Horm Metab Res 2011; 43(12): 865-871
DOI: 10.1055/s-0031-1291333
Humans, Clinical
© Georg Thieme Verlag KG Stuttgart · New York

Loss of PTEN Expression in Neuroendocrine Pancreatic Tumors

M. Krausch*
1   Department of General, Visceral and Pediatric Surgery, , Heinrich-Heine University of Düsseldorf, Germany
,
A. Raffel*
1   Department of General, Visceral and Pediatric Surgery, , Heinrich-Heine University of Düsseldorf, Germany
,
M. Anlauf
2   Institute of Pathology, Heinrich-Heine University of Düsseldorf, Germany
,
M. Schott
3   Department of Endocrinology, Diabetes and Rheumatology, Heinrich-Heine University of Düsseldorf, Germany
,
H. Willenberg
3   Department of Endocrinology, Diabetes and Rheumatology, Heinrich-Heine University of Düsseldorf, Germany
,
N. Lehwald
1   Department of General, Visceral and Pediatric Surgery, , Heinrich-Heine University of Düsseldorf, Germany
,
D. Hafner
4   Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine University of Düsseldorf, Germany
,
K. Cupisti
1   Department of General, Visceral and Pediatric Surgery, , Heinrich-Heine University of Düsseldorf, Germany
,
C. F. Eisenberger
1   Department of General, Visceral and Pediatric Surgery, , Heinrich-Heine University of Düsseldorf, Germany
,
W. T. Knoefel
1   Department of General, Visceral and Pediatric Surgery, , Heinrich-Heine University of Düsseldorf, Germany
› Author Affiliations
Further Information

Publication History

received 04 March 2011

accepted 13 October 2011

Publication Date:
21 November 2011 (online)

Abstract

PTEN (phosphatase and tensin homologue deleted from chromosome 10) is a well established tumor suppressor gene, which was cloned to chromosome 10q23. PTEN plays an important role in controlling cell growth, apoptosis, cell adhesion, and cell migration. In various studies, a genetic change as well as loss of PTEN expression by different carcinomas has been described. To date, the role of PTEN as a differentiation marker for neuroendocrine tumors (NET) and for the loss of PTEN expression is still unknown. It is assumed that loss of PTEN expression is important for tumor progression of NETs. We hypothesize that PTEN might be used as a new prognostic marker. We report 38 patients with a NET of the pancreas. Tumor tissues were surgically resected, fixed in formalin, and embedded in paraffin. PTEN expression was evaluated by immunohistochemistry and was correlated with several clinical and pathological parameters of each individual tumor. After evaluation of our immunohistochemistry data using a modified Remmele Score, a widely accepted method for categorizing staining results for reports and statistical evaluation, staining results of PTEN expression were correlated with the clinical and pathological parameters of each individual tumor. Our data demonstrates a significant difference in survival with existence of lymph node or distant metastases. Negative patients show a significant better survival compared with positive patients. Furthermore, we show a significant difference between PTEN expression and WHO or TNM classification. Taken together, our data shows a positive correlation between WHO classification and the new TNM classification of NETs, and loss of PTEN expression as well as survival. These results strongly implicate that PTEN might be helpful as a new prognostic factor.

*

*  These authors contributed equally to this study and share the first authorship.


