Skip to main content Accessibility help
×
Hostname: page-component-68945f75b7-gkscv Total loading time: 0 Render date: 2024-08-05T20:15:55.658Z Has data issue: false hasContentIssue false

Chapter 30 - The approach to metastatic endocrine tumors of unknown primary site

from Section III - Anatomical endocrine pathology

Published online by Cambridge University Press:  13 April 2017

Ozgur Mete
Affiliation:
University of Toronto
Sylvia L. Asa
Affiliation:
University of Toronto
Get access
Type
Chapter
Information
Endocrine Pathology , pp. 1028 - 1043
Publisher: Cambridge University Press
Print publication year: 2000

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Yao, JC, Hassan, M, Phan, A, et al. One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35 825 cases in the United States. J Clin Oncol 2008;26:30633072.CrossRefGoogle ScholarPubMed
Modlin, IM, Sandor, A. An analysis of 8305 cases of carcinoid tumors. Cancer 1997;79:813829.3.0.CO;2-2>CrossRefGoogle ScholarPubMed
Kirshbom, PM, Kherani, AR, Onaitis, MW, et al. Carcinoids of unknown origin: comparative analysis with foregut, midgut, and hindgut carcinoids. Surgery 1998;124:10631070.CrossRefGoogle ScholarPubMed
Kulke, MH, Siu, LL, Tepper, JE, et al. Future directions in the treatment of neuroendocrine tumors: consensus report of the National Cancer Institute neuroendocrine tumor clinical trials planning meeting. J Clin Oncol 2011;29:934943.Google Scholar
Bellizzi, AM. Assigning site of origin in metastatic neuroendocrine neoplasms: a clinically significant application of diagnostic immunohistochemistry. Adv Anat Pathol 2013 Sep;20:285314.CrossRefGoogle ScholarPubMed
Boudreaux, JP, Putty, B, Frey, DJ, et al. Surgical treatment of advanced-stage carcinoid tumors: lessons learned. Ann Surg 2005;241:839845.CrossRefGoogle ScholarPubMed
Givi, B, Pommier, SJ, Thompson, AK, et al. Operative resection of primary carcinoid neoplasms in patients with liver metastases yields significantly better survival. Surgery 2006;140:891897.Google Scholar
Moertel, CG, Kvols, LK, O'Connell, MJ, et al. Treatment of neuroendocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in the ana-plastic variants of these neoplasms. Cancer 1991;68:227232.Google Scholar
Strosberg, JR, Coppola, D, Klimstra, DS, et al. The NANETS consensus guidelines for the diagnosis and management of poorly differentiated (high-grade) extrapulmonary neuro-endocrine carcinomas. Pancreas 2010;39:799800.CrossRefGoogle Scholar
BL Neuroendocrine tumors. Fort Washington, PA: National Comprehensive Cancer Network, 2015 (http://www.nccn.org/professionals/physician_gls/f_guidelines.asp#site accessed 25 October 2015).Google Scholar
Soga, J, Tazawa, K. Pathologic analysis of carcinoids. Histologic reevaluation of 62 cases. Cancer 1971;28:990998.3.0.CO;2-K>CrossRefGoogle ScholarPubMed
Czeczok, TW, Gailey, MP, Hornick, JL, Bellizzi, AM. High grade neuroendocrine carcinomas are characterized by marked transcription factor lineage infidelity: an evaluation of 36 markers in 83 tumors. Mod Pathol 2014;27(suppl A):152A.Google Scholar
Silberg, DG, Swain, GP, Suh, ER, et al. Cdx1 and cdx2 expression during intestinal development. Gastroenterology 2000;119:961971.CrossRefGoogle ScholarPubMed
Moskaluk, CA, Zhang, H, Powell, SM, et al. Cdx2 protein expression in normal and malignant human tissues: an immunohistochemical survey using tissue microarrays. Mod Pathol 2003;16:913919.Google Scholar
Werling, RW, Yaziji, H, Bacchi, CE, et al. CDX2, a highly sensitive and specific marker of adenocarcinomas of intestinal origin: an immunohistochemical survey of 476 primary and metastatic carcinomas. Am J Surg Pathol 2003;27:303310.CrossRefGoogle ScholarPubMed
Barbareschi, M, Roldo, C, Zamboni, G, et al. CDX-2 homeobox gene product expression in neuroendocrine tumors: its role as a marker of intestinal neuroendocrine tumors. Am J Surg Pathol 2004;28:11691176.Google Scholar
Erickson, LA, Papouchado, B, Dimashkieh, H, et al. Cdx2 as a marker for neuroendocrine tumors of unknown primary sites. Endocr Pathol 2004;15:247252.Google Scholar
