Review Open Access
Copyright ©2010 Baishideng. All rights reserved.
World J Gastroenterol. Apr 14, 2010; 16(14): 1713-1719
Published online Apr 14, 2010. doi: 10.3748/wjg.v16.i14.1713
Pathologic research update of colorectal neuroendocrine tumors
Shu-Juan Ni, Wei-Qi Sheng, Xiang Du, Department of Pathology, Fudan University Cancer Hospital, Shanghai 200032, China; Department of Oncology, Shanghai Medical School, Fudan University, Shanghai 200032, China
Author contributions: Ni SJ, Sheng WQ and Du X all contributed to the data research for this review; Ni SJ conducted the literature review and wrote the manuscript; Sheng WQ developed the initial plan; Du X provided guidance throughout the preparation of this manuscript and corrected the final version.
Supported by The Science and Technology Commission of Shanghai Municipality
Correspondence to: Xiang Du, MD, PhD, Department of Pathology, Fudan University Cancer Hospital, 270 Dongan Road, Shanghai 200032, China. dx2008cn@yahoo.com.cn
Telephone: +86-21-64175590-8911 Fax: +86-21-64170067
Received: January 25, 2010
Revised: February 20, 2010
Accepted: February 27, 2010
Published online: April 14, 2010

Abstract

Colorectal neuroendocrine tumors (NETs) originate from neuroendocrine cells in the intestinal tract, and represent a small area within oncology, but one which has provided increasing new data during the past years. Although the World Health Organization has determined clinical and histological features to predict prognosis for such tumors, they may not be valid on an individual basis. We aim to give an overview of the recent findings with regard to pathology, molecular genetics and diagnosis of NETs.

Key Words: Neuroendocrine tumors, Carcinoid, Colorectal, World Health Organization classification, Tumor-node-metastases



INTRODUCTION

Neuroendocrine (NE) cells are distributed throughout the human body, including the gastrointestinal (GI) tract, pancreas, lung, thyroid, adrenal gland and many other organs[1-4]. The GI tract has the largest population of NE cells[5]. However, neuroendocrine tumors (NETs) of the colon and rectum are rare. Results from analyses of the Surveillance, Epidemiology and End Results database demonstrated that the age-adjusted incidence of carcinoids of the small intestine and digestive system has increased by 460% and 720%, respectively, in the past 30 years due to, at least in part, the improvements in diagnostic technology such as endoscopy, as well as doctors’ increased awareness[6]. In 2003, Modlin et al[7] reported that the incidence of NETs in the GI tract was 2.5-5 cases per 100 000 population annually. Males are more often affected than females, with a proportion of 1.8:1[8]. In the large intestine, NETs are more commonly found in the rectum (54%), then in the cecum (20%), sigmoid colon (7.5%), rectosigmoid colon (5.5%) and ascending colon (5%)[9].

Colon NETs usually appear as large tumors and often already have regional lymph nodes or liver metastasis at the time of diagnosis, therefore the prognosis is poor. On the other hand, NETs occurring in the rectum are often diagnosed incidentally during colonoscopy, therefore they are typically small, localized, non-functioning tumors with rare metastasis, probably as a result of early detection[6,8,10,11].

CLASSIFICATION

Colorectal NETs are traditionally classified as typical carcinoid, atypical carcinoid and undifferentiated cancer according to the differentiation of the disease[12,13]. The term carcinoid can no longer characterize the entire morphologic and biologic spectrum of neoplasms of the disseminated NE cell system. The updated World Health Organization (WHO) classification of 2000 adopted the neutral and inclusive terms, tumor and carcinoma. In this classification, it is clearly explained how to characterize well-differentiated NETs (benign behavior or uncertain malignant potential), well-differentiated NE carcinoma (low-grade malignancy), and poorly-differentiated NE carcinoma of high-grade malignancy [small cell carcinomas (SCCs)][14,15]. The morphologic/biologic criteria including tumor size, angioinvasion, proliferative activity, histological differentiation, metastasis, invasion, and hormonal activity (association with clinical syndromes or diseases) have also been added (Table 1).

