Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Oct 8.
Published in final edited form as: Ann Epidemiol. 2009 Jan;19(1):58–69. doi: 10.1016/j.annepidem.2008.10.004

The Epidemiology and Pathogenesis of Neoplasia in the Small Intestine

DAVID SCHOTTENFELD 1, JENNIFER L BEEBE-DIMMER 2,3, FAWN D VIGNEAU 4
PMCID: PMC3792582  NIHMSID: NIHMS521424  PMID: 19064190

Abstract

PURPOSE:

The mucosa of the small intestine encompasses about 90% of the luminal surface area of the digestive system, but only 2% of the total annual gastrointestinal cancer incidence in the United States.

METHODS:

The remarkable contrast in age-standardized cancer incidence between the small and large intestine has been reviewed with respect to the cell type patterns, demographic features, and molecular characteristics of neoplasms.

RESULTS:

Particularly noteworthy is the predominance of adenocarcinoma in the colon, which exceeds 98% of the total incidence by cell type, in contrast to that of 30% to 40% in the small intestine, resulting in an age-standardized ratio of rates exceeding 50-fold. The prevalence of adenomas and carcinomas is most prominent in the duodenum and proximal jejunum. The positive correlation in global incidence rates of small and large intestinal neoplasms and the reciprocal increases in risk of second primary adenocarcinomas suggest that there are common environmental risk factors. The pathophysiology of Crohn inflammatory bowel disease and the elevated risk of adenocarcinoma demonstrate the significance of the impaired integrity of the mucosal barrier and of aberrant immune responses to luminal indigenous and potentially pathogenic microorganisms.

CONCLUSION:

In advancing a putative mechanism for the contrasting mucosal susceptibilities of the small and large intestine, substantial differences are underscored in the diverse taxonomy, concentration and metabolic activity of anaerobic organisms, rate of intestinal transit, changing pH, and the enterohepatic recycling and metabolism of bile acids. Experimental and epidemiologic studies are cited that suggest that the changing microecology, particularly in the colon, is associated with enhanced metabolic activation of ingested and endogenously formed procarcinogenic substrates.

Keywords: Cancer Epidemiology, Molecular Biology, Neuroendocrine Tumor, Adenocarcinoma, Non-Hodgkin Lymphoma, Gastrointestinal Stromal Tumor, Crohn Disease, Celiac Disease, Microbial Flora, Bile Acid Metabolism

INTRODUCTION

Neoplasms of the small intestine are rare throughout the world. Global incidence is generally less than 1.0 per 100,000, ranging from 0.3 to 2.0 per 100,000, when age-standardized to the world population. In the United States, cancers of the small intestine represent 0.4% of total cancer cases and 0.2% of cancer deaths (1). The small intestine, an elongated tube consisting of the duodenum, jejunum and ileum, represents 75% of the length (i.e., about 5–6 meters), extending from the pylorus to the ileocecal valve, and 90% of the absorptive surface area of the esophago-gastrointestinal system. Remarkably only 2% of the total annual cancer incidence of the digestive system occurs in the small intestine. In contrast, approximately 57% of cancers in the digestive system are diagnosed each year in the large intestine, which measures about 1.5 meters in length.

The average annual age-adjusted incidence per 100,000 for cancer of the small intestine, when age-standardized to the U.S. 2000 population, is 1.9 in men, and 1.4 in women. From 1975 to 2000, the rates increased by almost 50%. This trend reflected increases for adenocarcinomas, malignant carcinoid tumors, and lymphomas in men, and for carcinoid tumors and lymphomas in women (2). The age-specific incidence increases for carcinoid tumors, adenocarcinomas, and lymphomas after age 30, whereas the incidence of sarcomas seems to level off after age 70. The incidence of carcinoid tumors and adenocarcinomas are slightly higher in blacks compared with whites, and the reverse is true for lymphomas and sarcomas (3). Adenocarcinomas are most common in the duodenum and proximal jejunum, whereas lymphomas and carcinoid tumors predominate in the ileum and distal jejunum (4). The international incidence of adenocarcinoma of the small intestine is positively correlated with the distribution of colon cancer incidence. The pattern of increased risk of second primary cancers of the small and of the large intestine suggest similar causal mechanisms, rather than the effects of surveillance bias or the carcinogenic effects of treatment of the index primary cancer (5, 6). The U.S. Surveillance, Epidemiology and End Results (SEER) cohort of male and female patients who had survived 5 or more years after the diagnosis of cancer of the small intestine, experienced more than a two-fold increase in risk (risk ratio [RR] = 2.62; 95% confidence interval [CI] = 1.77, 3.75) of colon cancer. Similarly, the cohort of patients with colon cancer exhibited an increased risk (RR = 3.44; 95% CI = 3.00, 3.95) of cancer of the small intestine (7). The reciprocal increases in risks were amplified in subcohorts in whom the index primary cancer was diagnosed before 60 years of age, suggesting interactions with genetic susceptibility factors (8). The increases in risk of second primary colorectal cancer have been described in association with a prior diagnosis of adenocarcinoma or carcinoid tumor in the small intestine (9-11).