La Rosa, S, Rigoli, E, Uccella, S, et al. CDX2 as a marker of intestinal EC-cells and related well-differentiated endocrine tumors. Virchows Arch 2004;445:248254.Google Scholar
Jaffee, IM, Rahmani, M, Singhal, MG, et al. Expression of the intestinal transcription factor CDX2 in carcinoid tumors is a marker of midgut origin. Arch Pathol Lab Med 2006;130: 15221526.Google Scholar
Lin, X, Saad, RS, Luckasevic, TM, et al. Diagnostic value of CDX-2 and TTF-1 expressions in separating metastatic neuroendocrine neoplasms of unknown origin. Appl Immunohistochem Mol Morphol 2007;15:407414.CrossRefGoogle ScholarPubMed
Schmitt, AM, Riniker, F, Anlauf, M, et al. Islet 1 (Isl1) expression is a reliable marker for pancreatic endocrine tumors and their metastases. Am J Surg Pathol 2008;32: 420425.CrossRefGoogle ScholarPubMed
Srivastava, A, Hornick, JL. Immunohistochemical staining for CDX-2, PDX-1, NESP-55, and TTF-1 can help distinguish gastrointestinal carcinoid tumors from pancreatic endocrine and pulmonary carcinoid tumors. Am J Surg Pathol 2009;33:626632.CrossRefGoogle ScholarPubMed
Hermann, G, Konukiewitz, B, Schmitt, A, et al. Hormonally defined pancreatic and duodenal neuroendocrine tumors differ in their transcription factor signatures: expression of ISL1, PDX1, NGN3, and CDX2. Virchows Arch 2011;459:147154.Google Scholar
Chan, ES, Alexander, J, Swanson, PE, et al. PDX-1, CDX-2, TTF-1, and CK7: a reliable immunohistochemical panel for pancreatic neuroendocrine neoplasms. Am J Surg Pathol 2012;36:737743.Google Scholar
Denby, KS, Briones, AJ, Bourne, PA, et al. IMP3, NESP55, TTF-1 and CDX2 serve as an immunohistochemical panel in the distinction among small-cell carcinoma, gastrointestinal carcinoid, and pancreatic endocrine tumor metastasized to the liver. Appl Immunohistochem Mol Morphol 2012;20:573579.CrossRefGoogle ScholarPubMed
Graham, RP, Shrestha, B, Caron, BL, et al. Islet-1 is a sensitive but not entirely specific marker for pancreatic neuroendocrine neoplasms and their metastases. Am J Surg Pathol 2013;37:399405.Google Scholar
Koo, J, Mertens, RB, Mirocha, JM, et al. Value of Islet 1 and PAX8 in identifying metastatic neuroendocrine tumors of pancreatic origin. Mod Pathol 2012;25:893901.Google Scholar
Rabban, JT, Lerwill, MF, McCluggage, WG, et al. Primary ovarian carcinoid tumors may express CDX-2: a potential pitfall in distinction from metastatic intestinal carcinoid tumors involving the ovary. Int J Gynecol Pathol 2009;28:4148.Google Scholar
Lau, SK, Luthringer, DJ, Eisen, RN. Thyroid transcription factor-1: a review. Appl Immunohistochem Mol Morphol 2002;10:97102.CrossRefGoogle ScholarPubMed
Ordóñez, NG. Thyroid transcription factor-1 is a marker of lung and thyroid carcinomas. Adv Anat Pathol 2000;7:123127.Google Scholar
Pelosi, G, Rodriguez, J, Viale, G, et al. Typical and atypical pulmonary carcinoid tumor overdiagnosed as small-cell carcinoma on biopsy specimens: a major pitfall in the management of lung cancer patients. Am J Surg Pathol 2005;29:179187.Google Scholar
Saqi, A, Alexis, D, Remotti, F, et al. Usefulness of CDX2 and TTF-1 in differentiating gastrointestinal from pulmonary carcinoids. Am J Clin Pathol 2005;123:394404.Google Scholar
Kaufmann, O, Dietel, M. Expression of thyroid transcription factor-1 in pulmonary and extrapulmonary small cell carcinomas and other neuroendocrine carcinomas of various primary sites. Histopathology 2000;36:415420.Google Scholar
Oliveira, AM, Tazelaar, HD, Myers, JL, et al. Thyroid transcription factor-1 distinguishes metastatic pulmonary from well-differentiated neuroendocrine tumors of other sites. Am J Surg Pathol 2001;25:815819.Google Scholar
Agoff, SN, Lamps, LW, Philip, AT, et al. Thyroid transcription factor-1 is expressed in extrapulmonary small cell carcinomas but not in other extrapulmonary neuroendocrine tumors. Mod Pathol 2000;13:238242.Google Scholar
Matoso, A, Singh, K, Jacob, R, et al. Comparison of thyroid transcription factor-1 expression by 2 monoclonal antibodies in pulmonary and nonpulmonary primary tumors. Appl Immunohistochem Mol Morphol 2010;18:142149.Google Scholar