Table 1 Classification of NETs of the colon and rectum[14,15].
Well-differentiated neuroendocrine tumor (carcinoid)
Benign: Non-functioning, confined to mucosa-submucosa, non-angioinvasive, < 1 cm in size (ileum) or ≤ 2 cm colon and rectum
Serotonin-producing tumor
Enteroglucagon-producing tumor
Benign or low-grade malignant (uncertain malignant potential): non-functioning, confined to mucosa-submucosa, angioinvasion, or < 1 cm in size (ileum) or ≤ 2 cm colon and rectum
Serotonin-producing tumor
Enteroglucagon-producing tumor
Well-differentiated neuroendocrine carcinoma (malignant carcinoid)
Low-grade malignant: invasion of the muscularis propria and beyond or metastases
Non-functioning or functioning serotonin-producing carcinoma (with carcinoid syndrome)
Non-functioning enteroglucagon-producing carcinoma
Poorly-differentiated neuroendocrine carcinoma
High-grade malignant

Well-differentiated NE tumor was referred to as typical carcinoid historically, and well-differentiated NE carcinomas were termed atypical carcinoid. Although this framework is helpful, lesions of uncertain behavior are poorly defined. In the WHO classification, SCC is synonymous with the term “poorly-differentiated NE carcinoma”, but there is no clear differentiation between large cell NE carcinomas (LCNECs) and mixed endocrine-glandular neoplasms. No coincident repeat diagnosis could be made by different pathologists or by different institutions, although the WHO classification of colorectal NETs has been used for several years. The main reason is that the WHO classification has not been closely adhered to[1]. Pathological recognition of these tumors is critical, as the treatment strategy, particularly chemotherapy for a particular subtype, largely depends on the underlying pathology[16,17].

Well-differentiated NE tumor-carcinoid

Colorectal carcinoids account for approximately 6% of all GI NETs, often accompanied by chronic inflammatory disease such as ulcerative colitis or Crohn’s disease. The tumors are usually multiple when complicated by these diseases[18]. In Crohn’s disease, the incidence of carcinoids may increase, by a recent estimation of 15-fold[19,20]. In most instances, colorectal carcinoids present without obvious signs or symptoms and remain undetectable for years[8]. Most of the diagnoses are made incidentally at the time of surgery for other abdominal disorders. Their relatively high incidence in large autopsy series has provided evidence for this observation[21].

The tumors with NE differentiation have classical histological architecture of trabecular, insular, or ribbon-like cell clusters, and have no or minimal cellular pleomorphism and sparse mitoses[8,22,23]. Colorectal carcinoids display moderate neurofilament staining. They stain positive for chromogranin A (CgA) in more than 70% of cases, positive for neuron specific enolase (NSE) in more than 50% of cases, and prostatic acid phosphatase is expressed in 80%-100% of cases. However, the staining pattern is variable[8]. Other markers such as synaptophysin (Syn), somatostatin, 5-HT, or CD56 may be present as well. The Ki-67 index is higher in carcinoids of a size more than 5 mm, as compared to those sized less than 5 mm in diameter[24].

Rectal carcinoids are usually discovered incidentally during colonoscopy examinations. The tumors are usually small, non-functional and without regional or distant metastasis[25,26]. The 5-year overall survival rate of patients with rectal carcinoids is 88%. However, the prognosis of colon carcinoids is worse than that of its rectal counterpart[8,10,26,27]. There are data showing that there is a high frequency of combined adenocarcinoma and NETs in the proximal colon[28,29]. Perhaps this is one of the reasons explaining the worse prognosis in colon carcinoids than for their rectal counterpart.

Well-differentiated NE carcinoma-malignant carcinoid

Malignant carcinoid was termed as atypical carcinoid, historically. Only a few cases have been reported as malignant carcinoid of the large intestine due to uncertainty of its definition.

Well-differentiated NE carcinomas have histological features and a biological behavior that fall between well-differentiated NE tumors and poorly-differentiated NE carcinomas[8]. These are aggressive lesions and represent forms poorly differentiated from carcinoid with increased mitotic activity and presence of necrosis. Mitoses range from 1 to 10/10 high power field (hpf). The presence of necrosis serves as an important character of malignant carcinoid[8,30,31]. The cells range in appearance from uniform, large, polygonal, or fusiform types with abundant eosinophilic granular cytoplasm and round to oval nuclei similar to the cells seen in typical carcinoid, to pleomorphic cells with scanty cytoplasm and hyperchromatic variably sized and shaped nuclei[32]. The tumors are nonargentaffinic but strongly argyrophilic and they are stained with the usual immunohistochemical markers of NE cell differentiation.

There are few reports of the clinical outcome of colorectal malignant carcinoids. It has been reported, however, that a patient with an atypical carcinoid of the lung has a significantly worse prognosis than those with typical carcinoids[33,34].

