.org/10.1158/1078-0432.CCR-06-1720
  54. Nikki Chenga, Dana M. Brantley, et al (2002). The ephrins and Eph receptors in angiogenesis. Cytokine Growth Factor Rev, 13, 75-85. https://doi.org/10.1016/S1359-6101(01)00031-4
  55. Paolo R, Barbara B, Andrea C (2009). Therapeutic prospect of Syk inhibitors. Exper Opin Ther Pat, 19, 1361-76. https://doi.org/10.1517/13543770903207039
  56. Panchamoorthy G, Fukazawa T, Miyake S, et al (1996). p120cbl is a major substrate of tyrosine phophorylation upon B cell antigen receptor stimulation and interacts in vivo with Fyn and Syk tyrosine kinases, Grb2 and shc adaptors, and the p85 subunit of phosphatidylinositol 3-kinase. J Biol Chem, 271, 3187-94. https://doi.org/10.1074/jbc.271.6.3187
  57. Ricardo C.T. A, Yoshihiro Y, Samir K, et al (1999). Freeman, Margaret A. Shipp. PTPROt: An Alternatively Spliced and Developmentally Regulated B-Lymphoid Phosphatase That Promotes G0/G1 Arrest. Blood, 94, 2403-13.
  58. Rolli V, Gallwitz M, Wossning T, et al (2002). Amplification of B cell antigen receptor signaling by a Syk/ ITAM positive feedback loop. Mol Cell, 10, 1057-69. https://doi.org/10.1016/S1097-2765(02)00739-6
  59. Roskoski R Jr (2012). ERK1/2 MAP kinases: structure, function, and regulation. Pharmacol Res, 66, 105-143. https://doi.org/10.1016/j.phrs.2012.04.005
  60. Rao PN, Levine E, Myers MO, et al (1999). Elevation of serum riboflavin carrier protein in breast cancer. Cancer Epidemiol Biomarkers Prev, 8, 985-90.
  61. Rane SG, Reddy EP (2000). Janus kinases: components of multiple signaling pathways. Oncogene, 19, 5662-79. https://doi.org/10.1038/sj.onc.1203925
  62. Raica M, Cimpean AM, Ribatti D (2009). Angiogenesis in premalignant conditions. Eur J Cancer, 45, 1924-1934. https://doi.org/10.1016/j.ejca.2009.04.007
  63. Ramaswamy B, Majumder S, Roy S, et al (2009). Estrogenmediated suppression of the gene encoding protein tyrosine phosphatase PTPRO in human breast cancer: mechanism and role in tamoxifen sensitivity[J]. Mol Endocrinol, 23, 176-87. https://doi.org/10.1210/me.2008-0211
  64. Shi W, Hou X, Peng H, et al (2014). MEK/ERK signaling pathway is required for enterovirus 71 replication in immature dendritic cells. Virol J, 11, 227. https://doi.org/10.1186/s12985-014-0227-7
  65. Sauer RT, Bolon DN, Burton BM, et al (2004). Sculpting the proteome with AAA(+) proteases and disassembly machines. Cell, 119, 9-18. https://doi.org/10.1016/j.cell.2004.09.020
  66. Stolz A, Hilt W, Buchberger A, et al (2011). Cdc48: a power machine in protein degradation. Trends Biochemical Sci, 36, 515-23. https://doi.org/10.1016/j.tibs.2011.06.001
  67. Surawska H, Ma PC, Salgia R (2004). The role of ephrins and Eph receptors in cancer. Cytokine Growth Factor Rev, 15, 419-33. https://doi.org/10.1016/j.cytogfr.2004.09.002
  68. Song L, Turkson J, Karras JG, et al (2003). Activation of Stat3 by receptor tyrosine kinases and cytokines regulates survival in human non-small cell carcinoma cells. Oncogene, 22, 4150-65. https://doi.org/10.1038/sj.onc.1206479
  69. Saha A, Blando J, Silver E, et al (2014). 6-Shogaol from dried ginger inhibits growth of prostate cancer cells both in vitro and in vivo through inhibition of STAT3 and NF-kB signaling. Cancer Prev Res, 7, 627-38. https://doi.org/10.1158/1940-6207.CAPR-13-0420
  70. Sung SY, Chung LW (2002). Prostate tumor-stroma interaction: molecular mechanisms and opportunities for therapeutic targeting. Differentiat, 70, 506-21. https://doi.org/10.1046/j.1432-0436.2002.700905.x
  71. Sung SY, Hsieh CL, Wu D, et al (2007). Tumor microenvironment promotes cancer progression, metastasis, and therapeutic resistance. Curr Probl Cancer, 31, 36-100. https://doi.org/10.1016/j.currproblcancer.2006.12.002
  72. Shao YL, Rong L, Yu LC, et al (2014). Aberrant PTPRO methylation in tumor tissues as a potential biomarker that predicts clinical outcomes in breast cancer patients. Biol Med Central Genet, 15, 67.
  73. Samson T J, Tasneem M (2005). Epigenetic regulation of protein tyrosine phosphatases: potential molecular targets for cancer therapy. Cancer Gene Therapy, 12, 665-72. https://doi.org/10.1038/sj.cgt.7700828
  74. Thomas P.E, Wharram B.L., Goyal M, et al (1994). GLEPP1, a renal glomerular epithelial cell (podocyte) membrane protein tyrosine phosphatase. Identification, molecular cloning, and characterization in rabbit. J Biol Chem, 269, 19953-62.