Fabbro, D, Di Loreto, C, Stamerra, O, et al. TTF-1 gene expression in human lung tumours. Eur J Cancer 1996;32A:512517.Google Scholar
Folpe, AL, Gown, AM, Lamps, LW, et al. Thyroid transcription factor-1: immunohistochemical evaluation in pulmonary neuroendocrine tumors. Mod Pathol 1999;12:58.Google Scholar
Cai, YC, Banner, B, Glickman, J, et al. Cytokeratin 7 and 20and thyroid transcription factor 1 can help distinguish pulmonary from gastrointestinal carcinoid and pancreatic endocrine tumors. Hum Pathol 2001;32:10871093.Google Scholar
Sturm, N, Rossi, G, Lantuéjoul, S, et al. Expression of thyroid transcription factor-1 in the spectrum of neuroendocrine cell lung proliferations with special interest in carcinoids. Hum Pathol 2002;33:175182.Google Scholar
Zamecnik, J, Kodet, R. Value of thyroid transcription factor-1 and surfactant apoprotein A in the differential diagnosis of pulmonary carcinomas: a study of 109 cases. Virchows Arch 2002;440:353361.CrossRefGoogle ScholarPubMed
Chang, YL, Lee, YC, Liao, WY, et al. The utility and limitation of thyroid transcription factor-1 protein in primary and metastatic pulmonary neoplasms. Lung Cancer 2004;44:149157.Google Scholar
Du, EZ, Goldstraw, P, Zacharias, J, et al. TTF-1 expression is specific for lung primary in typical and atypical carcinoids: TTF-1-positive carcinoids are predominantly in peripheral location. Hum Pathol 2004;35:825831.Google Scholar
Jerome Marson, V, Mazieres, J, Groussard, O, et al. Expression of TTF-1 and cytokeratins in primary and secondary epithelial lung tumours: correlation with histological type and grade. Histopathology 2004;45:125134.CrossRefGoogle ScholarPubMed
Hiroshima, K, Iyoda, A, Shida, T, et al. Distinction of pulmonary large cell neuroendocrine carcinoma from small cell lung carcinoma: a morphological, immunohistochemical, and molecular analysis. Mod Pathol 2006;19:13581368.Google Scholar
La Rosa, S, Chiaravalli, AM, Placidi, C, et al. TTF1 expression in normal lung neuroendocrine cells and related tumors: immunohistochemical study comparing two different monoclonal antibodies. Virchows Arch 2010;457:497507.CrossRefGoogle ScholarPubMed
Tsuta, K, Kalhor, N, Wistuba, II, et al. Clinicopathological and immunohistochemical analysis of spindle-cell carcinoid tumour of the lung. Histopathology 2011;59:526536.CrossRefGoogle ScholarPubMed
Masai, K, Tsuta, K, Kawago, M, et al. Expression of squamous cell carcinoma markers and adenocarcinoma markers in primary pulmonary neuroendocrine carcinomas. Appl Immunohistochem Mol Morphol 2013;21:292297.Google Scholar
Van Lommel, A. Pulmonary neuroendocrine cells (PNEC) and neuroepithelial bodies (NEB): chemoreceptors and regulators of lung development. Paediatr Respir Rev 2001 Jun;2:171176.Google Scholar
Toshitetsu, H, Mete, O. Head and neck paragangliomas: what does the pathologist need to know. Diagnostic Histopathol 2014;20: 316325.Google Scholar
Mete, O, Lopes, MB, Asa, SL. Spindle cell oncocytomas and granular cell tumors of the pituitary are variants of pituicytoma. Am J Surg Pathol 2013;37:16941699.Google Scholar
Saeed Kamil, Z, Sinson, G, Gucer, H, Asa, SL, Mete, O. TTF-1 expressing sellar neoplasm with ependymal rosettes and oncocytic change: mixed ependymal and oncocytic variant pituicytoma. Endocr Pathol 2014;25:436438.Google Scholar
Kristensen, MH, Nielsen, S, Vyberg, M. Thyroid transcription factor-1 in primary CNS tumors. Appl Immunohistochem Mol Morphol 2011;19:437443.Google Scholar
Macchia, PE, Lapi, P, Krude, H, et al. PAX8 mutations associated with congenital hypothyroidism caused by thyroid dysgenesis. Nat Genet 1998;19:8386.CrossRefGoogle ScholarPubMed
Mansouri, A, Chowdhury, K, Gruss, P. Follicular cells of the thyroid gland require PAX8 gene function. Nat Genet 1998;19:8790.CrossRefGoogle ScholarPubMed
Plachov, D, Chowdhury, K, Walther, C, et al. PAX8 a murine paired box gene expressed in the developing excretory system and thyroid gland. Development 1990;110:643651.CrossRefGoogle ScholarPubMed
Bouchard, M, Souabni, A, Mandler, M, et al. Nephric lineage specification by PAX2 and PAX8. Genes Dev 2002;16:29582970.Google Scholar