MORPHOLOGY AND MOLECULAR CHARACTERISTICS OF ENTERIC NEOPLASMS

The mucosal layer of the small intestine is replaced in 4–7 days (12) and consists of absorptive, glandular, and neuroendocrine cells that line the crypts and villi. The crypt epithelium functions in cell proliferation and cell renewal. Pluripotential stem cells are located at the base of each crypt, from which undifferentiated reserve cells migrate toward the lumen and differentiate into absorptive cells or enterocytes, or into mucin-secreting goblet cells, before undergoing apoptosis (13, 14). The villi are mucosal folds of columnar epithelial cells that amplify the absorptive surface area of the intestinal lumen. The tunica mucosa of the villi rests on a basement membrane, lamina propria, and muscularis mucosa. The lamina propria contains lymphocytes, macrophages, and plasma cells. The plasma cells secrete the immunoglobulin IgA that diffuses into the intestinal lumen or lamina propria. Mucosa-associated lymphoid cells are scattered throughout the mucosa of the small intestine, and in the ileum aggregate as macroscopic Peyer patches. Antigens that gain access to mucosal macrophages and Peyer patches activate B and T lymphocytes that function in antigen processing and immunosurveillance.

Neuroendocrine Tumors

Carcinoid tumors or argentaffinomas of the gastrointestinal tract are neuroendocrine tumors, derived from enterochromaffin or Kulchitsky cells that are capable of producing serotonin (5-hydroxytryptamine). Neuroendocrine tumors comprise a heterogeneous group of neoplasms that exhibit distinctive morphologic and biologic features. Such tumors originate from pancreatic islet cells, gastro-enteric tissues, gall bladder and biliary duct system, respiratory epithelium, and thyroid parafollicular cells (15, 16). The neuroendocrine cells are capable of producing various polypeptide hormones, including serotonin, gastrin, histamine, insulin, glucagon, vasoactive intestinal peptide, calcitonin and somatostatin. Endocrine secretion is regulated by G protein-coupled receptors, ion-gated receptors, and receptors with tyrosine kinase activity (17). Gastrointestinal neuroendocrine tumors may be classified according to their embryologic origin into tumors of the foregut (bronchopulmonary, stomach, pancreas, duodenum), midgut (jejunum, ileum, appendix, right colon), and hind gut (left colon, rectum). The classification "neuroendocrine" is derived from their functional relationships to multipotential stem cells that are associated with the secretion of marker proteins such as neurokinin A, chromogranin A, synaptophysin, and urinary 5-hydroxy indoleacetic acid. Neuroendocrine tumors of the mediastinum and of gastro-duodenopancreatic cells may be associated with the hereditary multiple endocrine neoplasia (MEN) syndrome (type-1), von-Hippel-Lindau disease, neurofibromatosis (type-1), hereditary nonpolyposis colorectal cancer, and tuberous sclerosis. The type-1 MEN syndrome is an autosomal dominant disorder with phenotypic features of multifocal parathyroid tumors, adenomas of the anterior pituitary, pancreatic and duodenal neuroendocrine tumors, and carcinoma of the thymus gland (18).

Within the small intestine, neuroendocrine carcinoid tumors occur predominately in the ileum, and rarely in the duodenum. In the duodenum, these tumors are classified as gastrinomas. In the SEER population, the average annual age-adjusted incidence per 1,000,000 based on the 1973–2005 registration for carcinoid tumors of the small intestine was 7.7 (white men), 5.5 (white women), 14.4 (black men), and 8.7 (black women). The incidence per 1,000,000 for carcinoid tumors of the colon was 2.7 (white men), 3.2 (white women), 4.7 (black men), and 4.0 (black women) (Table 1). Over the past 30 years, the SEER registries have documented a 400% increase in the age-adjusted incidence of carcinoid tumors of the small intestine (19). Carcinoid tumors accounted for 42% of the total of small intestinal tumors. In histopathologic studies based on surgical and autopsied patients with carcinoid tumors of the small intestine, up to one-third of cases were multifocal.

TABLE 1.

Incidence rates and frequency of cancers of the small and large intestine by histology, race, and sex, SEER (1973–2005)

Incidence rate (%*)
Primary site and histology White men Black men White
women
Black
women
Small intestine
 Adenocarcinoma 6.8 (30.8) 12.2 (37.2) 4.5 (32.7) 10.2 (45.5)
 Non-Hodgkin 4.9 (22.9) 3.4 (11.4) 2.5 (17.9) 1.5 (7.3)
 lymphoma
 Carcinoid 7.7 (36.0) 14.4 (43.9) 5.5 (39.2) 8.7 (39.7)
 Sarcoma 2.1 (10.3) 2.1 (7.4) 1.5 (10.2) 1.5 (7.5)
Colon
 Adenocarcinoma 447.3 (98.6) 498.7 (98.6) 345.9 (98.7) 402.6 (98.5)
 Non-Hodgkin 2.9 (0.7) 0.9 (0.2) 1.2 (0.3) 1.0 (0.2)
 lymphoma
 Carcinoid 2.7 (0.7) 4.7 (1.0) 3.2 (0.9) 4.0 (1.1)
 Sarcoma 0.5 (0.1) 0.6 (0.1) 0.3 (0.1) 0.5 (0.1)

Note: Rates are per 1,000,000 and age-adjusted to the 2000 US Std Population (19 age groups – Census P25-1130) standard.

Source: Surveillance, Epidemiology, and End Results (SEER) Program (www.seer.cancer.gov) SEER*Stat Database: Incidence – SEER 9 Regs Limited-Use, Nov 2007 Sub (1973–2005).

