Retrospective Study Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Mar 7, 2015; 21(9): 2700-2710
Published online Mar 7, 2015. doi: 10.3748/wjg.v21.i9.2700
Mucinous phenotype and CD10 expression of primary adenocarcinoma of the small intestine
Reiko Kumagai, Kenichi Kohashi, Shunsuke Takahashi, Hidetaka Yamamoto, Minako Hirahashi, Yoshinao Oda, Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
Kenichi Taguchi, Department of Pathology, National Hospital Organization Kyushu Cancer Center, Fukuoka 811-1395, Japan
Kenichi Nishiyama, Division of Pathology, Japanese Red Cross Fukuoka Hospital, Fukuoka 815-8555, Japan
Author contributions: Kumagai R and Kohashi K performed the experiments, and the statistical analysis and drafted the manuscript; Kumagai R and Takahashi S carried out the experiments and analyzed the data; Kumagai R and Nishiyama K collected the data; Yamamoto H gave technical support and conceptual advice; Hirahashi M, Taguchi K and Oda Y participated in the design of the study and helped to draft the manuscript.
Ethics approval: The study was reviewed and approved by the Institutional Review Board of Kyushu University (IRB#25-191).
Informed consent: Informed consent was not obtained, but this study is not an interventional study and we completely anonymized or omitted all the information that might disclose the identity of the subjects.
Conflict-of-interest: The authors declare that there are no conflicts of interest to disclose.
Data sharing: No additional data are available.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Dr. Yoshinao Oda, Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan. oda@surgpath.med.kyushu-u.ac.jp
Telephone: +81-92-6426061 Fax: +81-92-6425968
Received: October 11, 2014
Peer-review started: October 13, 2014
First decision: October 29, 2014
Revised: November 13, 2014
Accepted: December 20, 2014
Article in press: December 22, 2014
Published online: March 7, 2015

Abstract

AIM: To clarify the correlation with phenotypic expression, clinicopathological features, genetic alteration and microsatellite-instability status in small intestinal adenocarcinoma (SIA).

METHODS: The cases of 47 patients diagnosed with primary SIAs that were surgically resected at our institution in 1975-2005 were studied. We reviewed clinicopathological findings (age, gender, tumor size, gross appearance, histological morphologic type, invasion depth, lymphatic permeation, venous invasion, and lymph node metastasis), and the immunohistochemical expression of MUC5AC, MUC6, MUC2, CD10, and mismatch-repair (MMR) proteins (MLH1 and MSH2). We analyzed KRAS and BRAF gene mutations, and the microsatellite instability (MSI) status. The immunohistochemical staining of CD10, MUC2, MUC5AC and MUC6 was considered positive when distinct staining in > 5% of the adenocarcinoma cells was recorded. To evaluate of MMR protein expression, we used adjacent normal tissue including lymphoid follicles, inflammatory cells, and stromal cells as an internal positive control. Sections without nuclear staining in the tumor cells were considered to have lost the expression of the respective MMR protein.

RESULTS: There were 29 males and 18 females patients (mean age 59.9 years, range: 23-87 years). Tumors were located in the duodenum in 14 cases (30%), the jejunum in 21 cases (45%), and the ileum in 12 cases (25%). A phenotypic expression analysis revealed 20 MUC2-positive tumors (42.6%), 11 MUC5AC-positive (23.4%), 4 MUC6-positive (8.5%), and 7 CD10-positive (14.9%). The tumor sizes of the MUC2(+) tumors were significantly larger than those of the MUC2(-) tumors (mean, 5.7 ± 1.4 cm vs 4.7 ± 2.1 cm, P < 0.05). All three tumors with adenomatous component were positive for MUC2 (P < 0.05). Polypoid appearance was seen significantly more frequently in the CD10(+) group than in the CD10(-) group (P < 0.05). The tumor size was significantly larger in the CD10 (+) group than in the CD10(-) group (mean, 5.9 ± 1.4 cm vs 5.0 ± 2.1 cm, P < 0.05). Of 34 SIAs with successfully obtained MSI data, 4 were MSI-high. Of the 4 SIAs positive for both MUC5AC and MUC2, 3 showed MSI-H (75%) and 3 were mucinous adenocarcinoma (75%). KRAS mutations were detected in 4 SIAs. SIAs had KRAS mutation expressed only MUC2, but were negative for MUC5AC, MUC6 and CD10.

CONCLUSION: These findings suggest that the phenotypic expression of SIAs is correlated with their biological behavior, genetic alteration, and MSI status.

Key Words: Small intestine, Adenocarcinoma, Mucin, CD10, Microsatellite instability

Core tip: This study analyzed the immunohistochemical expression of mucin core proteins (MUC5AC, MUC6 and MUC2), CD10 and mismatch-repair proteins (MLH1, MSH2), microsatellite instability (MSI), and the mutational status of KRAS and BRAF in 47 primary small intestinal adenocarcinoma. We suggest that the mucin phenotype and CD10 expression of small intestinal adenocarcinoma correlates with biological behavior, genetic alteration, and MSI status. Notably, the preservation of CD10 expression may be correlated with favorable biological behavior. The SIA with co-expression of MUC5AC and MUC2 was correlated with MSI-H status and mucinous adenocarcinoma, similar to colorectal carcinoma.



INTRODUCTION

Primary small intestinal adenocarcinomas (SIAs) are rare, accounting for only about 0.8% of all malignant gastrointestinal tumors, although the small intestine represents 75% of the length and 90% of the mucosal surface of the gastrointestinal tract[1,2]. Despite major advances in imaging and therapeutic techniques, the prognosis of SIAs remains dismal, with an overall 5-year survival rate of 25%-30%[3,4].