Poorly-differentiated NE carcinoma (small cell carcinoma)

Colorectal SCCs constitute 0.2%-0.8% of all colorectal tumors, mostly located in the right colon[1,35]. They are clinically aggressive; therefore have extremely poor prognosis, even when diagnosed at an early stage. Most patients present with overt distant metastases. Sometimes they are discovered in the background of colon inflammatory diseases, or in the background of NE cell proliferation[36,37].

Colorectal SCCs are identical to lung SCCs morphologically. These small blue cell tumors present the character of densely packed, small, and oval-, spindle-, fusiform-shaped anaplastic cells with minimal amounts of cytoplasm and granular nuclear chromatin. The size of the nuclei measures approximately twice the size of mature lymphocytes[38]. Solid sheets, nests, and rosettes as well as ribbon-like structures composed of small cells and intermediate cells form, which all exhibit more than 10/10 hpf mitoses[1].

In cases with classic histological features, it is unnecessary to demonstrate NE differentiation to establish the diagnosis. However, further analysis may be necessary in puzzling cases. SCCs typically express CgA, Syn, NSE, and CD56 by immunohistochemistry. Pan cytokeratin (AE1/AE3) and low molecular weight keratin are also reported to be positive in all SCCs[1,39]. Shida et al[40] reported that 70% of GI SCCs expressed human achaete-scute homologue gene-1 protein (hASH1), which is usually absent in normal GI NE cells, carcinoid and adenocarcinoma. The sensibility and specificity of hASH1 is better than other classic markers, such as CgA and Syn, and this protein may serve as a new biomarker for GI SCC diagnosis. There are also some reports demonstrating that cytokeratin is positive in these tumors and the Ki-67 index beyond 75% is often seen in SCCs[1].

As mentioned above, the prognosis of patients with GI SCCs is usually extremely poor. Most patients present with regional or overt distant metastases. Up to 80% of patients already have regional lymph nodes and/or distant metastases at the time of diagnosis[41]. When tumor size is larger than 5 cm, the median survival time for patients is usually less than 4 mo, and the median survival of those with smaller tumor is around 12 mo[1].

LCNEC

LCNECs are rare, poorly-differentiated NE carcinomas. LCNECs are amongst the worst studied group of colorectal NETs and their diagnostic criteria are briefly described in many standard texts[14]. The features of these tumors are similar to their counterpart in the lung[42]. They are malignant neoplasms composed of large cells structured in organoid, nested, trabecular, rosette-like, and palisading patterns that suggest NE differentiation. Compared to SCCs, LCNECs have more cytoplasm, nuclei are more vesicular, and nucleoli are prominent[42]. Focal necrosis is often observed. These tumors exist alone or associated with adjacent adenomas or conventional adenocarcinomas[43]. LCNECs are defined through both NE morphology and immunohistochemical positivity of NE markers.

CK20 is expressed in colorectal LCNECs; a generally acknowledged marker in adenocarcinoma which is rarely found in other NETs[44,45]. This phenomenon may indicate that there is some relationship between LCNECs and adenocarcinomas.

LCNECs have a similar prognosis to colorectal SCCs. Kumarasinghe et al[46] suggested that LCNECs should be included in the poorly-differentiated NE carcinoma category based on their poor prognosis.

Mixed endocrine-glandular neoplasm

Mixed endocrine-glandular neoplasms constitute a heterogeneous group of rare neoplasms. These tumors are composed of at least two distinct tumor populations, with the NE part containing at least 30% of obviously endocrine cells[14,47,48]. The degree of NE cell differentiation varies between tumors and between different tumor areas. The most common component is carcinoid, while poorly-differentiated NE type also exists, and another component can be adenocarcinoma or squamous carcinoma[14,15,49]. The NE component is usually well-differentiated, and easily recognized by its suggestive histological features; the NE nature of the tumor cells can be verified by the immunodetection of specific NE markers (such as CgA and Syn). Especially when the NE component is poorly differentiated, the demonstration of NE markers to confirm the diagnosis is needed[50]. More often, the diagnosis cannot be made until the tumors are stained with NE cell markers when the endocrine cells are inconspicuous and the quantity present is not obvious[1].

Because of their rarity and unusual presentation, the optimal strategy of management of mixed endocrine tumors is largely unknown. The more aggressive component of the mixed endocrine tumors must be taken into account when considering their treatment. Mixed tumors containing a well-differentiated NE component and an adenocarcinoma component are suggested to be treated as adenocarcinoma. Mixed tumors containing a poorly-differentiated NE component must be treated as poorly-differentiated NE carcinomas[50]. Some researchers report mixed NETs as having worse prognosis, thus the ascertainment of the NE component is important to predict the prognosis[48].