*

Percent of histologic groups presented – other histologies not included.

Histologic codes include: 8800–8805, 8810, 8811, 8890–8897, 8930, 8933–8936, 8990–8891, 9044, 9120, 9140, 9180, 9540, 9560, 9755, 9758, 9930.

Molecular studies of sporadic gastroenteropancreatic neuroendocrine tumors have demonstrated that chromosomal instability, copy number aberrations, point mutations, methylation abnormalities, and ablation of putative tumor-suppressor pathways are associated with neoplastic transformation and tumor progression. The prominent mutations associated with intestinal adenocarcinomas (e.g., TP53, K-ras, p16, APC and DCC alleles) are not salient molecular events in the pathogenesis of gastrointestinal neuroendocrine tumors (20, 21). Comparative genomic hybridization, loss of heterozygosity, and mutational analyses suggest genetic pathways that are selectively involved in the pathogenesis of gastrointestinal neuroendocrine tumors (22). Hereditary and sporadic gastrointestinal carcinoid tumors that are associated with point mutations or deletions of the MEN 1 gene on chromosome 11q 13 tend to be gastrin cell tumors of duodenal or proximal jejunal origin (23). The MEN 1 gene encodes a protein product "menin" that is expressed ubiquitously in endocrine and non-endocrine tissues. Menin exhibits tumor suppressor activity, presumably by modulating histone methylation in promoters of HOX genes and various cyclin-dependent kinase inhibitors. Distal jejunal and ileal carcinoid tumors tend to be associated more specifically with inactivation or losses in chromosomes 18p, 18q, 11q, 16q, and 9p, and to be associated with overexpression of transforming growth factors α and β, epidermal growth factor, fibroblast growth factor, and neural cell adhesion molecules and their receptors (24, 25).

Adenomas, Epithelial Dysplasia and Adenocarcinoma

In the SEER population, the average annual age-adjusted incidence per 1,000,000, based on the 1973–2005 registration, for adenocarcinoma of the small intestine was 6.8 (white men), 4.5 (white women), 12.2 (black men), and 10.2 (black women). The incidence per 1,000,000 for adenocarcinoma of the colon (excluding rectum) was 447.3 (white men), 345.9 (white women), 498.7 (black men), and 402.6 (black women) (Table 1). Therefore, the ratio of the rates for adenocarcinoma of the colon in white men, when compared with that for adenocarcinoma of the small intestine, was 65.8. Adenocarcinoma composed about 30% to 40% of the total of registered cancers in the small intestine in whites and blacks, respectively, whereas the proportion of total cancers classified as adenocarcinoma in the colon exceeded 98%. The predominant locations in the small intestine for adenocarcinoma are the duodenum and proximal jejunum (Table 2, Fig. 1). The classification of adenocarcinoma of the periampullary region encompasses tumors located in the duodenum, and in the distal common bile and pancreatic ducts. Adenocarcinomas in the duodenal ampulla of Vater may manifest morphologic and biologic features of enteric mucosal or pancreaticobiliary duct epithelium, and are associated with increases in incidence of colorectal adenomas and carcinomas (26, 27). The temporal patterns for the incidence of adenocarcinoma in the small intestine and colon both reflect the higher rates in men compared to women (Figs. 2 and 4) and blacks compared to whites (Figs. 3 and 5). In the survey by Severson et al. (28) that compared average annual rates per 1,000,000 in the SEER database, the incidence of adenocarcinoma of the small intestine increased among black men from 8.2 (1973–1977) to 16.0 (1987–1999), and among white men, the incidence increased from 6.2 to 8.5; among black women, the incidence increased from 5.1 to 15.7, and among white women, from 4.8 to 5.1. In the most recent 5 years of SEER data collection, 2000–2005, the incidence varied slightly among white men from 7.1 in 2000 to 8.9 in 2005, and among black men, from 11.1 to 12.7. Among white women, the incidence varied from 3.9 to 5.4, but among black women, decreased slightly from 11.8 to 10.3. The corresponding annual percentage change increase in the incidence of adenocarcinoma across the 32-year period was lowest among white women (0.9%), 1.4% among white men, 2.4% among black men, and highest among black women (3.0%).

TABLE 2.

Incidence and proportion of cancers of the small intestine by anatomic subsite and histology, SEER (1973–2005)

Anatomic subsite and histology Rate Count %
Duodenum
 Adenocarcinoma 3.0 2,062 58.7
 NHL 0.5 340 9.7
 Carcinoid 0.8 578 16.5
 Sarcoma* 0.3 221 6.3
 Other 0.5 309 8.8
3,510 100.0
Jejunum
 Adenocarcinoma 1.2 839 42.1
 NHL 0.6 446 22.4
 Carcinoid 0.4 279 14.0
 Sarcoma* 0.4 322 16.2
 Other 0.1 105 5.3
1,991 100.0
Ileum
 Adenocarcinoma 0.9 603 15.1
 NHL 1.0 681 17.1
 Carcinoid 3.2 2,282 57.3
 Sarcoma* 0.4 264 6.6
 Other 0.2 153 3.8
3,983 100.0
Other small intestine
 Adenocarcinoma 0.8 576 15.4
 NHL 1.4 979 26.1
 Carcinoid 2.1 1,471 39.2
 Sarcoma* 0.7 501 13.4
 Other 0.3 223 5.9
3, 750 100.0

NHL = non-Hodgkin lymphoma.