Mucins are a family of high-molecular-weight, heavily glycosylated proteins that are widely produced by epithelial tissue. In the alimentary canal, MUC2 is produced by goblet cells of the small and large intestinal mucosa, and MUC5AC is produced by gastric foveolar epithelium. MUC6 is produced by the gastric pyloric gland and duodenal Brunner’s glands. Compared to normal mucosa, adenocarcinomas occasionally produce different types of mucin. Some researchers have reported that in gastric carcinoma (GC) or colorectal carcinoma (CRC), the mucin phenotype was related to the tumor growth pattern, appearance, aggressiveness, prognosis, genetic alternation and microsatellite instability (MSI) status. For example, it was reported that MUC2-positive CRCs have a relatively favorable prognosis[5], whereas loss of MUC2 was an adverse prognostic factor in CRCs[6]. MUC5AC was often found to be expressed in CRCs with MSI-high (MSI-H) and/or mucinous adenocarcinomas, and intriguingly, this type of CRCs often express MUC2 as well[7,8].

The “serrated pathway” is an evolving pathway in the carcinogenesis of CRC, characterized by BRAF gene mutations and MSI-H[9]. Sessile serrated adenomas, considered precursors of MSI-H CRCs, often express both MUC2 and MUC5AC, the same as MSI-H CRCs do[10].

It has been suggested that not only mucin expression but also CD10 expression, which is usually seen in the brush border of the small intestinal villi, are correlated with aggressive biological behavior of CRCs. CD10-positive CRCs in particular were suggested to be prone to the development of metastasis to the liver[11,12]. Analyses of the mucin core proteins and CD10 expression in adenocarcinomas of the alimentary canal are thus important to determine these tumors’ biological behavior and carcinogenesis.

In SIAs, however, the phenotypic expressions of mucin core proteins and CD10 have not been established, and the clinicopathological features and pathogenesis of SIAs are less known than those of CRCs.

The purposes of the present study were to evaluate the mucin core proteins and CD10 expression of primary SIAs and to clarify their correlation with the phenotypic expression, clinicopathological features, biological behavior, genetic alterations and MSI status.

MATERIALS AND METHODS
Patients

The cases of 47 patients diagnosed with primary SIAs that were surgically resected and diagnosed at the Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University (Fukuoka, Japan) between January 1975 and December 2005 were studied.

To prevent confusion with the findings of carcinomas in neighboring organs, all carcinomas involving the pylorus, ileocecal valve, and ampulla of Vater were excluded from the analysis. The cases of patients diagnosed with hereditary nonpolyposis colorectal cancer (HNPCC), familial adenomatous polyposis (FAP), celiac disease or inflammatory bowel disease (IBD) were also excluded. The study was approved by the Institutional Review Board of Kyushu University (IRB#25-191).

Histological evaluation

The surgically resected small intestine specimens were each fixed in 10% formalin and examined macroscopically. The entire tumor tissue was sliced in a serial fashion at approx. 3 to 5 mm thickness, and the tissue slices were routinely processed to paraffin blocks and stained with hematoxylin and eosin (HE). Clinicopathologic findings (patient age, gender, tumor size, gross appearance, histological morphologic type, invasion depth, lymphatic permeation, venous invasion, and lymph node metastasis) were reviewed. Representative sections were also used for the subsequent immunohistochemical stains and molecular analysis.

Immunohistochemistry

Immunohistochemical staining was performed using 4-μm-thick formalin-fixed, paraffin-embedded tissue sections and the primary antibodies for CD10 (56C6, dilution 1:100, Leica, Newcastle, United Kingdom), MUC2 (Ccp58, dilution 1:200, Leica), MUC5AC (CLH1, dilution 1:200, Leica), MUC6 (CLH5, dilution 1:200, Leica,), MLH1 (G168-728, dilution 1:50, BD Bioscience, San Diego, CA) and MSH2 (FE11, dilution 1:200, Oncogene Research Products, La Jolla, CA). A biotin-free, horseradish peroxidase (HRP) enzyme-labeled polymer method (Envision+ system; Dako, Carpinteria, CA) was used, with hematoxylin counterstaining.

The immunohistochemical staining of CD10, MUC2, MUC5AC and MUC6 was considered positive when distinct staining in more than 5% of the adenocarcinoma cells was recorded, in reference to the phenotypic expressions of previous studies[13,14]. The adenomatous area of the SIAs was excluded in the evaluation of immunohistochemical staining.

For the evaluation of mismatch-repair (MMR) protein expression (i.e., MLH1 and MSH2), adjacent normal tissue including lymphoid follicles, inflammatory cells, and stromal cells was used as an internal positive control. Sections without nuclear staining in the tumor cells were considered to have lost the expression of the respective MMR protein.

Sections were viewed by three independent observers (R.K., K.K. and M.H.) with conflicts resolved using a conference microscope.

Mutational analysis for KRAS and BRAF

Genomic DNA was extracted from paraffin sections using the QIAamp DNA Mini Kit (Qiagen, Valencia, CA), according to the manufacturer’s instructions. To detect KRAS mutation in exon 2, and BRAF mutation in exon 15, we performed a polymerase chain reaction (PCR) followed by sequencing, as described[15]. The primer sets used are listed in Table 1.