Marx, A, Shimosato, Y, Kuo, TT, et al. Thymic neuroendocrine tumours. In Travis, WD, Brambilla, E, Muller-Hermelink, HK, Harris, CC, eds. World Health Organization Classification of Tumours: Pathology and Genetics of Tumours of the Lung, Pleura, Thymus and Heart, 3rd edn. Lyon: International Agency for Research on Cancer, 2004:188195.Google Scholar
Matias-Guiu, X, DeLellis, R, Moley, JF, et al. Medullary thyroid carcinoma. In DeLellis, RA, Lloyd, RV, Heitz, PU, Eng, C, eds. World Health Organization Classification of Tumours: Pathology and Genetics of Tumours of Endocrine Organs. Lyon: International Agency for Research on Cancer, 2004:8691.Google Scholar
Long, KB, Srivastava, A, Hirsch, MS, et al. PAX8 expression in well-differentiated pancreatic endocrine tumors: correlation with clinicopathologic features and comparison with gastrointestinal and pulmonary carcinoid tumors. Am J Surg Pathol 2010;34:723729.CrossRefGoogle ScholarPubMed
Sangoi, AR, Ohgami, RS, Pai, RK, et al. PAX8 expression reliably distinguishes pancreatic well-differentiated neuroendocrine tumors from ileal and pulmonary well-differentiated neuroendocrine tumors and pancreatic acinar cell carcinoma. Mod Pathol 2011;24:412424.Google Scholar
Ozcan, A, Shen, SS, Hamilton, C, et al. PAX 8 expression in non-neoplastic tissues, primary tumors, and metastatic tumors: a comprehensive immunohistochemical study. Mod Pathol 2011;24:751764.Google Scholar
Haynes, CM, Sangoi, AR, Pai, RK. PAX8 is expressed in pancreatic well-differentiated neuroendocrine tumors and in extrapancreatic poorly differentiated neuroendocrine carcinomas in fine-needle aspiration biopsy specimens. Cancer Cytopathol 2011;119:193201.Google Scholar
Laury, AR, Perets, R, Piao, H, et al. A comprehensive analysis of PAX8 expression in human epithelial tumors. Am J Surg Pathol 2011;35:816826.CrossRefGoogle ScholarPubMed
Tacha, D, Zhou, D, Cheng, L. Expression of PAX8 in normal and neoplastic tissues: a comprehensive immunohistochemical study. Appl Immunohistochem Mol Morphol 2011;19:293299.Google Scholar
Lorenzo, PI, Jimenez Moreno, CM, Delgado, I, et al. Immunohistochemical assessment of Pax8 expression during pancreatic islet development and in human neuroendocrine tumors. Histochem Cell Biol 2011;136:595607.Google Scholar
Jimenez Moreno, CM, Lorenzo, PI, Delgado, I, et al. Pax8 detection in well-differentiated pancreatic endocrine tumors: how reliable is it? Am J Surg Pathol 2011;35: 19061907.Google Scholar
Ordóñez, NG. Value of PAX 8 immunostaining in tumor diagnosis: a review and update. Adv Anat Pathol 2012;19:140151.Google Scholar
Rivera, M, Sang, C, Gerhard, R, et al. Anaplastic thyroid carcinoma: morphologic findings and PAX-8 expression in cytology specimens. Acta Cytol 2010;54:668672.Google Scholar
Bishop, JA, Sharma, R, Westra, WH. PAX8 immunostaining of anaplastic thyroid carcinoma: a reliable means of discerning thyroid origin for undifferentiated tumors of the head and neck. Hum Pathol 2011;42:18731877.Google Scholar
Smallridge, RC, Ain, KB, Asa, SL, et al. for the American Thyroid Association Anaplastic Thyroid Cancer Guidelines Taskforce. American Thyroid Association guidelines for management of patients with anaplastic thyroid cancer. Thyroid 2012;22:11041139.Google Scholar
DeLellis, RA, Lloyd, RV, Heitz, PU, Eng, C, eds. World Health Organization Classification of Tumours: Pathology and Genetics of Tumours of Endocrine Organs. Lyon: International Agency for Research on Cancer, 2004.Google Scholar
Agaimy, A, Erlenbach-Wunsch, K, Konukiewitz, B, et al. ISL1 expression is not restricted to pancreatic well-differentiated neuroendocrine neoplasms, but is also commonly found in well and poorly differentiated neuroendocrine neoplasms of extrapancreatic origin. Mod Pathol 2013;26:9951003.CrossRefGoogle Scholar
Thor, S, Ericson, J, Brannstrom, T, et al. The homeodomain LIM protein Isl-1 is expressed in subsets of neurons and endocrine cells in the adult rat. Neuron 1991;7:881889.Google Scholar
Park, JY, Hong, SM, Klimstra, DS, et al. Pdx1 expression in pancreatic precursor lesions and neoplasms. Appl Immunohistochem Mol Morphol 2011;19:444449.CrossRefGoogle ScholarPubMed