Note: Rates are per 1,000,000 and age-adjusted to the 2000 US Std Population (19 age groups – Census P25-1130) standard.

Source: Surveillance, Epidemiology, and End Results (SEER) Program (www.seer.cancer.gov) SEER*Stat Database: Incidence – SEER 9 Regs Limited-Use, Nov 2007 Sub (1973–2005).

*

Histologic codes include: 8800–8805, 8810, 8811, 8890–8897, 8930, 8933–8936, 8990–8891, 9044, 9120, 9140, 9180 9540, 9560, 9755, 9758, 9930.

Other site includes small intestine (not otherwise specified), Meckel’s diverticulum, overlapping segments.

FIGURE 1.

FIGURE 1

Age-adjusted incidence rates of adenocarcinoma of the small intestine and colon by cancer site (SEER, 1973–2005).

FIGURE 2.

FIGURE 2

Age-adjusted incidence rates of adenocarcinoma of the small intestine by gender; all races (SEER, 1973–2005).

FIGURE 4.

FIGURE 4

Age-adjusted incidence rates of adenocarcinoma of the colon by gender, all races (SEER, 1973–2005).

FIGURE 3.

FIGURE 3

Age-adjusted incidence rates of adenocarcinoma of the small intestine by race; all races (SEER, 1973–2005).

FIGURE 5.

FIGURE 5

Age-adjusted incidence rates of adenocarcinoma of the colon by race (SEER, 1973–2005).

Adenocarcinomas may appear as polypoid, infiltrating, or as annular constricting lesions. As in the colon, the adenoma in the small intestine is a precursor of adenocarcinoma (29). Residual adenomatous tissue is observed commonly at the margins of sporadic carcinomas, and of carcinomas in patients with familial adenomatous polyposis. The spatial distribution of enteric adenomas parallels the distribution of carcinomas in enteric sites (30). In familial adenomatous polyposis, the prevalence of adenomas throughout the duodenum is markedly increased, and the relative risk of adenocarcinoma in the duodenum is increased to more than 300 times that of a referent population (31, 32). The morphology of enteric adenomas tends to be more villous, or tubulovillous, than the predominant tubular pattern evidenced in the colon. The risk potential for neoplastic transformation of adenomas increases with components of villous histopathology, increasing size in cross-sectional diameter (e.g., ≥1 cm), and with a higher grade of dysplasia.

Genetic syndromes predisposing to neoplasia in the small intestine are characterized by a familial history, although the type of cancer may appear de novo in the absence of a family history, and the associated cancers are generally diagnosed at a younger age than sporadic tumors. Such cancer-associated syndromes may be attributed to a single predominating nucleotide mutation, or to multiple genes on the same or different chromosomes, and are associated with multicentric precursor and neoplastic lesions in other gastrointestinal or other systemic organs (33). The pathogenesis of sporadic invasive adenocarcinoma in the small intestine involves mutational and epigenetic pathways that simulate the multistep events in colorectal neoplasia. Mutations have been reported in adenomatous polyposis coli, β-catenin, E-cadherin, K-ras, p53, and 18q alleles that presumably participate in the adenoma-carcinoma histogenesis pathway (34-36). In studies of adenocarcinoma of the ampulla of Vater, the nuclear accumulation of β-catenin, and K-ras mutations, has been associated with increased cyclin D1 expression (37, 38). Truncating mutations at the adenomatous polyposis coli 5q locus occur infrequently in small intestinal neoplasia, in contrast to their higher prevalence in colorectal adenomas and carcinomas. The truncated adenomatous polyposis coli protein results in aberrations in critical functions of cell adhesion, cell migration, apoptosis, unregulated intracellular accumulation of β-catenin, and the augmented transcription of specific oncogenes such as cMYC and cyclin D1 (39, 40). Losses at chromosome 18q 21–22 would negatively affect growth mediation by transforming growth factor-β signaling (41). Consistent with colorectal cancer, 10% to 15% of cases of adenocarcinomas of the small intestine exhibit high microsatellite instability indicative of DNA mismatch repair dysfunction (42). Most germ line mutations reside at hMLH1 and hMSH2 loci (43). Individuals affected with hereditary nonpolyposis colorectal cancer are at increased risk of developing extra-colonic carcinomas in the small intestine, endometrium, pancreas, renal pelvis, stomach, liver and biliary tract, and ovary, and central nervous system gliomas. The lifetime risk of small intestinal neoplasms in patients with hereditary nonpolyposis colorectal cancer has been estimated to range from 1% to 4% (44, 45). The tumors are located primarily in the duodenum. The tumors that manifest a high level of microsatellite instability tend to have a distinctive phenotype of poor differentiation, prominent lymphocyte infiltration, and mucin production (46). Microsatellites contain a limited number of DNA sequences repeated in tandem and exhibit polymorphisms in length throughout the genome. Because they are repetitive, microsatellites are prone to strand slippage and errors in replication.

A hallmark of the Peutz-Jeghers syndrome is the presence of hamartomatous polyps in the small intestine, colon, and stomach. The syndrome is an autosomal dominant disorder caused by a germ line mutation in the serine/threonine kinase gene, STK11/ LKB1, a tumor suppressor gene located on chromosome 19p13.3 (47-49). Most mutations are small deletions, insertions, or base substitutions resulting in an abnormal truncated protein. Another prominent feature is the distribution of melanin pigmented lesions on the lips, perioral region, hands, and buccal mucosa. Patients with the syndrome are at increased risk of small bowel, other gastrointestinal, breast, ovarian, uterine cervical, ovarian, testicular, and lung cancers (50-52).