Table 1 Primers used in this study.
Primer
KRASForward5’-GGCCTGCTGAAAATGACTGA-3’
Reverse5’-GTTGGATCATATTCGTCCAC-3’
BRAFForward5’-CCTTTACTTACTACACCTCAG-3’
Reverse5’-CATCCACAAAATGGATCCAG-3’
BAT25Forward15’-TCGCCTCCAAGAATGTAAGT-3’
Reverse5’-TCTGCATTTTAACTATGGCTC-3’
BAT26Forward15’-TGACTACTTTTGACTTCAGCC-3’
Reverse5’-AACCATTCAACATTTTTAACCC-3’
D2S123Forward15’-GAAACAGGATCCTGCCTTTA-3’
Reverse5’-GGACTTTCCACCTATGGGAC-3’
D5S346Forward15’-GACTCACTCTAGTATAAATCGGG-3’
Reverse5’-GAGCAGATAAGACAGTATTACTAGTT-3’
D17S250Forward15’-GGGAAGAATCAAATAGACAAT-3’
Reverse5’-GCTGGCCATATATATATTTAAACC-3’
MSI analysis

DNA isolated from the adenocarcinomas and non-neoplastic normal mucosa was amplified. The MSI status was determined using the Bethesda panel established by the United States National Cancer Institute, including mononucleotide (BAT25 and BAT26) and dinucleotide (D2S123, D5S346, D17S250) repeats[16]. The primer sets used are listed in Table 1. PCR products were separated on an ABI 310 automated sequencer (Applied Biosystems, Warrington, United Kingdom). The data analysis was done by GeneScan software (Applied Biosystems). Tumors with two or more of the markers exhibiting instability were classified as MSI-H. Tumors with only one marker exhibiting instability were classified as MSI-low (MSI-L), and those showing no markers with instability were classified as microsatellite stable (MSS).

Statistical analysis

All calculations were made using JMP Statistical Discovery Software (version 8.0; SAS, Cary, NC). We used Fisher’s exact test, the χ2 test or Wilcoxon signed-rank test to evaluate the association between the phenotypic expression and clinicopathological features or genetic alteration. The results were considered significant when the P value was < 0.05. The statistical methods of this study were reviewed by Mototsugu Shimokawa from Clinical Research Institute, National Hospital Organization Kyushu Cancer Center.

RESULTS
Clinicopathological findings

The clinicopathological findings of the 47 SIAs are shown in Table 2. There were 29 male and 18 female patients with a mean age of 59.9 years (range 23-87 years). Tumors were located in the duodenum in 14 cases (30%), the jejunum in 21 cases (45%), and the ileum in 12 cases (25%). The tumor showed polypoid growth in 12 cases, and ulcerative growth in 35 cases. The tumor size ranged from 1.5 to 10.0 cm (mean, 5.1 cm). Two tumor invaded the lamina propria mucosae or the submucosa (pT1, classified invasion depth according to the UICC TNM classification of malignant tumors[17]), 4 tumors invaded the muscularis propria (pT2), 31 tumors invaded into the subserosa (pT3), and 10 tumors had perforated the visceral serosa or directly invaded other organs (pT4). Of the 25 tumors resected with lymph nodes, 16 had lymph node metastases (data not shown). Thirty-three tumors were well or moderately differentiated (tubular), 9 were poorly differentiated, and 5 were mucinous adenocarcinomas. Lymphatic permeation was detected in 33 tumors (70%), and venous invasion was detected in 12 tumors (26%). Three tumors had an adjacent adenomatous component, which showed a tubulo-villous structure.

Table 2 Clinicopathological findings of all 47 cases of small intestinal adenocarcinomas n (%).
All cases (n = 47)
Age (yr)
Mean (range)59.9 (23-87)1
Gender
Male29
Female18
Gross appearance
Polypoid12
Ulcerative35
Size (cm) (mean)5.13
Location
Duodenum14
Jejunum21
Ileum12
Histological subtype
Tubular233
Poorly9
Mucinous5
Invasion depth3
T12
T24
T318
T423
Lymphatic permeation33 (70)
Venous invasion12 (26)
Adenomatous component3 (6)
Expressions of mucin core proteins and CD10

The phenotypic expression analysis of the 47 SIAs revealed 20 tumors (42.6%) positive for MUC2, 11 tumors (23.4%) positive for MUC5AC, 4 tumors (8.5%) positive for MUC6, and 7 tumors (14.9%) positive for CD10. The correlations between clinicopathological features and the expressions of mucin core proteins and CD10 are summarized in Table 3.

Table 3 Clinicopathological features and expression of mucin core proteins and CD10.
MUC5AC
MUC6
MUC2
CD10
Positive(n = 12)Negative(n = 35)Positive(n = 4)Negative(n = 43)Positive(n = 20)Negative(n = 27)Positive(n = 7)Negative(n = 40)
Age (yr)
Mean ± SD63.4 ± 15.1158.8 ± 16.576.3 ± 10.0158.7 ± 15.962.3 ± 15.358.0 ± 16.7160.1 ± 15.159.8 ± 16.51
(range)(35-85)(23-87)(67-87)(23-85)(38-87)(23-81)(38-87)(23-85)
Gender (male/female)10/219/163/126/179/1118/93/427/14
Gross appearance
(polypoid/ulcerated)3/99/262/210/336/146/214/3a8/32a
Size (cm)5.3 ± 2.25.1 ± 1.95.0 ± 1.85.1 ± 2.05.7 ± 1.6a4.7 ± 2.2a5.9 ± 1.4a5.0 ± 2.1a
Location
(duodenum/jejunum/ileum)2/5/512/16/72/1/112/20/116/11/38/10/94/2/110/19/11
Histological subtype
(tubular2/poorly/mucinous)6/3/327/6/23/0/130/9/413/2/510/7/07/0/029/9/5
Invasion depth3
T10202202a0a
T22222222a2a
T32160186121a17a
T481522110132a21a
Lymphatic permeation n (%)10 (83)23 (66)2 (50)31 (72)12 (60)21 (78)3 (43)30 (75)
Venous invasion n (%)3 (25)9 (26)1 (25)11 (26)5 (25)7 (26)2 (29)10 (25)
Adenomatous component n (%)0 (0)3 (9)1 (25)2 (5)3 (15)a0 (0)a2 (29)a1 (3)a