 
  • References

  • 1 Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, Puc J, Miliaresis C, Rodgers L, McCombie R, Bigner SH, Giovanella BC, Ittmann M, Tycko B, Hibshoosh H, Wigler MH, Parsons R. PTEN a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 1997; 275: 1943-1947
  • 2 Li DM, Sun H. TEP1, encoded by a candidate tumor suppressor locus, is a novel protein tyrosine phosphatase regulated by transforming growth factor beta. Cancer Res 1997; 57: 2124-2129
  • 3 Steck PA, Pershouse MA, Jasser SA, Yung WK, Lin H, Ligon AH, Langford LA, Baumgard ML, Hattier T, Davis T, Frye C, Hu R, Swedlund B, Teng DH, Tavtigian SV. Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat Genet 1007; 15: 356-362
  • 4 Zhao H, Dupont J, Yakar S, Karas M, LeRoith D. PTEN inhibits egulating cell proliferation and induces apoptosis by downrcell surface IGF-IR expression in prostate cancer cells. Oncogene 2004; 23: 786-794
  • 5 Konopka B, Janiec-Jankowska A, Paszko Z, Goluda M. The coexistence of ERBB2, INT2, and CMYC oncogene amplifications and PTEN gene mutations in endometrial carcinoma. J Cancer Res Clin Oncol 2004; 130: 114-121
  • 6 Brown KS, Blair D, Reid SD, Nicholson EK, Harnett MM. FcgammaRIIb-mediated negative regulation of BCR signalling is associated with the recruitment of the MAPKinasephosphatase, Pac-1, and the 3′-inositol phosphatase, PTEN. Cell Signal 2004; 16: 71-80
  • 7 Cho SH, Lee CH, Ahn Y, Kim H, Kim H, Ahn CY, Yang KS, Lee SR. Redox regulation of PTEN and protein tyrosine phosphatases in H(2)O(2) mediated cell signaling. FEBS Lett 2004; 560: 7-13
  • 8 Wishart MJ, Dixon JE. PTEN and myotubularin phosphatases: from 3-phosphoinositide dephosphorylation to disease. Trends Cell Biol 2002; 12: 579-585
  • 9 Mills GB, Kohn E, Lu Y, Eder A, Fang X, Wang H, Bast RC, Gray J, Jaffe R, Hortobagyi G. Linking molecular diagnostics to molecular therapeutics: targeting the PI3K pathway in breast cancer. Semin Oncol 2003; 30 (Suppl. 16) 93-104
  • 10 Orchiston EA, Bennett D, Leslie NR, Clarke RG, Winward L, Downes CP Safrany ST PTEN M-CBR3 a versatile and selective regulator of inositol 1,3,4,5,6-pentakisphosphate (Ins (1,3,4,5,6) P5). Evidence for Ins (1,3,4,5,6)P5 as a proliferative signal. J Biol Chem 2004; 279: 1116-1122
  • 11 Wang L, Ignat A, Axiotis CA. Differential expression of the PTEN tumor suppressor protein in fetal and adult neuroendocrine tissues and tumors: progressive loss of PTEN expression in poorly differentiated neuroendocrine neoplasms. Appl Immunohistochem Mol Morphol 2002; 10: 139-146
  • 12 Li XH, Zheng HC, Takahashi H, Masuda S, Yang XH, Takano Y. PTEN expression and mutation in colorectal carcinomas. Oncol Rep 2009; 22: 757-764
  • 13 Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A, Schulick RD, Tang LH, Wolfgang CL, Choti MA, Velculescu VE, Diaz Jr LA, Vogelstein B, Kinzler KW, Hruban RH, Papadopoulos N. DAXX/ATRX, MEN1, and mTOR Pathway Genes Are Frequently Altered in Pancreatic Neuroendocrine Tumors. Science 2011; 331 (6021) 1199-1203
  • 14 Perren A, Komminoth P, Saremaslani P, Matter C, Feurer S, Lees JA, Heitz PU, Eng C. Mutation and expression analysis reveal differential subcellular compartmentalization of PTEN in endocrine pancreatic tumors compared to normal islet cells. Am J Pathol 2000; 157: 1097-1103
  • 15 Rindi G, Klöppel G, Alhman H, Caplin M, Couvelard A, de Herder WW, Erikssson B, Falchetti A, Falconi M, Komminoth P, Körner M, Lopes JM, McNicol AM, Nilsson O, Perren A, Scarpa A, Scoazec JY, Wiedenmann B. and all other Frascati Consensus Conference participants; European Neuroendocrine Tumor Society (ENETS) . TNM staging of foregut (neuro)endocrine tumors: a consensus proposal including a grading system. Virchows Arch 2006; 449: 395-401
  • 16 McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM. Statistics Subcommittee of the NCI-EORTC Working Group on Cancer Diagnostics . Reporting recommendations for tumor marker prognostic studies (remark). Exp Oncol 2006; 28: 99-105
  • 17 Lohmann DR, Funk A, Niedermeyer HP, Haupel S, Hofler H. Identification of p53 gene mutations in gastrointestinal and pancreatic carcinoids by nonradioisotopic SSCA. Virchows Arch B Cell Pathol 1993; 64: 293-296
  • 18 Hofler H, Ruhri C, Putz B, Wirnsberger G, Hauser H. Oncogene expression in endocrine pancreatic tumors. Virchows Arch B Cell Pathol 1988; 55: 355-361
  • 19 Speel EJ, Richter J, Moch H, Egenter C, Saremaslani P, Rutimann K, Zhao J, Barghorn A, Roth J, Heitz PU, Komminoth P. Genetic differences in endocrine pancreatic tumor subtypes detected by comparative genomic hybridization. Am J Pathol 1999; 155: 1787-1794