Non-Hodgkin Lymphomas

The non-Hodgkin lymphomas comprise a broad spectrum of lymphoproliferative neoplasms arising from B cells, T cells, and natural killer cells. Primary lymphomas of the small intestine include mucosa-associated lymphoid tissue lymphoma, diffuse large B-cell lymphoma, immunoproliferative lymphoma, also classified as immunoproliferative small intestinal disease or IPSID, and enteropathy-associated T-cell lymphoma (EATL). Diffuse large B-cell, immunoblastic, and Burkitt lymphomas arising in the small intestine have been reported in patients with human immunodeficiency virus infection (53). In the SEER population, the average annual age-adjusted incidence per 1,000,000, based on 1973–2005 registration, for non-Hodgkin lymphomas was 4.9 (white men), 3.4 (black men), 2.5 (white women), and 1.5 (black women) (Table 1). The lymphomas accounted for approximately 18% to 23% of enteric neoplasms in whites, and 7% to 11% in blacks. In contrast, non-Hodgkin lymphomas in the colon accounted for less than 1% of all registered neoplasms.

The majority of enteric lymphomas are B-cell lymphomas. The evolution and progression of B-cell lymphomas involves complex interactions of host immune cells to foreign antigens, accompanied by transforming molecular events, such as chromosomal translocations, and constitutive activation of signaling pathways (54, 55). Extranodal lymphomas arising from marginal zone lymphoid tissue are associated with chronic antigenic stimulation by microbial pathogens, e.g., Helicobacter pylori (56), Chlamydia psittaci (57), Campylobacter jejuni (58), hepatitis C virus (59), or autoantigens. The proximal small intestine is the most common site of origin of immunoproliferative Ig α heavy chain disease, or mucosa-associated lymphoid tissue-type lymphoma. The prevalence of IPSID is concentrated in the young adult population in the Middle East and North Africa (60, 61). The early histopathology shows a lymphocytic and plasmacytic infiltration in the bowel wall that may evolve into diffuse large B-cell lymphoma. The limited geographic distribution of IPSID and the apparent efficacy of treatment with antimicrobial agents have suggested that chronic infection is causal in an antigen-stimulated lymphoproliferative disease. In this regard, persistent infection with C. jejuni has been implicated based on DNA sequencing, fluorescence in situ hybridization and histochemical studies of biopsy specimens (58).

Patients with a history of celiac disease are at increased risk of T-cell and, with lesser frequency, B-cell non-Hodgkin lymphoma, and adenocarcinoma in the small intestine (62, 63). The relative risk of enteropathy-associated T-cell lymphoma in the small intestine is increased approximately 10-to 20-fold in patients with protracted clinical celiac disease (64, 65). Serologic screening using antigliadin and antiendomysial antibody testing, followed by confirmatory endoscopic biopsy, has estimated that the prevalence of celiac disease in Europe and the United States is about 0.5% to 1% (66, 67). Celiac disease is an autoimmune disorder triggered by the ingestion of gluten peptides contained in wheat, rye, and barley. The alcohol-soluble fraction of gluten, namely gliadin, and similar prolamins in rye and barley, are toxic in genetically susceptible individuals (68). Predisposition to gluten sensitivity has been mapped to the HLA-D region on chromosome 6 (69). In patients with celiac disease, the immune reaction to gliadin evokes a chronic inflammatory response concentrated primarily in the mucosa of the proximal small intestine that is manifested by intra-epithelial T-cell lymphocytosis, hyperplasia of crypt epithelium, and villous atrophy (70, 71). The extent of mucosal damage is correlated with serum titers of IgA endomysial antibodies and tissue transglutaminase antibodies. Celiac patients are at increased risk of a wide spectrum of autoimmune disorders, including dermatitis herpetiformis (72-74).

Mesenchymal Tumors

The average annual incidence per 1,000,000 persons of sarcomas of the small intestine in men (2.1) exceeds that in women (1.5). The most commonly occurring sarcoma in the small intestine is the gastrointestinal stromal tumor or GIST. These mesenchymal tumors arise most often in the stomach, but have been diagnosed in the colon, esophagus, mesentery, omentum, and peritoneum (75). Before the use of immunohistochemistry and the demonstration of expression of cell markers such as CD34 and CD117/ KIT, many of these tumors were misclassified as leiomyosarcomas or neurogenic tumors (76, 77). The cell of origin of GIST neoplasms is the interstitial cell of Cajal, which is interspersed between the circular and longitudinal muscle layers of the gastrointestinal tract, and normally functions within the autonomic nervous system to regulate intestinal motility. The Cajal pleomorphic cell expresses KIT tyrosine kinase receptor (CD117), which on phosphorylation activates downstream signaling pathways (78). Gain-of-function mutations in the KIT proto-oncogene have been shown in more than 90% of GIST tumors (79, 80). Most KIT mutations have originated in somatic cells, however, GIST tumors arising in the small intestine have been reported in patients with type I neurofibromatosis (78).