All mucin core proteins were expressed in the cytoplasm of the tumor cells. The tumor sizes of the MUC2(+) tumors were significantly larger than those of the MUC2(-) tumors (mean, 5.7 ± 1.6 cm vs 4.7 ± 2.2 cm, P < 0.05). All 3 tumors with adenomatous component were positive for MUC2 (P < 0.05). The MUC5AC(+) SIAs were more prevalent in the males than the females (9 males vs 2 females). The ages of the patients in the MUC6(+) group tended to be higher than those of the MUC6(-) group (mean, 76.3 ± 10.0 years vs 58.7±15.9 years).

CD10 expression was observed along the luminal surface of the tumor glands. In the CD10(+) group, CD10 was more frequently expressed in the superficial areas of the adenocarcinomas than in their deep area, and the deep area of the CD10-negative adenocarcinomas showed the same tubular shape but more high-grade atypia compared to the surperficial area of the CD10-positive adenocarcinomas (Figure 1). Polypoid appearance was seen significantly more frequently in the CD10(+) group compared to the CD10(-) group (P < 0.05). The tumor size was significantly larger in the CD10(+) group than in the CD10(-) group (mean, 5.9 ± 1.4 cm vs 5.0 ± 2.1 cm, P < 0.05). The invasion depth was significantly deeper in the CD10(-) group than in the CD10(+) group (T1 vs T2-T4, P < 0.05). The incidence of lymphatic permeation tended to be less in the CD10(+) group than in the CD10(-) group (Figure 2).

Figure 1
Figure 1 Representative CD10-positive small intestinal adenocarcinoma case. A: Well differentiated adenocarcinoma (pointed by arrows) (HE stain, × 12.5); B: CD10-positive glands were seen in the superficial area and the edge of the adenocarcinoma (left, pointed by arrows), while CD10-negative glands were seen in the center or deep area of the tumor (right) (CD10, × 12.5); C, D: These glands in the surface area were positive for CD10 (HE stain and CD10, × 200); E, F: Other glands in the deeper invasive area were negative for CD10 and showed more evident loss of nuclear polarity compared to the CD10-positive glands (HE stain and CD10, × 200).
Figure 2
Figure 2 Representative CD10-negative small intestinal adenocarcinoma case. A: Normal small intestinal mucosa was observed on the right side, whereas the adenocarcinoma was observed on the left side (HE stain, × 12.5); B: Normal mucosa was positive for CD10, but adenocarcinoma was negative for CD10 (CD10, × 12.5); C: The brush border of the normal mucosa was positive for CD10 (CD10, × 100); D: This SIA case showed a poorly differentiated adenocarcinoma (HE satin, × 200); E: Lymphatic permeation was frequently seen (HE stain, × 200); F: The carcinoma cells were negative for CD10 (CD10, × 200).

We found 16 SIAs that expressed only intestinal markers (MUC2 and/or CD10), 7 SIAs that expressed only gastric markers (MUC5AC and/or MUC6), and 6 SIAs that expressed both gastric and intestinal markers, including 4 SIAs that were positive for both MUC5AC and MUC2.

Expression of MLH1 and MSH2

Four of the 47 cases could not be evaluated for MLH1 due to poor staining quality. One of the remaining 43 tumors showed loss of MLH1, and 2 of the 47 tumors showed loss of MSH2.

MSI analysis

MSI data were obtained from 34 tumors. The phenotypes were MSI-H in 4 tumors (12%), MSI-L in 8 tumors (24%), and MSS in 22 tumors (64%). Immunohistochemical loss of MMR protein was detected in 2 of 4 MSI-H tumors (1 tumor showed loss of MLH1, and the other showed loss of MSH2), 1 of 8 MSI-L tumors (loss of MSH2), and no MMS tumors (Table 4).

Table 4 Summarized data of microsatellite instability status, expression of mismatch repair proteins and KRAS mutations.
MSIExpression of MMR protein
KRAS mutationbase exchange (codon)AgeGender
MLH1MSH2
Case 6MSI-H++-55M
Case 36MSI-H+--85M
Case 31MSI-H-+-67M
Case 34MSI-H++-71M
Case 12MSI-L++GGT>GAT(12)69M
Case 28MSI-L+--41M
Case 24MSI-L++-60M
Case 40MSI-L++-35M
Case 25MSI-L++-53F
Case 5MSI-L++-23F
Case 44MSI-L++-87F
Case 38MSI-L++-67F
Case 41MSS++GGC>GAC(13)45F
Case 20MSS++GGT>GAT(12)59F
Case 46MSS++GGT>GTT(12)40F

The data of MSI status and clinicopathological features is shown in Table 5. The MSI-H SIAs were composed of 2 well differentiated adenocarcinomas and 2 mucinous adenocarcinomas (Figure 3). The tumor size was significantly larger in MSI-H SIAs than in MSI-L and MSS SIAs (mean 7.7 ± 1.1 cm vs 5.1 ± 2.0 cm, P < 0.05).