MOLECULAR GENETICS OF COLORECTAL NETs

The molecular mechanisms of NET tumorigenesis are unclear; however, these aspects have been focused on in many recent reports[51]. Increased knowledge of the molecular background for the development of NETs may improve the management of these tumors in the future. A number of genetic syndromes including multiple endocrine neoplasia syndrome-type 1 (MEN1), vov Hippel-Lindau syndrome, and neurofibromatosis-type 1 may be associated with intestinal NETs[8,52]. Some studies have demonstrated neither classic oncogenes (scr, ras, myc, fos, jun) nor suppressor genes (P53, RB) present in NETs[52,53]. However, there are reports of other gene site deletions. For example, PDCD4 was found to be absent in NETs; this is a gene which is interrelated with cell proliferation, located at 11q13 close to MEN1[54].

The microsatellite instability (MSI) caused by mismatch repair (MMR) damage is an important cause of tumorigenesis in hereditary nonpolyposis colorectal cancer (HNPCC)[55-58]. Stelow et al[59] discovered the loss of MMR genes in colorectal SCCs, but not along with HNPCC. The chromosome instability caused by the loss of MMR genes possibly follows a different pathway from MSI in SCC tumorigenesis. Arnold et al[60] studied 34 poorly-differentiated colorectal NETs, 38 well-differentiated benign or malignant fore-/midgut NETs, and 150 sporadic colorectal cancers with known MSI status and found that 20/34 (59%) colorectal NETs vs 11/38 (29%) fore-/midgut NETs were CpG island methylator phenotype (CIMP) positive. The Ki-67 index was significantly higher in poorly-differentiated colorectal NETs compared with the less malignant fore-/mid-gut NETs. However the CIMP status did not correlate with survival.

Research in NET molecular genetics should target early detection, prognosis predicting, or treatment selection. Currently, new technologies should be used to find new sensitive and specific biochemical and tissue markers for colorectal NETs.

PROPOSED TNM STAGE AND RISK FACTORS

NETs are a challenging group of diseases, and the lack of a widely accepted staging system limits the clinician’s ability to provide meaningful prognostic information to the patients. In October 2009, the 7th edition of the AJCC for the first time gave a detailed TNM description for NETs, classifying primary tumor (T), regional lymph nodes (N), and distant metastasis (M), respectively. However, no clarified TNM staging has been established for colon and rectal NETs[61].

In 2008, Landry et al[62,63] proposed a different staging system from that of conventional colorectal adenocarcinomas in the study of 4710 rectal NETs and 2459 colon NETs (Table 2). In their report [62], it was revealed that rectal NETs were the only primary malignancy in 82% of patients, 17% patients had one additional malignancy and 1% had two or more additional malignancies. The mean and median sizes of primary tumor were 1 and 0.6 cm, respectively. About 4.1% had regional lymph node metastases, and 2.4% presented with distant metastases at the time of diagnosis. The 5-year survival rates for patients with stages I and II disease were 97% and 84%, respectively. The 5-year survival rate for stage III patients was 27% with a median survival time of 45 mo. The 5-year survival rate for stage IV patients was 20% with a median survival time of 31 mo. The 10-year survival rates of the disease for stages I through IV were 91%, 56%, 14%, and 2.5%, respectively.

Table 2 Proposed staging system for rectal and colon NETs[62,63].
RectalColon
Depth of invasion and size
T1Up to and into muscularis propria, ≤ 1 cmAny depth of invasion, ≤ 1 cm
Up to muscularis propria, > 1 to ≤ 2 cmUp to or including muscularis propria, > 1 to ≤ 4 cm
T2Beyond muscularis propria, ≤ 1 cmBeyond muscularis propria, > 1 to ≤ 4 cm
Into muscularis propria, > 1 to ≤ 2 cmUp to or including muscularis propria, > 4 cm
Up to and into muscularis propria, > 2 cm
T3Invasion beyond muscularis propria, > 1 cmBeyond muscularis propria, > 4 cm
Lymph node
N0No lymph node metastasis
N1Regional lymph node metastasis
Distant metastasis
M0No distant metastasis
M1Distant metastasis
Stage
 IT1; N0; M0
IIT1; N1; M0 or T2; Any N; M0
IIIT3; N0; M0 or T3; N1; M0
IVAny T; Any N; M1

In another study[63] of colon NETs, the researchers demonstrated that there were regional lymph node metastases in 48% of patients, and distant metastases in 24% of patients at the time of diagnosis. The 5-year survival rates for stages I and II were 97% and 69%, respectively. Similarly, the 5-year survival rate for stage III disease was 21%, with a median survival time of 27 mo. The 5-year survival rate for stage IV disease was 17%, with a median survival time of 20 mo. The 10-year survival rates for stages I through IV were 92%, 47%, 15%, and 5.4%, respectively.