LIFESTYLE RISK FACTORS

Tobacco and Alcohol

Risk factors such as the regular use of tobacco (81, 82), excessive alcohol consumption and the interaction with folate deficiency (83), obesity (84, 85), and nutrition (86-88) have been studied extensively in patients with colon cancer. In contrast, studies of patients with adenocarcinoma or malignant carcinoid tumors of the small intestine have been constrained because of the rarity and biologic heterogeneity of the major types of tumor. Studies of tobacco and alcohol that were based solely on the review of medical records (9, 89), or on next-of-kin surrogate interviews (90), have not explored the effects of average concentrations of exposure, cumulative lifetime exposures, induction-latency intervals, or dose-response trends. In a population-based study conducted in Europe, Kaerlev et al. (91) reported modest estimates of risk for malignant carcinoid tumor associated with smoking (OR = 1.9; 95% CI = 1.1, 3.2), but no association with adenocarcinoma of the small intestine (92). In a population-based study conducted in Los Angeles County, Wu et al. (93) reported that the odds ratio for smoking cigarettes and adenocarcinoma of the small intestine, after adjusting for use of ethanol, was 2.6 (95% CI = 0.6, 11.6) in men and 1.1 (95% CI = 0.4, 2.7) in women. The estimations of risk were not altered in comparisons of former with current smokers, or in relation to age at starting smoking, number of cigarettes smoked per day, or total years of cigarette smoking.

Consumption of alcohol has also been associated with small bowel adenocarcinomas independently of smoking history (9, 92, 93). In the study by Wu et al. (93) of the consumption of all forms of alcoholic beverages, expressed in grams of ethanol per day, and adjusted for cigarette smoking, the risk of adenocarcinoma was increased 2.9-fold (95% CI = 1.2, 7.1) for patients who consumed more than 80 g (> 6 beverage drinks) per day. Kaerlev et al. (92) found intake of beer and spirits (≥24 g/day), but not wine, was associated with an approximate 3.5-fold increase in risk for adenocarcinoma. The preliminary findings of Chen et al., indicating an association between alcohol intake and carcinoid tumors, have not been replicated (91, 94). Various tumorigenic mechanisms have been attributed to alcohol and its genotoxic metabolite, acetaldehyde, including impaired DNA methylation, inflammation-mediated oxidative stress, and perturbation of the intestinal mucosal immune response (95, 96).

Diet and Body Mass

Dietary factors have been purported to play a role in small intestine neoplasia given that global incidence rates for small and large bowel cancers parallel one another and correlate with per capita consumption of dietary fat and red meat (3, 90). Negri et al. (97) reported an elevated risk of adenocarcinoma of the small intestine among the highest consumers of sugar, refined carbohydrates, and red meat, and a reduced risk associated with higher intake of coffee, fish, fruits, and vegetables (97). Likewise, Chow et al. (90) reported an elevated risk of dying from small bowel cancer associated with intake of red meat and cured or smoked foods.

Increased body mass is consistently linked to cancer of the colon, yet the few studies examining the relationship between obesity and cancer of the small intestine have produced mixed results (90, 94, 97-100). Wolk et al. (98) observed significant elevation in risk of small bowel cancer among obese men and women (standardized incidence ratio = 2.8; 95% CI = 1.6, 4.5). In a large cohort of U.S. veterans, obesity (body mass index = ≥30 kg/m2) was associated with an approximate 60% increase in risk of cancer of the small intestine among white men (RR = 1.58; 95% CI = 1.18, 2.12), but not among black men (RR = 1.07; 95% CI = 0.54, 2.08). This risk was further elevated among whites when restricted to duodenal cancers (RR = 2.10; 95% CI = 1.38, 3.22) (99). Similarly, in a Norwegian cohort study of 1,162 cases in 2 million men and women, obese men were 60% more likely to be diagnosed with small bowel cancer(RR= 1.59; 95% CI = 1.13, 2.23) compared to men with normal body mass index (18.5–24.9 kg/m2). However, no relationship between BMI and cancer risk was observed among women, except for cancers of the duodenum (RR = 1.67; 95% CI = 1.00, 2.80) (100). Other studies of obesity and cancer of the small intestine have shown either no association (90) or an inverse relationship with body mass (94, 97).

INTERSECTING PATHOGENIC MECHANISMS

Chronic Inflammation and a Cascade of Cytokines

Chronic inflammatory bowel disease includes two clinically and pathologically distinctive autoimmune entities, Crohn disease (CD) and ulcerative colitis. Crohn disease is a chronic inflammatory condition presenting principally in the distal small intestine, but may involve the colon, mainly the proximal colon, rectum, anus, and perianal tissues, other digestive organs, and tissues outside of the digestive tract. Nearly 50% of patients manifest inflammatory disease that involves both ileum and colon. Focal intestinal inflammation associated with superficial mucosal ulcerations, with segmental distribution or "skip lesions," are early characteristic pathologic features. With increasing chronicity, the ulcerating lesions coalesce and the inflammatory response tends to be transmural and granulomatous (101). The granulomatous inflammation of CD comprises aggregates of macrophages, lymphocytes, plasma cells, and multinucleated giant cells that are formed in response to the release of cytokines such as tumor necrosis factor (102, 103).