Figure 3
Figure 3 Small intestinal adenocarcinoma with microsatellite instability-high (case 36). A: The tumor produced abundant mucin (HE stain, × 20); B: The tumor focally showed signet ring-like carcinoma cells (HE stain, × 200); C: The carcinomatous gland was composed of columnar cells with intracytoplasmic mucin (HE stain, × 200); D: These glands were positive for MUC2 (MUC2, × 200); E: positive for MUC5AC (MUC5AC, × 200); F: But negative for MUC6 (MUC6, × 200). In these microsatellites, the tumor was unstable. Microsatellite analysis of paired normal and tumor DNA using G: BAT25; H: BAT26; I: D17S250.
Table 5 Clinicopathological features, microsatellite instability status and KRAS mutation.
MSI
KRAS
MSI-H(n = 4)MSI-L/MSS(n = 30)Mutational type(n = 4)Wild type(n = 33)
Age (yr)69.5 ± 12.359.6 ± 15.9153.3 ± 13.260.8 ± 16.21
Gender (male/female)4/016/141/322/11
Gross appearance (polypoid/ulcerated)1/36/242/29/24
Size (cm)7.7 ± 1.1a5.1 ± 2.0a4.4 ± 1.05.5 ± 2.0
Location (duodenum/jejunum/ileum)0/2/27/15/81/2/19/15/9
Histological subtype (tubular2/poorly/mucinous)2/0/222/6/24/0/022/8/3
Invasion depth3
T10101
T20203
T3213113
T4214315
Lymphatic permeation n (%)3 (75)23 (77)4 (100)22 (67)
Venous invasion n (%)1 (25)9 (30)1 (25)10 (30)
Adenomatous component n (%)0 (0)1 (3)0 (0)2 (6)
Frequency and associations of tumor gene mutations

The mutation analysis was successfully conducted in 37 of the 47 cases. The genetic mutations and MSI status findings are summarized in Table 4. KRAS mutations were prevalent in 11% (4 of 37) of the tumors. The most prevalent KRAS mutation was GGT > GAT(G12D) within codon 12, which was detected in 2 cases. One case showed GGT>GTT(G12V) within codon 12, and one case showed GGC>GAC(G13D) within codon 13. The data of KRAS mutation and clinicopathological features is shown in Table 5. No BRAF V600E mutation was found in any cases.

Correlations among the mucin phenotype, CD10 expression, genetic alteration and MSI status

Three of the 4 SIAs with KRAS mutation were positive for only MUC2, but negative for MUC5AC, MUC6 and CD10 (75%). Three of the 4 SIAs positive for both MUC5AC and MUC2 showed MSI-H (75%) (Table 6). One of the 4 MSI-H SIA was immunohistochemically negative for all antibodies (MUC5AC, MUC6, MUC2 and CD10).

Table 6 Summarized data of 4 small intestinal adenocarcinomas with co-expression of MUC5AC and MUC2.
AgeGenderLocationHistological subtypeInvasion depthLymphatic permeationVenous invasionLymph node metastasisMSI
case 655mJejunumTubularT3---MSI-H
case 2961mJejunumMucinousT4+-No dataMSS
case 3167mIleumMucinousT4+-No dataMSI-H
case 3685mIleumMucinousT4+--MSI-H
DISCUSSION

CD10 is a membrane-bound, zinc-containing metalloendopeptidase, that is expressed in a wide variety of normal and neoplastic cells including pre-cursor B and T cells, follicle center B cells in the hematopoietic system, myoepithelial cells of the breast and salivary glands, basaloid cells of the prostatic gland, glomerular cells of the kidney, and stromal cells in the endometrium[18,19]. In the alimentary canal, normal small intestine shows immunoreactivity for CD10 in the brush border. On the other hand, except for intestinal metaplasia of the stomach, the normal gastric foveolae and the colorectal epithelium show no CD10 expression.

In our evaluation of the distribution of CD10, we observed CD10 expression in the superficial area of the SIAs. The expression diminished with greater depth and was not detected in the deepest area of almost all of the CD10(+) SIAs. Yao et al[13] reported that the CD10 expression of CRCs was more frequently observed in the center or deep area of the tumors than their periphery. The distribution of CD10-positive areas seemed to be opposite in SIAs and CRCs.

Regarding the CD10 expression of advanced CRCs, the significant correlations have been reported between CD10 expression and a high incidence of liver metastasis[11,12,20], and between CD10 expression and venous invasion[13]. Carl-McGrath et al[21] reported that CD10 was significantly up-regulated in GCs and lymph node metastases. Although the concrete function of CD10 in carcinogenesis remains unclear, the acquisition of CD10 expression correlates with the malignant potential in CRCs and GCs. In the present study, the incidence of lymphatic permeation tended to be lower in the CD10(+) SIAs than in the CD10(-) SIAs, regardless of the tendency for larger tumor sizes in the CD10(+) SIAs. The CD10-positive adenocarcinoma glands showed lower atypia than the CD10-negative adenocarcinoma glands did in the same SIAs. These results suggest that CD10(+) SIAs are less aggressive than CD10(-) SIAs, and that the loss of CD10 expression in SIAs may be correlated with the phenotypic dedifferentiation and malignant phenotype.