Some studies have suggested some risk factors of colorectal NETs related to poor outcome, such as tumor size, depth of wall penetration, presence of lymphatic or venous invasion, and mitotic rate of metastasis in colorectal NETs[64-69]. The tumor size is related to prognosis. Most rectal NETs are less than 1 cm at the time of diagnosis[70,71]. The malignancy rate increases when the tumor grows larger than 1 cm. However, for small-sized tumors less than 1 cm at detection with predominant submucosal invasion there is usually a relatively high incidence of distant metastasis, about 1.7%-3.4%[71,72]. Some reports have shown that cancer-specific survival of patients with colorectal NETs without metastasis was better than that of those with adenocarcinomas. However, the survival rate is similar between carcinoid and adenocarcinoma if the tumors have lymph node or distant metastases[65]. On the other hand, colorectal NE carcinomas behave aggressively and are associated with a worse prognosis than that of conventional colorectal adenocarcinomas of the same stage[16,39].

CONCLUSION

The complexity, heterogeneity and scarcity of NETs, rare colorectal neoplasia, account for almost no improvement of survival rates in the past 30 years. The diagnosis for colorectal NETs mostly depends on pathology with the aid of immunohistochemistry. As we have summarized here, the 2000 WHO classification of colorectal NETs provides a solid basis for localization, biology and prognosis of individual NETs, but there are still some types of tumors not properly described. More studies will be needed with regard to diagnosis, treatment and prognosis.

Footnotes

Peer reviewer: Jutta Elisabeth Lüttges, Professor, Doctor of Medicine and Professor of Pathology, Head of Department of Pathology, General Hospital Saarbrücken, Herchenbacher str. 60, Riegelsberg, 66292, Germany