The determination of the true incidence of CD is problematic because of the lack of population-based registration data and the difficulty in distinguishing early onset CD from other inflammatory diseases of the small or large intestine. The estimated incidence of CD, ranging from 1 to 15 per 100,000, is highest in North America, northern and western European countries, and in American- or European-born Jews. The incidence of adenocarcinoma in the small or large intestine is increased in patients with CD (104-106). As in ulcerative colitis, the risk of cancer in patients with CD is correlated with early age at onset, duration, and extent of disease in the intestinal organ, and the prevalence of precancerous dysplastic lesions. In population-based studies of patients with CD in the small intestine, the relative risk of carcinoma in the small intestine has been estimated to be increased 10- to 40-fold (107). The cumulative risk at 10 years was estimated as 0.2% (95% CI = 0.0–0.8), and at 25 years, 2.2% (95% CI = 0.7–6.4) (108). In a meta-analysis of six population-based cohort studies, the age-standardized incidence rate ratio for colorectal cancer in patients with CD of the colon was estimated as 2.5 (95% CI = 1.7–3.5) (109).

The integrity of the mucosal barrier lining the intestinal tract, which is maintained by tight junctions between adjacent epithelial cells and relative impermeability of the apical villous brush border, serves an important function in the innate immune system. The intestinal tract is exposed continuously to commensal indigenous microbial flora and potentially to pathogenic organisms (110). Enteric epithelial cells, in particular Paneth cells secrete antimicrobial substances such as mucus, lysozymes, cysteine-rich defensins, and IgA (111). The constitutive and inducible production of various defensin peptides in the intestinal mucosa limits the invasion and adherence of commensal and pathogenic microorganisms (112). The pathophysiology of CD underscores the pathogenic significance of permeability of the mucosal barrier and of the aberrant immune response by effector lymphocytes and proinflammatory cytokines to indigenous luminal microorganisms or foreign antigens (113-115). The inflammatory response mediated by CD4+ Th1-cells is exaggerated and cytotoxic. A salient immunopathogenic mechanism in CD is the resistance of activated T cells to undergo apoptosis (116).

The pathogenesis of CD as shown in animal models and human studies would suggest that protracted intestinal inflammation and its sequelae arise from the interactions of the luminal microflora, immune-mediated tissue injury, and genetic susceptibility (117, 118). Genome-wide linkage analyses have identified 10 suspected loci associated with susceptibility to inflammatory bowel disease (119, 120). The genetic basis for susceptibility to CD is most consistent with a polygenic mode of inheritance (36). A potentially significant gene-phenotype expressed in CD has been linked with at least three variants of the caspase recruitment domain gene positioned at chromosome 16q12 (121). The gene product of caspase recruitment domain 15 is the NOD2 protein, an intracellular receptor for a component of the bacterial cell wall that activates cells of the innate immune system (122). Aberrant NOD2 protein has been shown to result in defective pattern recognition and clearance of invasive bacteria by macrophages and diminished production of microbicidal defensins (123, 124).

As evidenced by the multistep pathogenesis of CD and dysplasia-adenocarcinoma of the small intestine, recurrent or persistent inflammation, whether due to an autoimmune disease or chronic exposure to an infectious or chemical agent, may induce or promote tumorigenesis in a susceptible host and target tissue (125, 126). Phases of cancer promotion and progression are intimately linked to a dysfunctional cytokine network. Cytokines represent a family of biologic response modifiers including interleukins, chemokines, growth factors, and extracellular proteases that interact with cell surface receptors and target genes that influence clonal neoplastic cell proliferation, angiogenesis and migration of tumor cells through the basement membrane and into the stromal matrix (127). In addition, the metabolic products of activated inflammatory cells are accompanied by excessive formation of reactive oxygen and nitrogen species that are potentially damaging to DNA and the integrity of cell surface membranes (128).

Microbial Flora Interacting With Endogenous and Ingested Substrates

In addition to the consideration of the mechanisms linked with chronic inflammation and immune dysfunction, the contrasting incidence of adenocarcinoma of the small and large intestine may be viewed in the context of the differences in microbial density and taxonomy of anaerobic microorganisms in the fecal stream, the enterohepatic metabolism of bile acids and their potential role in tumorigenesis, and rate of intestinal transit. The contents of the fecal stream remain liquid throughout the small intestine. Chyme is transported through the small intestine by peristaltic and segmenting waves, requiring about 3–5 hours for passage from the pylorus to the ileocecal valve. The right colon carries out complex functions that include mixing and fermentation of the ileal effluent, and excretion and desiccation of the intraluminal contents to form stool; the left colon serves as a conduit for desiccation and transport of stool to the rectosigmoid region. Although the rate of transit in the small intestine is correlated directly with the rate of transit in the colon, transit time through the small intestine is generally more rapid per unit length than in the colon and rectum. The principal points of delay in the large intestine are in the cecum, ascending colon, sigmoid colon, and rectum (129). The electrical rhythm of the small and large intestine is controlled by the enteric nervous system and neuro-hormonal mechanisms, which in turn are modulated by the dendritic and T-cells of the immune system (130). Slowing of the rate of intestinal transit augments the enterohepatic recycling of bile acids, the excreted fecal mass of microorganisms, anaerobic bacterial fermentation of polysaccharides and production of short chain fatty acids, and the rising pH of the fecal stream (131, 132).

A diverse and dynamic microbial ecosystem is normally resident within the lumen or adherent to the mucosal surface of the small and large intestine. The prevalence of microbiota in different anatomic areas of the gastrointestinal tract is influenced by the pH, bowel motility, symbiotic or antagonistic interactions among the various species of microorganisms, mucosal immune mechanisms, oxidationreduction potential of the luminal contents and mucosa, and nutritional metabolites. Decreased oxidation-reduction potential predisposes to colonization with obligate anaerobic bacteria (132).