The relationship between mucin phenotype and clinicopathological or biological features has been investigated in various types of GC and CRC[7,8,11-14,22-25]. It is well established that MUC2 expression is related to the progression or prognosis of CRC[5,6,25], an association that suggests that the expression of MUC2 is also related to the biological behavior of SIA. Although in the present study the tumor sizes of the MUC2(+) SIAs were significantly larger than those of the MUC2(-) SIAs, the frequency of lymphatic permeation tended to be lower in the MUC2(+) SIAs than in the MUC2(-) SIAs. Poorly differentiated adenocarcinomas were prevalent in the MUC2(-) SIA group. From these findings, we infer that MUC2(-) SIAs are more aggressive than MUC2(+) SIAs. Loss of MUC2 expression in SIAs may also be correlated with malignant potential.

The serrated neoplastic pathway of CRCs is characterized by BRAF mutation, abnormal DNA methylation, and MSI-H status, and some authors suggested that serrated polyps were the precursor of MSI-H CRCs[9,26]. The MUC5AC+/ MUC2+ mucinous phenotype is common in serrated polyps (including hyperplastic polyps and serrated adenomas) and MSI-H CRCs[10,27,28]. Arai et al[28] also demonstrated a higher proportion of MSI-H in mucinous adenocarcinoma. In the present study, we observed the 4 SIAs that expressed both MUC5AC and MUC2, and these 4 SIAs contained 3 MSI-H tumors and 3 mucinous adenocarcinomas. The data suggested that the co-expression of MUC5AC and MUC2 in SIAs was also correlated with MSI-H tumors and mucinous adenocarcinomas as in CRCs.

We did not detect any serrated polyps or any mutations in codon 600 of BRAF gene. Fu et al[29] investigated 99 duodenal adenocarcinomas and detected no BRAF mutation among them, and therefore they suggested that BRAF mutations are not involved in MSI-H duodenal adenocarcinomas. In other investigations, the BRAF mutations were rare in SIAs[30,31]. In the carcinogenesis of SIAs, it is still unclear whether there is a serrated neoplastic pathway as same as in CRCs.

The present study revealed that 4 of 34 (12%). SIAs had microsatellite instability. This result is in line with reports by other investigators, who described the frequency of MSI-H of SIAs of 5% to 30%[32-35]. We also found that 2 of 4 MSI-H tumors showed the immunohistochemical loss of MMR protein expression, which affected MLH1 in 1 case and MSH2 in 1 case. Mangold et al[36] reported that 32% of CRCs from MLH1 mutation-carrier patients showed partly positive or weak staining of MLH1. In the present study, we considered only the completely negative staining of MLH1 as the loss of MLH1 protein, and thus some of the SIAs we assessed as positive for MLH1 protein could be false-positive. Planck et al[35] investigated a large population-based series of 89 SIAs, and eight of 15 MSI-H tumors expressed both MLH1 and MSH2. We suggest that the immunohistochemical staining of MLH1 and MSH2 proteins is generally useful to detect MSI-H SIAs, but a molecular analysis is necessary to detect some MSI-H SIAs.

Previous studies detected KRAS mutations in 14%-83% of SIA cases[32,34,37-39]. We detected mutations in KRAS codon 12 and codon 13 in 4 of 37 SIAs. The frequency and the location were in line with those of the previous studies. Three of the 4 present SIAs with KRAS mutations were positive for only MUC2 (75%). We found a tendency for the SIAs with KRAS mutations to have the colonic phenotype.

In conclusion, we suggest that the mucinous phenotype and CD10 expression of SIAs also correlates with the tumors’ biological behavior, genetic alteration, and MSI status. In particular, the preservation of CD10 expression, correlated with larger tumor size, polypoid appearance and less lymphatic permeation, features that are suggestive of less aggressive biological behavior. The co-expression of MUC5AC and MUC2 of the SIAs was correlated with the MSI-H status and mucinous adenocarcinoma, as in CRCs.

ACKNOWLEDGMENTS

We are grateful to Dr. Shimokawa M, National Hospital Organization Kyushu Cancer Center, for reviewing the statistical method of this study.

COMMENTS
Background

Mucins are a family of high-molecular-weight, heavily glycosylated proteins that are widely produced by epithelial tissue. In the alimentary canal, MUC2 is produced by goblet cells of the small and large intestinal mucosa, and MUC5AC is produced by gastric foveolar epithelium.

Research frontiers

The purposes of the present study were to evaluate the mucin core proteins and CD10 expression of primary small intestinal adenocarcinomas (SIAs) and to clarify their correlation with the phenotypic expression, clinicopathological features, biological behavior, genetic alterations and microsatellite instability status.

Innovations and breakthroughs

The authors considered only the completely negative staining of MLH1 as the loss of MLH1 protein, and thus some of the SIAs we assessed as positive for MLH1 protein could be false-positive.

Peer-review

The paper by Kumagai et al entitled “Mucinous phenotype and CD10 expression of primary adenocarcinoma of the small intestine: Possible association with biological behavior, genetic alteration and microsatellite instability status” investigates the expression of CD10, MUC5A, MUC2, MUC6 and proteins related to microsatellite instability/mismatch repair proteins (MLH1 and MSH2) in 47 cases of small bowel adenocarcinoma, evaluated by immunohistochemistry. Then, Authors correlated the immunohistochemical pattern of expression of these molecules to size, grade and stage of the tumor.