  75. Takafumi S, Masaru I, Hiraki S, et al (2006). Eph receptors are negatively controlled by protein tyrosine phosphatase receptor type O. Nature Neuroscience, 9, 761-9. https://doi.org/10.1038/nn1697
  76. Tabernero L, Aricescu AR, Jones EY, et al (2008). Protein tyrosine phosphatases: structure-function relationships. FEBS J, 275, 867-82. https://doi.org/10.1111/j.1742-4658.2008.06251.x
  77. Takafumi S, Masaru I, Hiraki S, et al (2006). Eph receptors are negatively controlled by protein tyrosine phosphatase receptor type O. Nature Neurosci, 9, 761-9. https://doi.org/10.1038/nn1697
  78. Tasneem M, Kutay H, Ghoshal K, et al (2004). Protein tyrosine phosphatase receptor-type O(PTPRO) exhibits characteristics of a candidate tumor suppressor in human lung cancer. Med Sci, 38, 13844-9.
  79. Tasneem M, Kalpana G, Das A, et al (2003). Suppression of the protein tyrosine phosphatase receptor type O gene (PTPRO) by methylation in hepatocellular carcinomas. Oncogene, 22, 6319-31. https://doi.org/10.1038/sj.onc.1206750
  80. Udayakumar TS, Stratton MS, Nagle RB, et al (2002). Fibroblast growth factor-1 induced promatrilysin expression through the activation of extracellular-regulated kinases and STAT3. Neoplasia, 4, 60-7. https://doi.org/10.1038/sj.neo.7900207
  81. Vaiopoulos AG, Athanasoula KCh, Papavassiliou AG (2013). NF-kB in colorectal cancer. J mol Med, 91, 1029-37. https://doi.org/10.1007/s00109-013-1045-x
  82. ValdeZ BC, Murray D, Ramdas L, et al (2008). Altered gene expression in busulfan-resistant human myeloid leukemia. LeuK Res, 32, 1684-97. https://doi.org/10.1016/j.leukres.2008.01.016
  83. Wei HL, Chia HC, Kuo SH, et al (2013). Protein tyrosine phosphatase receptor type O (Ptpro) regulates cerebellar formation during zebrafish development through modulating Fgf signaling. Cell Mol Life Sci, 70, 2367-81. https://doi.org/10.1007/s00018-013-1259-7
  84. Wang Q, Song C, Li CC (2004). Molecular perspectives on p97-VCP: progress in understanding its structure and diverse biological functions. J Structural Biol, 146, 44-57. https://doi.org/10.1016/j.jsb.2003.11.014
  85. Yoji M, Munemasa M, Takenori K, et al (2010). Tyrosine phosphorylation of R3 subtype receptor-type protein tyrosine phosphatases and their complex formations with Grb2 or Fyn. Genes to Cells, 15, 513-24.
  86. Yang DH, Goyal M, Sharif K, et al (1996). Glomerular epithelial protein1 and podocalyxin-like protein 1 in inflammatory glomerular disease (crescentic nephritis) in rabbit and man. Lab. Invest, 74, 571-84.
  87. Yamamoto S, Tomita Y, Nakamori S, et al (2003). Elevated expression of valosin-containing protein(p97) in hepatocellular carcinoma is correlated with increased of tumor recurrence. J Clin Oncol, 21, 447-52. https://doi.org/10.1200/JCO.2003.06.068
  88. Yamamoto S, Tomita Y, Nakamori S, et al (2004). Expression level of valosin-containing protein(p97) is associated with prognosis of esophageal carcinoma. Clin Cancer Res, 10, 5558-65. https://doi.org/10.1158/1078-0432.CCR-0723-03
  89. Yamamoto S, Tomita Y, Nakamori S, et al (2003). Expression level of valosin-containing protein(p97) is strongly associated with prognosis of gastric carcinoma. J Clin Oncol, 21, 2537-44. https://doi.org/10.1200/JCO.2003.12.102
  90. Yamamoto S, Tomita Y, Nakamori S, et al (2005). Increased expression of valosin-containing protein(p97) is correlated with disease recurrence in follicular thyroid cancer. Ann Surg Oncol, 12, 925-34. https://doi.org/10.1245/ASO.2005.07.002
  91. Yokogami K, Wakisaka S, Avruch J, et al (2000). Serine phosphorylation and maximal activation of STAT3 during CNTF signaling is mediated by the rapamycin target mTOR. Curr Biol, 10, 47-50. https://doi.org/10.1016/S0960-9822(99)00268-7
  92. You YJ, Chen YP, Zheng XX, et al (2012). Aberrant methylation of the PTPRO gene in periheral blood biomarker in esophageal cell carcinoma patients. Cancer Letters, 315, 138-44. https://doi.org/10.1016/j.canlet.2011.08.032
  93. Yuriko M, Jing Y, Fumiaki S, et al (2004). Identification of Genes Uniquely Involved in Frequent Microstaellite instability Colon Cancinogenesis by Expression Profiling Combined with Epigenetic Scanning. American Associat Cancer, 64, 2434-8.
  94. Yu Du1, Jennifer R. Grandis (2015). Receptor-type protein tyrosine phosphatases in cancer. Chinese J Cancer, 34, 61-9.
  95. Yi TH, Fei FL, Chen K, et al (2013). PTPRO promoter methylation is predictive of poorer outcome for HER2-positive breast cancer: indication for personalized therapy. J Translational Med, 11, 245-52. https://doi.org/10.1186/1479-5876-11-245
  96. Zhao L, JIA JIE H, Li DONG R, el at (2015). Protein tyrosine phosphatase receptor type O expression in the tumor niche correlates with reduced tumor growth, angiogenesis, circulating tumor cells and metastasis of breast cancer. Oncol Reports, 3862, 1-7.