S- Editor Wang YR L- Editor Logan S E- Editor Zheng XM

References
1.  Brenner B, Tang LH, Klimstra DS, Kelsen DP. Small-cell carcinomas of the gastrointestinal tract: a review. J Clin Oncol. 2004;22:2730-2739.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Li AF, Tsay SH, Liang WY, Li WY, Chen JY. Clinical significance of p16INK4a and p53 overexpression in endocrine tumors of the gastrointestinal tract. Am J Clin Pathol. 2006;126:856-865.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Staren ED, Gould VE, Warren WH, Wool NL, Bines S, Baker J, Bonomi P, Roseman DL, Economou SG. Neuroendocrine carcinomas of the colon and rectum: a clinicopathologic evaluation. Surgery. 1988;104:1080-1089.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Vilallonga R, Espín Basany E, López Cano M, Landolfi S, Armengol Carrasco M. [Neuroendocrine carcinomas of the colon and rectum. A unit's experience over six years]. Rev Esp Enferm Dig. 2008;100:11-16.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Sundler F, Böttcher G, Ekblad E, Håkanson R. [The neuroendocrine system of the gastrointestinal tract]. Nord Med. 1988;103:8-11.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Modlin IM, Oberg K, Chung DC, Jensen RT, de Herder WW, Thakker RV, Caplin M, Delle Fave G, Kaltsas GA, Krenning EP. Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol. 2008;9:61-72.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Modlin IM, Lye KD, Kidd M. A 5-decade analysis of 13,715 carcinoid tumors. Cancer. 2003;97:934-959.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Modlin IM, Kidd M, Latich I, Zikusoka MN, Shapiro MD. Current status of gastrointestinal carcinoids. Gastroenterology. 2005;128:1717-1751.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Modlin IM, Sandor A. An analysis of 8305 cases of carcinoid tumors. Cancer. 1997;79:813-829.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Kaltsas GA, Besser GM, Grossman AB. The diagnosis and medical management of advanced neuroendocrine tumors. Endocr Rev. 2004;25:458-511.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Spread C, Berkel H, Jewell L, Jenkins H, Yakimets W. Colon carcinoid tumors. A population-based study. Dis Colon Rectum. 1994;37:482-491.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Torre M, Barberis M, Barbieri B, Bonacina E, Belloni P. Typical and atypical bronchial carcinoids. Respir Med. 1989;83:305-308.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Pinchot SN, Holen K, Sippel RS, Chen H. Carcinoid tumors. Oncologist. 2008;13:1255-1269.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Solcia E, Klöppel G, Sobin LH. Histological typing of endocrine tumors. 2nd ed. Berlin: Springer 2000; 61-62, 66-67.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Solcia E, Sobin LH, Arnold R. Endocrine tumors of the colon and rectum. Pathology and genetics of tumours of the digestive system. World Healht Organization classification of tumors. Lyon: IARC Press 2000; 137-139.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Bernick PE, Klimstra DS, Shia J, Minsky B, Saltz L, Shi W, Thaler H, Guillem J, Paty P, Cohen AM. Neuroendocrine carcinomas of the colon and rectum. Dis Colon Rectum. 2004;47:163-169.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Moertel CG, Kvols LK, O'Connell MJ, Rubin J. Treatment of neuroendocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in the anaplastic variants of these neoplasms. Cancer. 1991;68:227-232.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Gledhill A, Hall PA, Cruse JP, Pollock DJ. Enteroendocrine cell hyperplasia, carcinoid tumours and adenocarcinoma in long-standing ulcerative colitis. Histopathology. 1986;10:501-508.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Freeman HJ. Colorectal cancer risk in Crohn's disease. World J Gastroenterol. 2008;14:1810-1811.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  West NE, Wise PE, Herline AJ, Muldoon RL, Chopp WV, Schwartz DA. Carcinoid tumors are 15 times more common in patients with Crohn's disease. Inflamm Bowel Dis. 2007;13:1129-1134.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Berge T, Linell F. Carcinoid tumours. Frequency in a defined population during a 12-year period. Acta Pathol Microbiol Scand A. 1976;84:322-330.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Pilichowska M, Kimura N, Ouchi A, Lin H, Mizuno Y, Nagura H. Primary hepatic carcinoid and neuroendocrine carcinoma: clinicopathological and immunohistochemical study of five cases. Pathol Int. 1999;49:318-324.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Mani S, Modlin IM, Ballantyne G, Ahlman H, West B. Carcinoids of the rectum. J Am Coll Surg. 1994;179:231-248.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Shimizu T, Tanaka S, Haruma K, Kitadai Y, Yoshihara M, Sumii K, Kajiyama G, Shimamoto F. Growth characteristics of rectal carcinoid tumors. Oncology. 2000;59:229-237.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Ramage JK, Goretzki PE, Manfredi R, Komminoth P, Ferone D, Hyrdel R, Kaltsas G, Kelestimur F, Kvols L, Scoazec JY. Consensus guidelines for the management of patients with digestive neuroendocrine tumours: well-differentiated colon and rectum tumour/carcinoma. Neuroendocrinology. 2008;87:31-39.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Massironi S, Sciola V, Peracchi M, Ciafardini C, Spampatti MP, Conte D. Neuroendocrine tumors of the gastro-entero-pancreatic system. World J Gastroenterol. 