Footnotes

P- Reviewer: Boscá L, Ierardi E S- Editor: Qi Y L- Editor: A E- Editor: Wang CH

References
1.  Delaunoit T, Neczyporenko F, Limburg PJ, Erlichman C. Pathogenesis and risk factors of small bowel adenocarcinoma: a colorectal cancer sibling? Am J Gastroenterol. 2005;100:703-710.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 62]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
2.  Schottenfeld D, Beebe-Dimmer JL, Vigneau FD. The epidemiology and pathogenesis of neoplasia in the small intestine. Ann Epidemiol. 2009;19:58-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 202]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
3.  Dabaja BS, Suki D, Pro B, Bonnen M, Ajani J. Adenocarcinoma of the small bowel: presentation, prognostic factors, and outcome of 217 patients. Cancer. 2004;101:518-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 357]  [Cited by in F6Publishing: 316]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
4.  Howe JR, Karnell LH, Menck HR, Scott-Conner C. The American College of Surgeons Commission on Cancer and the American Cancer Society. Adenocarcinoma of the small bowel: review of the National Cancer Data Base, 1985-1995. Cancer. 1999;86:2693-2706.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Elzagheid A, Emaetig F, Buhmeida A, Laato M, El-Faitori O, Syrjänen K, Collan Y, Pyrhönen S. Loss of MUC2 expression predicts disease recurrence and poor outcome in colorectal carcinoma. Tumour Biol. 2013;34:621-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 26]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
6.  Lugli A, Zlobec I, Baker K, Minoo P, Tornillo L, Terracciano L, Jass JR. Prognostic significance of mucins in colorectal cancer with different DNA mismatch-repair status. J Clin Pathol. 2007;60:534-539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
7.  Biemer-Hüttmann AE, Walsh MD, McGuckin MA, Simms LA, Young J, Leggett BA, Jass JR. Mucin core protein expression in colorectal cancers with high levels of microsatellite instability indicates a novel pathway of morphogenesis. Clin Cancer Res. 2000;6:1909-1916.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Kocer B, Soran A, Erdogan S, Karabeyoglu M, Yildirim O, Eroglu A, Bozkurt B, Cengiz O. Expression of MUC5AC in colorectal carcinoma and relationship with prognosis. Pathol Int. 2002;52:470-477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 53]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
9.  Patai AV, Molnár B, Tulassay Z, Sipos F. Serrated pathway: alternative route to colorectal cancer. World J Gastroenterol. 2013;19:607-615.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 55]  [Cited by in F6Publishing: 52]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
10.  Fujita K, Hirahashi M, Yamamoto H, Matsumoto T, Gushima M, Oda Y, Kishimoto J, Yao T, Iida M, Tsuneyoshi M. Mucin core protein expression in serrated polyps of the large intestine. Virchows Arch. 2010;457:443-449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
11.  Yao T, Takata M, Tustsumi S, Nishiyama K, Taguchi K, Nagai E, Tsuneyoshi M. Phenotypic expression of gastrointestinal differentiation markers in colorectal adenocarcinomas with liver metastasis. Pathology. 2002;34:556-560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 34]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
12.  Ohji Y, Yao T, Eguchi T, Yamada T, Hirahashi M, Iida M, Tsuneyoshi M. Evaluation of risk of liver metastasis in colorectal adenocarcinoma based on the combination of risk factors including CD10 expression: multivariate analysis of clinicopathological and immunohistochemical factors. Oncol Rep. 2007;17:525-530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
13.  Yao T, Tsutsumi S, Akaiwa Y, Takata M, Nishiyama K, Kabashima A, Tsuneyoshi M. Phenotypic expression of colorectal adenocarcinomas with reference to tumor development and biological behavior. Jpn J Cancer Res. 2001;92:755-761.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 36]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
14.  Lichtenstein IL, Amid PK, Shulman AG. The iliopubic tract. The key to inguinal herniorrhaphy? Int Surg. 1990;75:244-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
15.  Fujita K, Yamamoto H, Matsumoto T, Hirahashi M, Gushima M, Kishimoto J, Nishiyama K, Taguchi T, Yao T, Oda Y. Sessile serrated adenoma with early neoplastic progression: a clinicopathologic and molecular study. Am J Surg Pathol. 2011;35:295-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 83]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
16.  Boland CR, Thibodeau SN, Hamilton SR, Sidransky D, Eshleman JR, Burt RW, Meltzer SJ, Rodriguez-Bigas MA, Fodde R, Ranzani GN. A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res. 1998;58:5248-5257.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Sobin LH, Gospodarowicz MK, Wittekind C.  UICC TNM classification of Malignant tumors. 7th ed. New York: John Wiley & Sons 2009; .  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Iwase T, Kushima R, Mukaisho K, Mitsufuji S, Okanoue T, Hattori T. Overexpression of CD10 and reduced MUC2 expression correlate with the development and progression of colorectal neoplasms. Pathol Res Pract. 2005;201:83-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
19.  Yasuda M, Johbu I, Yoshihiro S, Nobue K, Keiichi T, Naoki O, Yoshiyuki O. Availability of CD10 as a Histopathological Diagnostic Marker. Acta Histochem Cytochem. 2005;38:17-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
20.  Fujimoto Y, Nakanishi Y, Sekine S, Yoshimura K, Akasu T, Moriya Y, Shimoda T. CD10 expression in colorectal carcinoma correlates with liver metastasis. Dis Colon Rectum. 2005;48:1883-1889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 49]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