Fendrich, V, Ramerth, R, Waldmann, J, et al. Sonic hedgehog and pancreatic-duodenal homeobox 1 expression distinguish between duodenal and pancreatic gastrinomas. Endocr Relat Cancer 2009;16:613622.Google Scholar
Zhang, L, Smyrk, TC, Oliveira, AM, et al. KIT is an independent prognostic marker for pancreatic endocrine tumors: a finding derived from analysis of islet cell differentiation markers. Am J Surg Pathol 2009;33:15621569.Google Scholar
Rasmussen, P, Lindholm, J. Ectopic pituitary adenomas. Clin Endocrinol (Oxf) 1979 Jul;11:6974.Google Scholar
Nishiike, S, Tatsumi, KI, Shikina, T, Masumura, C, Inohara, H. Thyroid-stimulating hormone-secreting ectopic pituitary adenoma of the nasopharynx. Auris Nasus Larynx 2014;pii:S0385-814600115.Google Scholar
Thompson, LD, Seethala, RR, Müller, S. Ectopic sphenoid sinus pituitary adenoma (ESSPA) with normal anterior pituitary gland: a clinicopathologic and immunophenotypic study of 32 cases with a comprehensive review of the English literature. Head Neck Pathol 2012;6:75100.Google Scholar
Liu, B, Zhuang, Z, Luo, J, Wang, Y. A case report of an ectopic clival growth hormone adenoma associated with an empty sella and a review of the literature. Clin Neurol Neurosurg 2013;115:25332536.Google Scholar
Wu, XF, Wen, M. CT finding of parapharyngeal space ectopic pituitary adenoma: case report and review of literature. Head Neck 2015;37:E120E124.Google Scholar
Al-Bazzaz, S, Karamchandani, J, Mocarski, E, Horvath, E, Rotondo, F, Kovacs, K. Ectopic prolactin-producing pituitary adenoma in a benign ovarian cystic teratoma. Endocr Pathol 2014;25:321323.Google Scholar
Mete, O, Asa, SL. Clinicopathological correlations in pituitary adenomas. Brain Pathol 2012;22:443453.Google Scholar
Mete, O, Asa, SL. Therapeutic implications of accurate classification of pituitary adenomas. Semin Diagn Pathol 2013;30:158640.Google Scholar
Nonaka, D. Study of parathyroid transcription factor Gcm2 expression in parathyroid lesions. Am J Surg Pathol 2011;35:145151.Google Scholar
Ordóñez, NG. Value of GATA3 immunostaining in the diagnosis of parathyroid tumors. Appl Immunohistochem Mol Morphol 2014;22:756761.Google Scholar
Kamp, K, Feelders, RA, van Adrichem, RC, et al. Parathyroid hormone-related peptide (PTHrP) secretion by gastroenteropancreatic neuroendocrine tumors (GEP-NETs): clinical features, diagnosis, management, and follow-up. J Clin Endocrinol Metab 2014;99:30603069.Google Scholar
So, JS, Epstein, JI. GATA3 expression in paragangliomas: a pitfall potentially leading to misdiagnosis of urothelial carcinoma. Mod Pathol 2013;26:13651370.CrossRefGoogle ScholarPubMed
Yeh Chu, PG, Weiss, LM. Keratin expression in human tissues and neoplasms. Histopathology 2002;40:403439.Google Scholar
Yeh Tot, T. Cytokeratins 20 and 7 as biomarkers: usefulness in discriminating primary from metastatic adenocarcinoma. Eur J Cancer 2002;38:758763.Google Scholar
Moll, R, Lowe, A, Laufer, J, et al. Cytokeratin 20 in human carcinomas. A new histodiagnostic marker detected bymonoclonal antibodies. Am J Pathol 1992;140:427447.Google ScholarPubMed
Miettinen, M. Keratin 20: immunohistochemical marker for gastrointestinal, urothelial, and Merkel cell carcinomas. Mod Pathol 1995;8:384388.Google Scholar
Chu, P, Wu, E, Weiss, LM. Cytokeratin 7 and cytokeratin 20 expression in epithelial neoplasms: a survey of 435 cases. Mod Pathol 2000;13:962972.Google Scholar
Broers, JL, Ramaekers, FC, Rot, MK, et al. Cytokeratins in different types of human lung cancer as monitored by chain-specific monoclonal antibodies. Cancer Res 1988;48:32213229.Google Scholar
Alsaad, KO, Serra, S, Schmitt, A, et al. Cytokeratins 7 and 20 immunoexpression profile in goblet cell and classical carcinoids of appendix. Endocr Pathol 2007;18:1622.Google Scholar
Jiang, Y, Long, H, Wang, W, et al. Clinicopathological features and immunoexpression profiles of goblet cell carcinoid and typical carcinoid of the appendix. Pathol Oncol Res 2011;17:127132.Google Scholar