2008;14:5377-5384.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Ramage JK, Davies AH, Ardill J, Bax N, Caplin M, Grossman A, Hawkins R, McNicol AM, Reed N, Sutton R. Guidelines for the management of gastroenteropancreatic neuroendocrine (including carcinoid) tumours. Gut. 2005;54 Suppl 4:iv1-iv16.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Thomas RM, Sobin LH. Gastrointestinal cancer. Cancer. 1995;75:154-170.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Habal N, Sims C, Bilchik AJ. Gastrointestinal carcinoid tumors and second primary malignancies. J Surg Oncol. 2000;75:310-316.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Sharma R, McLeod D, Clarke SJ. Small blue cell tumors of the rectum. Case 3. Atypical carcinoid of the rectum. J Clin Oncol. 2005;23:914-915.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Valli M, Fabris GA, Dewar A, Hornall D, Sheppard MN. Atypical carcinoid tumour of the lung: a study of 33 cases with prognostic features. Histopathology. 1994;24:363-369.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Pelosi G, Rodriguez J, Viale G, Rosai J. 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;29:179-187.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  García-Yuste M, Matilla JM, Cueto A, Paniagua JM, Ramos G, Cañizares MA, Muguruza I. Typical and atypical carcinoid tumours: analysis of the experience of the Spanish Multi-centric Study of Neuroendocrine Tumours of the Lung. Eur J Cardiothorac Surg. 2007;31:192-197.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Toma C, Ionescu N, Nicolae M. Typical and atypical carcinoid tumors of lung: two distinct clinicomorphologic entities. Rom J Morphol Embryol. 1993;39:113-116.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Kim JH, Lee SH, Park J, Kim HY, Lee SI, Nam EM, Park JO, Kim K, Jung CW, Im YH. Extrapulmonary small-cell carcinoma: a single-institution experience. Jpn J Clin Oncol. 2004;34:250-254.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Rubin A, Pandya PP. Small cell neuroendocrine carcinoma of the rectum associated with chronic ulcerative colitis. Histopathology. 1990;16:95-97.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Grassia R, Bodini P, Dizioli P, Staiano T, Iiritano E, Bianchi G, Buffoli F. Neuroendocrine carcinomas arising in ulcerative colitis: coincidences or possible correlations? World J Gastroenterol. 2009;15:4193-4195.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Fenoglio-Preiser CM. Gastrointestinal neuroendocrine/neuroectodermal tumors. Am J Clin Pathol. 2001;115 Suppl:S79-S93.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Gaffey MJ, Mills SE, Lack EE. Neuroendocrine carcinoma of the colon and rectum. A clinicopathologic, ultrastructural, and immunohistochemical study of 24 cases. Am J Surg Pathol. 1990;14:1010-1023.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Shida T, Furuya M, Nikaido T, Kishimoto T, Koda K, Oda K, Nakatani Y, Miyazaki M, Ishikura H. Aberrant expression of human achaete-scute homologue gene 1 in the gastrointestinal neuroendocrine carcinomas. Clin Cancer Res. 2005;11:450-458.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Hung SS. Small cell carcinoma of the colon. A case report and literature review. J Clin Gastroenterol. 1989;11:335-339.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Travis WD, Linnoila RI, Tsokos MG, Hitchcock CL, Cutler GB Jr, Nieman L, Chrousos G, Pass H, Doppman J. Neuroendocrine tumors of the lung with proposed criteria for large-cell neuroendocrine carcinoma. An ultrastructural, immunohistochemical, and flow cytometric study of 35 cases. Am J Surg Pathol. 1991;15:529-553.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Shia J, Tang LH, Weiser MR, Brenner B, Adsay NV, Stelow EB, Saltz LB, Qin J, Landmann R, Leonard GD. Is nonsmall cell type high-grade neuroendocrine carcinoma of the tubular gastrointestinal tract a distinct disease entity? Am J Surg Pathol. 2008;32:719-731.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Kato T, Terashima T, Tomida S, Yamaguchi T, Kawamura H, Kimura N, Ohtani H. Cytokeratin 20-positive large cell neuroendocrine carcinoma of the colon. Pathol Int. 2005;55:524-529.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Driessen A, Nafteux P, Lerut T, Van Raemdonck D, De Leyn P, Filez L, Penninckx F, Geboes K, Ectors N. Identical cytokeratin expression pattern CK7+/CK20- in esophageal and cardiac cancer: etiopathological and clinical implications. Mod Pathol. 2004;17:49-55.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Kumarasinghe MP, Weng EK. Pathological features and their prognostic implications in colorectal endocrine cell tumours: a long-term follow-up study. Pathology. 2005;37:204-210.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Furlan D, Cerutti R, Genasetti A, Pelosi G, Uccella S, La Rosa S, Capella C. Microallelotyping defines the monoclonal or the polyclonal origin of mixed and collision endocrine-exocrine tumors of the gut. Lab Invest. 2003;83:963-971.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Capella C, La Rosa S, Uccella S, Billo P, Cornaggia M. Mixed endocrine-exocrine tumors of the gastrointestinal tract. Semin Diagn Pathol. 2000;17:91-103.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Rossi G, Bertolini F, Sartori G, Bigiani N, Cavazza A, Foroni M, Valli R, Rindi G, De Gaetani C, Luppi G. Primary mixed adenocarcinoma and small cell carcinoma of the appendix: a clinicopathologic, immunohistochemical, and molecular study of a hitherto unreported tumor. Am J Surg Pathol. 