  • 20 O’Connor R. Regulation of IGF-I receptor signaling in tumor cells. Horm Metab Res 2003; 35: 771-777
  • 21 Podsypanina K, Ellenson LH, Nemes A, Gu J, Tamura M, Yamada KM, Cordon-Cardo C, Catoretti G, Fisher PE, Parsons R. Mutation of Pten/Mmac1 in mice causes neoplasia in multiple organ systems. Proc Natl Acad Sci USA 1999; 96: 1563-1568
  • 22 Suzuki A, de la Pompa JL, Stambolic V, Elia AJ, Sasaki T, del Barco Barrantes I, Ho A, Wakeham A, Itie A, Khoo W, Fukumoto M, Mak TW. High cancer susceptibility and embryonic lethality associated with mutation of the PTEN tumor suppressor gene in mice. Curr Biol 1998; 8: 1169-1178
  • 23 Cheng JQ, Godwin AK, Bellacosa A, Taguchi T, Franke TF, Hamilton TC, Tsichlis PN Testa JR AKT2 a putative oncogene encoding a member of a subfamily of protein-serine/threonine kinases, is amplified in human ovarian carcinomas. Proc Natl Acad Sci USA 1992; 89: 9267-9271
  • 24 Boehle AS, Kurdow R, Boenicke L, Schniewind B, Faendrich F, Dohrmann P, Kalthoff H. Wortmannin inhibits growth of human non-small-cell lung cancer in vitro and in vivo. Langenbecks Arch Surg 2002; 387: 234-239
  • 25 Lee SR, Yang KS, Kwon J, Lee C, Jeong W, Rhee SG. Reversible inactivation of the tumor suppressor PTEN by H2O2 . J Biol Chem 2002; 277: 20336-20342
  • 26 Gericke A, Munson M, Ross AH. Regulation of the PTEN phosphatase. Gene 2006; 374: 1-9
  • 27 Missiaglia E, Dalai I, Barbi S, Beghelli S, Falconi M, della Peruta M, Piemonti L, Capurso G, Di Florio A, delle Fave G, Pederzoli P, Croce CM, Scarpa A. Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol 2010; 28: 245-255
  • 28 Liliental J, Moon SY, Lesche R, Mamillapalli R, Li D, Zheng Y, Sun H, Wu H. Genetic deletion of the Pten tumor suppressor gene promotes cell motility by activation of Rac1 and Cdc42 GTPases. Curr Biol 2000; 10: 401-404
  • 29 Suzuki A, Kaisho T, Ohishi M, Tsukio-Yamaguchi M, Tsubata T, Koni PA, Sasaki T, Mak TW, Nakano T. Critical roles of Pten in B cell homeostasis and immunoglobulin class switch recombination. J Exp Med 2003; 197: 657-667
  • 30 Suzuki A, Kaishu T, Ohishi M, Tsukio-Yamaguchi M, Tsubata T, Koni PA, Sasaki T, Mak TW, Nakano T. Critical roles of Pten in B cell homeostasis and immunoglobulin class switch recombination. J Exp Med 2003; 197: 657-667
  • 31 Tamura M, Gu J, Matsumoto K, Aota S, Parsons R, Yamada KM. Inhibition of cell migration, spreading, and focal adhesions by tumor suppressor PTEN. Science 1998; 280: 1614-1617
  • 32 Lacalle RA, Gomez-Mouton C, Barber DF, Jimenez-Baranda S, Mira E, Martinez AC, Carrera AC, Manes S. PTEN regulates motility but not directionality during leukocyte chemotaxis. J Cell Sci 2004; 117: 6207-6215
  • 33 Sanchez T, Thangada S, Wu MT, Kontos CD, Wu D, Wu H, Hla T. PTEN as an effector in the signaling of antimigratory G protein-coupled receptor. Proc Natl Acad Sci USA 2005; 102: 4312-4317
  • 34 Chalhoub N, Baker JS. PTEN and the PI3-Kinase Pathway in Cancer. Annu Rev Pathol Mech Dis 2009; 4: 127-150
  • 35 Jiang BH, Liu LZ. PI3K/PTEN signalling in angiogenesis and tumoroginesis. Adv Cancer Res 2009; 102: 19-65
  • 36 Sawai H, Yasuda A, Ochi N, Ma J, Matsuo Y, Wakasugi T, Takahashi H, Funahashi H, Sato M, Takeyama H. Loss of PTEN expression is associated with colorectal cancer liver metastasis and poor patient survival. BMC Gastroenterology 2008; 8: 56
  • 37 Bardelli A, Pierotti MA. Phosphatase protein homologue to tensin expression and phosphatidylinositol-3 phosphate kinase mutations in colorectal cancer. Cancer Res 2005; 65: 11227
  • 38 Frattini M, Saletti P, Romagnani E, Martin V, Molinari F, Ghisletta M, Camponovo A, Etienne LL, Cavalli F, Mazzucchelli L. PTEN loss of expression predicts cetuximab efficacy in metastatic colorectal cancer patients. Br J Cance 2007; 97: 1139-1145
  • 39 Jhawer M, Goel S, Wilson AJ, Montagna C, Ling YH, Byun DS, Nasser S, Arango D, Shin J, Klampfer L, Augenlicht LH, Soler RP, Mariadason JM. PIK3CA mutation/PTEN expression status predicts response of colon cancer cells to the epidermal growth factor receptor inhibitor cetuximab. Cancer Res 2008; 68: 1953-1961
  • 40 Jang K-S, Song YS, Jang S-H, Min K-W, Na W, Jang SM, Jun YJ, Lee KH, Choi D, Paik SS. Clinicopathological significance of nuclear PTEN expression in colorectal adenocarcinoma. Histopathology 2010; 56: 229-239
  • 41 O’Toole D, Couvelard A, Rebours V, Zappa M, Hentic O, Hammel P, Levy P, Bedossa P, Raymond E, Ruszniewski P. Molecular markers associated with response to chemotherapy in gastro-entero-pancreatic neuroendocrine tumors. Endocr Relat Cancer 2010; 17: 847-856