Di Loreto, C, Di Lauro, V, Puglisi, F, et al. Immunocytochemical expression of tissue specific transcription factor-1 in lung carcinoma. J Clin Pathol 1997;50:3032.Google Scholar
Byrd-Gloster, AL, Khoor, A, Glass, LF, et al. Differential expression of thyroid transcription factor 1 in small cell lung carcinoma and Merkel cell tumor. Hum Pathol 2000;31:5862.Google Scholar
Hanly, AJ, Elgart, GW, Jorda, M, et al. Analysis of thyroid transcription factor-1 and cytokeratin 20 separates Merkel cell carcinoma from small cell carcinoma of lung. J Cutan Pathol 2000;27:118120.CrossRefGoogle ScholarPubMed
Ordóñez, NG. Value of thyroid transcription factor-1 immunostaining in distinguishing small cell lung carcinomas from other small cell carcinomas. Am J Surg Pathol 2000;24:12171223.Google Scholar
Cheuk, W, Kwan, MY, Suster, S, et al. Immunostaining for thyroid transcription factor 1 and cytokeratin 20 aids the distinction of small cell carcinoma from Merkel cell carcinoma, but not pulmonary from extrapulmonary small cell carcinomas. Arch Pathol Lab Med 2001;125:228231.Google Scholar
Shin, SJ, DeLellis, RA, Rosen, PP. Small cell carcinoma of the breast: additional immunohistochemical studies. Am J Surg Pathol 2001;25:831832.Google Scholar
Leech, SN, Kolar, AJ, Barrett, PD, et al. Merkel cell carcinoma can be distinguished from metastatic small cell carcinoma using antibodies to cytokeratin 20 and thyroid transcription factor 1. J Clin Pathol 2001;54:727729.Google Scholar
Sturm, N, Lantuéjoul, S, Laverriere, MH, et al. Thyroid transcription factor 1 and cytokeratins 1, 5, 10, 14 (34betaE12) expression in basaloid and large-cell neuroendocrine carcinomas of the lung. Hum Pathol 2001;32:918925.Google Scholar
Chhieng, DC, Cangiarella, JF, Zakowski, MF, et al. Use of thyroid transcription factor 1, PE-10, and cytokeratins 7 and 20 in discriminating between primary lung carcinomas and metastatic lesions in fine-needle aspiration biopsy specimens. Cancer 2001;93:330336.Google Scholar
Wu, M, Wang, B, Gil, J, et al. p63 and TTF-1 immunostaining. A useful marker panel for distinguishing small cell carcinoma of lung from poorly differentiated squamous cell carcinoma of lung. Am J Clin Pathol 2003;119:696702.Google Scholar
Myong, NH. Thyroid transcription factor-1 (TTF-1) expression in human lung carcinomas: its prognostic implication and relationship with wxpressions of p53 and Ki-67 proteins. J Korean Med Sci 2003;18:494500.Google Scholar
Yamamoto, J, Ohshima, K, Ikeda, S, et al. Primary esophageal small cell carcinoma with concomitant invasive squamous cell carcinoma or carcinoma in situ. Hum Pathol 2003;34: 11081115.Google Scholar
Yang, DT, Holden, JA, Florell, SR. CD117, CK20, TTF-1, and DNA topoisomerase II-alpha antigen expression in small cell tumors. J Cutan Pathol 2004;31:254261.CrossRefGoogle ScholarPubMed
Liu, J, Farhood, A. Immunostaining for thyroid transcription factor-1 on fine-needle aspiration specimens of lung tumors: a comparison of direct smears and cell block preparations. Cancer 2004;102:109114.Google Scholar
Nagao, T, Gaffey, TA, Olsen, KD, et al. Small cell carcinoma of the major salivary glands: clinicopathologic study with emphasis on cytokeratin 20 immunoreactivity and clinical outcome. Am J Surg Pathol 2004;28:762770.Google Scholar
Soriano, P, Navarro, S, Gil, M, et al. Small-cell carcinoma of the urinary bladder. A clinico-pathological study of ten cases. Virchows Arch 2004;445:292297.Google Scholar
McCluggage, WG, Oliva, E, Connolly, LE, et al. An immunohistochemical analysis of ovarian small cell carcinoma of hypercalcemic type. Int J Gynecol Pathol 2004;23:330336.Google Scholar
Liu, J, Farhood, A. Thyroid transcription factor-1 immunocytochemical staining of pleural fluid cytocentrifuge preparations for detection of small cell lung carcinoma. Acta Cytol 2004;48:635640.Google Scholar
Zhang, H, Liu, J, Cagle, PT, et al. Distinction of pulmonary small cell carcinoma from poorly differentiated squamous cell carcinoma: an immunohistochemical approach. Mod Pathol 2005;18:111118.Google Scholar