2004;28:1233-1239.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Hervieu V, Scoazec JY. [Mixed endocrine tumors]. Ann Pathol. 2005;25:511-528.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Liu L, Broaddus RR, Yao JC, Xie S, White JA, Wu TT, Hamilton SR, Rashid A. Epigenetic alterations in neuroendocrine tumors: methylation of RAS-association domain family 1, isoform A and p16 genes are associated with metastasis. Mod Pathol. 2005;18:1632-1640.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Oberg K. Neuroendocrine tumors of the gastrointestinal tract: recent advances in molecular genetics, diagnosis, and treatment. Curr Opin Oncol. 2005;17:386-391.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Rindi G, Villanacci V, Ubiali A. Biological and molecular aspects of gastroenteropancreatic neuroendocrine tumors. Digestion. 2000;62 Suppl 1:19-26.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Göke R, Gregel C, Göke A, Arnold R, Schmidt H, Lankat-Buttgereit B. Programmed cell death protein 4 (PDCD4) acts as a tumor suppressor in neuroendocrine tumor cells. Ann N Y Acad Sci. 2004;1014:220-221.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Blanes A, Diaz-Cano SJ. Complementary analysis of microsatellite tumor profile and mismatch repair defects in colorectal carcinomas. World J Gastroenterol. 2006;12:5932-5940.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Wu Y, Berends MJ, Mensink RG, Kempinga C, Sijmons RH, van Der Zee AG, Hollema H, Kleibeuker JH, Buys CH, Hofstra RM. Association of hereditary nonpolyposis colorectal cancer-related tumors displaying low microsatellite instability with MSH6 germline mutations. Am J Hum Genet. 1999;65:1291-1298.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Müller A, Fishel R. Mismatch repair and the hereditary non-polyposis colorectal cancer syndrome (HNPCC). Cancer Invest. 2002;20:102-109.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Fishel R. The selection for mismatch repair defects in hereditary nonpolyposis colorectal cancer: revising the mutator hypothesis. Cancer Res. 2001;61:7369-7374.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Stelow EB, Moskaluk CA, Mills SE. The mismatch repair protein status of colorectal small cell neuroendocrine carcinomas. Am J Surg Pathol. 2006;30:1401-1404.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Arnold CN, Nagasaka T, Goel A, Scharf I, Grabowski P, Sosnowski A, Schmitt-Gräff A, Boland CR, Arnold R, Blum HE. Molecular characteristics and predictors of survival in patients with malignant neuroendocrine tumors. Int J Cancer. 2008;123:1556-1564.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A. Neuroendocrine tumors. Part III Digestive System. AJCC Cancer Staging Manual, 7th ed. New York: Springer-Verlag 2010; 181-190.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Landry CS, Brock G, Scoggins CR, McMasters KM, Martin RC 2nd. A proposed staging system for rectal carcinoid tumors based on an analysis of 4701 patients. Surgery. 2008;144:460-466.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Landry CS, Brock G, Scoggins CR, McMasters KM, Martin RC 2nd. Proposed staging system for colon carcinoid tumors based on an analysis of 2,459 patients. J Am Coll Surg. 2008;207:874-881.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Fujimoto Y, Oya M, Kuroyanagi H, Ueno M, Akiyoshi T, Yamaguchi T, Muto T. Lymph-node metastases in rectal carcinoids. Langenbecks Arch Surg. 2010;395:139-142.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Konishi T, Watanabe T, Kishimoto J, Kotake K, Muto T, Nagawa H. Prognosis and risk factors of metastasis in colorectal carcinoids: results of a nationwide registry over 15 years. Gut. 2007;56:863-868.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Shebani KO, Souba WW, Finkelstein DM, Stark PC, Elgadi KM, Tanabe KK, Ott MJ. Prognosis and survival in patients with gastrointestinal tract carcinoid tumors. Ann Surg. 1999;229:815-821; discussion 822-823.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Fahy BN, Tang LH, Klimstra D, Wong WD, Guillem JG, Paty PB, Temple LK, Shia J, Weiser MR. Carcinoid of the rectum risk stratification (CaRRs): a strategy for preoperative outcome assessment. Ann Surg Oncol. 2007;14:1735-1743.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Hassan MM, Phan A, Li D, Dagohoy CG, Leary C, Yao JC. Risk factors associated with neuroendocrine tumors: A U.S.-based case-control study. Int J Cancer. 2008;123:867-873.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Hemminki K, Li X. Incidence trends and risk factors of carcinoid tumors: a nationwide epidemiologic study from Sweden. Cancer. 2001;92:2204-2210.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Klöppel G, Anlauf M. Epidemiology, tumour biology and histopathological classification of neuroendocrine tumours of the gastrointestinal tract. Best Pract Res Clin Gastroenterol. 2005;19:507-517.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Tomoda H, Furusawa M, Hayashi I, Okumura K. A rectal carcinoid tumor of less than 1 cm in diameter with lymph node metastasis: a case report and a review of the literature. Jpn J Surg. 1990;20:468-471.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Soga J. Carcinoids of the rectum: an evaluation of 1271 reported cases. Surg Today. 1997;27:112-119.  [PubMed]  [DOI]  [Cited in This Article: ]