21.  Carl-McGrath S, Lendeckel U, Ebert M, Wolter AB, Roessner A, Röcken C. The ectopeptidases CD10, CD13, CD26, and CD143 are upregulated in gastric cancer. Int J Oncol. 2004;25:1223-1232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
22.  Yasugi A, Yashima K, Hara A, Koda M, Kawaguchi K, Harada K, Andachi H, Murawaki Y. Fhit, Mlh1, P53 and phenotypic expression in the early stage of colorectal neoplasms. Oncol Rep. 2008;19:41-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
23.  Koseki K. Subclassification of Well Differentiated Gastric Cancer with Reference to Biological Behavior and Malignancy, Gastric Type vs. Intestinal Type, and Papillary Carcinoma vs. Tublar Carcinoma. Stomach and Intestine. 1999;34:507-512.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Kabashima A, Yao T, Sugimachi K, Tsuneyoshi M. Gastric or intestinal phenotypic expression in the carcinomas and background mucosa of multiple early gastric carcinomas. Histopathology. 2000;37:513-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 48]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
25.  Velcich A, Yang W, Heyer J, Fragale A, Nicholas C, Viani S, Kucherlapati R, Lipkin M, Yang K, Augenlicht L. Colorectal cancer in mice genetically deficient in the mucin Muc2. Science. 2002;295:1726-1729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 681]  [Cited by in F6Publishing: 688]  [Article Influence: 31.3]  [Reference Citation Analysis (0)]
26.  Bettington M, Walker N, Clouston A, Brown I, Leggett B, Whitehall V. The serrated pathway to colorectal carcinoma: current concepts and challenges. Histopathology. 2013;62:367-386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 309]  [Cited by in F6Publishing: 323]  [Article Influence: 29.4]  [Reference Citation Analysis (0)]
27.  Jass JR. Mucin core proteins as differentiation markers in the gastrointestinal tract. Histopathology. 2000;37:561-564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
28.  Arai T, Kasahara I, Sawabe M, Kanazawa N, Kuroiwa K, Honma N, Aida J, Takubo K. Microsatellite-unstable mucinous colorectal carcinoma occurring in the elderly: comparison with medullary type poorly differentiated adenocarcinoma. Pathol Int. 2007;57:205-212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
29.  Fu T, Pappou EP, Guzzetta AA, Jeschke J, Kwak R, Dave P, Hooker CM, Morgan R, Baylin SB, Iacobuzio-Donahue CA. CpG island methylator phenotype-positive tumors in the absence of MLH1 methylation constitute a distinct subset of duodenal adenocarcinomas and are associated with poor prognosis. Clin Cancer Res. 2012;18:4743-4752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 396]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
30.  Bläker H, Helmchen B, Bönisch A, Aulmann S, Penzel R, Otto HF, Rieker RJ. Mutational activation of the RAS-RAF-MAPK and the Wnt pathway in small intestinal adenocarcinomas. Scand J Gastroenterol. 2004;39:748-753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 56]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
31.  Schönleben F, Qiu W, Allendorf JD, Chabot JA, Remotti HE, Su GH. Molecular analysis of PIK3CA, BRAF, and RAS oncogenes in periampullary and ampullary adenomas and carcinomas. J Gastrointest Surg. 2009;13:1510-1516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 44]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
32.  Arai M, Shimizu S, Imai Y, Nakatsuru Y, Oda H, Oohara T, Ishikawa T. Mutations of the Ki-ras, p53 and APC genes in adenocarcinomas of the human small intestine. Int J Cancer. 1997;70:390-395.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Michel S, Kloor M, Singh S, Gdynia G, Roth W, von Knebel Doeberitz M, Schirmacher P, Bläker H. Coding microsatellite instability analysis in microsatellite unstable small intestinal adenocarcinomas identifies MARCKS as a common target of inactivation. Mol Carcinog. 2010;49:175-182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 14]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
34.  Warth A, Kloor M, Schirmacher P, Bläker H. Genetics and epigenetics of small bowel adenocarcinoma: the interactions of CIN, MSI, and CIMP. Mod Pathol. 2011;24:564-570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 49]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
35.  Planck M, Ericson K, Piotrowska Z, Halvarsson B, Rambech E, Nilbert M. Microsatellite instability and expression of MLH1 and MSH2 in carcinomas of the small intestine. Cancer. 2003;97:1551-1557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 83]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
36.  Mangold E, Pagenstecher C, Friedl W, Fischer HP, Merkelbach-Bruse S, Ohlendorf M, Friedrichs N, Aretz S, Buettner R, Propping P. Tumours from MSH2 mutation carriers show loss of MSH2 expression but many tumours from MLH1 mutation carriers exhibit weak positive MLH1 staining. J Pathol. 2005;207:385-395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 78]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
37.  Rashid A, Hamilton SR. Genetic alterations in sporadic and Crohn’s-associated adenocarcinomas of the small intestine. Gastroenterology. 1997;113:127-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 81]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
38.  Sutter T, Arber N, Moss SF, Findling RI, Neugut AI, Weinstein IB, Holt PR. Frequent K-ras mutations in small bowel adenocarcinomas. Dig Dis Sci. 1996;41:115-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 31]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
39.  Nishiyama K, Yao T, Yonemasu H, Yamaguchi K, Tanaka M, Tsuneyoshi M. Overexpression of p53 protein and point mutation of K-ras genes in primary carcinoma of the small intestine. Oncol Rep. 2002;9:293-300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]