Nassar, H, Albores-Saavedra, J, Klimstra, DS. High-grade neuroendocrine carcinoma of the ampulla of Vater: a clinicopathologic and immunohistochemical analysis of 14 cases. Am J Surg Pathol 2005;29:588594.Google Scholar
Llombart, B, Monteagudo, C, Lopez-Guerrero, JA, et al. Clinicopathological and immunohistochemical analysis of 20 cases of Merkel cell carcinoma in search of prognostic markers. Histopathology 2005;46:622634.Google Scholar
Jones, TD, Kernek, KM, Yang, XJ, et al. Thyroid transcription factor 1 expression in small cell carcinoma of the urinary bladder: an immunohistochemical profile of 44 cases. Hum Pathol 2005;36:718723.Google Scholar
Wu, M, Szporn, AH, Zhang, D, et al. Cytology applications of p63 and TTF-1 immunostaining in differential diagnosis of lung cancers. Diagn Cytopathol 2005;33:223227.Google Scholar
Bobos, M, Hytiroglou, P, Kostopoulos, I, et al. Immunohistochemical distinction between Merkel cell carcinoma and small cell carcinoma of the lung. Am J DermatoPathol 2006;28:99104.Google Scholar
Yao, JL, Madeb, R, Bourne, P, et al. Small cell carcinoma of the prostate: an immunohistochemical study. Am J Surg Pathol 2006;30:705712.CrossRefGoogle ScholarPubMed
Kalhor, N, Zander, DS, Liu, J. TTF-1 and p63 for distinguishing pulmonary small-cell carcinoma from poorly differentiated squamous cell carcinoma in previously Pap-stained cytologic material. Mod Pathol 2006;19:11171123.CrossRefGoogle ScholarPubMed
Yun, JP, Zhang, MF, Hou, JH, et al. Primary small cell carcinoma of the esophagus: clinicopathological and immunohistochemical features of 21 cases. BMC Cancer 2007;7:38.Google Scholar
Carlson, JW, Nucci, MR, Brodsky, J, et al. Biomarker-assisted diagnosis of ovarian, cervical and pulmonary small cell carcinomas: the role of TTF-1, WT-1 and HPV analysis. Histopathology 2007;51:305312.Google Scholar
El Demellawy, D, Khalifa, MA, Ismiil, N, et al. Primary colorectal small cell carcinoma: a clinicopathological and immunohistochemical study of 10 cases. Diagn Pathol 2007;2:35.Google Scholar
Alijo Serrano, F, Sanchez-Mora, N, Angel Arranz, J, et al. Large cell and small cell neuroendocrine bladder carcinoma: immunohistochemical and outcome study in a single institution. Am J Clin Pathol 2007;128:733739.Google Scholar
Kargi, A, Gurel, D, Tuna, B. The diagnostic value of TTF-1, CK5/6, and p63 immunostaining in classification of lung carcinomas. Appl Immunohistochem Mol Morphol 2007;15: 415420.Google Scholar
Lu, J, Xue, LY, Lu, N, et al. Superficial primary small cell carcinoma of the esophagus: clinicopathological and immunohistochemical analysis of 15 cases. Dis Esophagus 2010;23:153159.Google Scholar
Wang, W, Epstein, JI. Small cell carcinoma of the prostate. A morphologic and immunohistochemical study of 95 cases. Am J Surg Pathol 2008;32:6571.Google Scholar
Ralston, J, Chiriboga, L, Nonaka, D. MASH1: a useful marker in differentiating pulmonary small cell carcinoma from Merkel cell carcinoma. Mod Pathol 2008;21:13571362.Google Scholar
McCluggage, WG, Kennedy, K, Busam, KJ. An immunohistochemical study of cervical neuroendocrine carcinomas: neoplasms that are commonly TTF1 positive and which may express CK20 and P63. Am J Surg Pathol 2010;34:525532.Google Scholar
Li, AF, Li, AC, Hsu, CY, et al. Small cell carcinomas in gastrointestinal tract: immunohistochemical and clinicopathological features. J Clin Pathol 2010;63:620625.Google Scholar
Lewis, JS Jr., Duncavage, E, Klonowski, PW. Oral cavity neuroendocrine carcinoma: a comparison study with cutaneous Merkel cell carcinoma and other mucosal head and neck neuroendocrine carcinomas. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2010;110:209217.Google Scholar
Sidiropoulos, M, Hanna, W, Raphael, SJ, et al. Expression of TdT in Merkel cell carcinoma and small cell lung carcinoma. Am J Clin Pathol 2011;135:831838.Google Scholar
Fernández-Aceñero, MJ, Cordova, S, Manzarbeitia, F, et al. Immunohistochemical profile of urothelial and small cell carcinomas of the bladder. Pathol Oncol Res 2011;17:519523.Google Scholar
Thompson, S, Cioffi-Lavina, M, Chapman-Fredricks, J, et al. Distinction of high-grade neuroendocrine carcinoma/small cell carcinoma from conventional urothelial carcinoma of urinary bladder: an immunohistochemical approach. Appl Immunohistochem Mol Morphol 2011;19:395399.Google Scholar
Quinn, AM, Blackhall, F, Wilson, G, et al. Extrapulmonary small cell carcinoma: a clinicopathological study with identification of potential diagnostic mimics. Histopathology 2012;61:454464.Google Scholar
Holzinger, A, Dingle, S, Bejarano, PA, et al. Monoclonal antibody to thyroid transcription factor-1: production, characterization, and usefulness in tumor diagnosis. Hybridoma 1996;15:4953.Google Scholar
Jakobsen, AM, Ahlman, H, Kolby, L, et al. NESP55, a novel chromogranin-like peptide, is expressed in endocrine tumours of the pancreas and adrenal medulla but not in ileal carcinoids. Br J Cancer 2003;88:17461754.Google Scholar
Srivastava, A, Padilla, O, Fischer-Colbrie, R, et al. Neuroendocrine secretory protein-55 (NESP-55) expression discriminates pancreatic endocrine tumors and pheochromocytomas from gastrointestinal and pulmonary carcinoids. Am J Surg Pathol 2004;28: 13711378.Google Scholar
Ischia, R, Lovisetti-Scamihorn, P, Hogue-Angeletti, R, et al. Molecular cloning and characterization of NESP55, a novel chromogranin-like precursor of a peptide with 5-HT1B receptor antagonist activity. J Biol Chem 1997;272:1165711662.Google Scholar
Gucer, H, Mete, O. Endobronchial gangliocytic paraganglioma: not all keratin-positive endobronchial neuroendocrine neoplasms are pulmonary carcinoids. Endocr Pathol 2014 Sep;25:356358.Google Scholar
Mete, O, Asa, SL. Composite medullary and papillary thyroid carcinoma in a patient with MEN2B: case report and review of c-cell lesions of the thyroid. Pathol Case Rev 2009;14:208213.Google Scholar
Mete, O, Asa, SL. Pitfalls in the diagnosis of follicular epithelial proliferations of the thyroid. Adv Anat Pathol 2012;19:363373.Google Scholar
Alumets, J, Alm, P, Falkmer, S, et al. Immunohistochemical evidence of peptide hormones in endocrine tumors of the rectum. Cancer 1981;48:24092415.Google Scholar
O'Briain, DS, Dayal, Y, DeLellis, RA, et al. Rectal carcinoids as tumors of the hindgut endocrine cells: a morphological and immunohistochemical analysis. Am J Surg Pathol 1982;6:131142.Google Scholar
Yang, K, Ulich, T, Cheng, L, et al. The neuroendocrine products of intestinal carcinoids. An immunoperoxidase study of 35 carcinoid tumors stained for serotonin and eight polypeptide hormones. Cancer 1983;51:19181926.Google Scholar
Anlauf, M, Hamscher, G, Weihe, E, et al. Xenin-immunoreactive cells and extractable xenin in neuroendocrine tumors of duodenal origin. Gastroenterology 2002;123:16161626.Google Scholar
Federspiel, BH, Burke, AP, Sobin, LH, et al. Rectal and colonic carcinoids. A clinicopathologic study of 84 cases. Cancer 1990;65:135140.Google Scholar
Burke, AP, Thomas, RM, Elsayed, AM, et al. Carcinoids of the jejunum and ileum: an immunohistochemical and clinicopathologic study of 167 cases. Cancer 1997;79:10861093.Google Scholar
Xu, B, Chetty, R, Pérez-Ordoñez, B. Neuroendocrine neoplasms of the head and neck: some suggestions for the new WHO classification of head and neck tumors. Head Neck Pathol 2014 Mar;8:2432.Google Scholar
Agaimy, A, Lell, M, Schaller, T, Märkl, B, Hornung, J. “Neuroendocrine” middle ear adenoma: consistent expression of the transcription factor ISL1 (Islet-1) further supports their neuroendocrine derivation. Histopathology 2015;66:182191.Google Scholar
Doglioni, C, Gambacorta, M, Zamboni, G, et al. Immunocytochemical localization of progesterone receptors in endocrine cells of the human pancreas. Am J Pathol 1990;137:9991005.Google Scholar
Viale, G, Doglioni, C, Gambacorta, M, et al. Progesterone receptor immunoreactivity in pancreatic endocrine tumors. An immunocytochemical study of 156 neuroendocrine tumors of the pancreas, gastrointestinal and respiratory tracts, and skin. Cancer 1992;70:22682277.Google Scholar
Wang, SC, Parekh, JR, Zuraek, MB, et al. Identification of unknown primary tumors in patients with neuroendocrine liver metastases. Arch Surg 2010;145:276280.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×