The upper two-thirds of the small intestine contain low concentrations of aerophilic Gram-positive bacteria such as lactobacilli and enterococci. The density of microbiota increases markedly in the distal ileum, and may contain up to one million anaerobic organisms per gram of luminal contents. The large intestine harbors over 500 species of bacteria, mainly obligate anaerobic Gram-negative and Gram-positive organisms, such as bacteroides, bifidobacteria, Clostridia, and Enterobacteriaceae (133). The anaerobic microbial ecosystem in the cecum, at a concentration in excess of one billion bacteria per gram of luminal contents, flourishes because of the relative stagnation of the fecal stream, and the very low oxidation-reduction potential (134). It is of interest that the increased risk of adenocarcinoma in patients with CD occurring in surgically bypassed loops of chronically inflamed ileum has been attributed to stagnation of intestinal contents accompanied by overgrowth of anaerobic bacteria (135).

The enormous number and diversity of microorganisms in the intestinal tract are important in the development of gut-associated lymphoid tissue and immune capacity (136-138). Conversely, the intestinal bacteria provide the metabolic capacity to facilitate xenobiotic transformation and potential synthesis of carcinogenic metabolites. Intestinal anaerobic microorganisms generate various metabolizing enzymes, such as β-glucuronidase, β-glucosidase, sulfatase, nitrate and nitro reductases, and decarboxylases, which act on various substrates such as the bile acids, fatty acids, and steroid molecules. The conversion of ingested chemical agents to genotoxic molecules has been investigated in antibiotic-treated, germ-free rodents. For example, cycasin, found in Cycadeceae plants, a methylazoxymethanol glucoside, is not tumorigenic when administered parenterally or orally to germ-free rats. However, when fed orally to rats with normal intestinal microbial flora, less than 50% of the conjugated compound was recovered in the feces and urine, and adenocarcinomas were induced in the large intestine, liver, biliary duct system, and kidney. It was apparent that microbial β-glucosidase had converted cycasin to unconjugated methylazoxymethanol, an active mutagenic and tumorigenic metabolite (139). Similarly, tumor induction by dimethylhydrazine or 2,3-dimethyl 4-aminobiphenyl is enhanced by the β-glucuronidase enzymatic activity of intestinal bacteria (140-142).

Greater than 95% of the bile salts that are synthesized in the liver are reabsorbed either by passive diffusion in the proximal jejunum, or by active transport in the distal ileum. The bile salts are then transported via the portal vein back to the liver where they are absorbed by hepatic cells and again secreted as bile. The enterohepatic recirculation of bile salts recycles about 6–8 times daily (143). The bile salts are the ionized form of the bile acid molecule. The carboxyl group in the side chain of the bile salt molecule when activated can react with glycine or taurine forming amides known as conjugated bile salts. In glycocholic and glycochenocholic acids, the bile acids are conjugated with glycine, whereas taurocholic and taurochenocholic acids are conjugated with taurine. Intestinal anaerobic bacteria, for example species of the Bacteroides fragilis group, deconjugate and dehydroxylate the bile salts by removing glycine and taurine residues and the hydroxyl group at position 7 (144). The primary bile salts are then biochemically transformed into the secondary bile acids, deoxycholic acid and lithocholic acid. The deconjugated and dehydroxylated bile salts are less soluble in intestinal chyme and are therefore less readily absorbed from the intestinal lumen than the bile salts that have not been subjected to bacterial metabolism. Based on both experimental and observational epidemiologic studies, deoxycholic acid has been classified as a potential tumor promoter in conjunction with other genotoxic agents (145-147). Studies of concentration levels of deoxycholic acid in both fecal and serum samples have been associated with colorectal adenomas and cancer (148-150). The relatively prominent distribution of adenocarcinoma in the duodenum and proximal jejunum, particularly after cholecystectomy, has been attributed to proximity to the juncture of the common bile duct (151). Review of physiologic, pathologic, experimental, and epidemiologic studies suggest that the changing microecology, particularly in the colon, is associated with enhanced metabolic activation of ingested and endogenously-formed procarcinogenic substrates.

Acknowledgments

This study was supported by National Institutes of Health Grant 1K07CA127214-01A1 and SEER contract NO1PC-35145.

Selected Abbreviations and Acronyms

SEER

Surveillance, Epidemiology and End Results

MEN

multiple endocrine neoplasia

GIST

gastrointestinal stromal tumor

CD

Crohn disease

REFERENCES

  • 1.Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, et al. Cancer statistics, 2008. CA Cancer J Clin. 2008;58:71–96. doi: 10.3322/CA.2007.0010. [DOI] [PubMed] [Google Scholar]
  • 2.Chow JS, Chen CC, Ahsan H, Neugut AI. A population-based study of the incidence of malignant small bowel tumors: SEER, 1973-1990. Int J Epidemiol. 1996;25:722–728. doi: 10.1093/ije/25.4.722. [DOI] [PubMed] [Google Scholar]
  • 3.Haselkorn T, Whittemore AS, Lilienfeld DE. Incidence of small bowel cancer in the United States and worldwide: geographic, temporal, and racial differences. Cancer Causes Control. 2005;16:781–787. doi: 10.1007/s10552-005-3635-6. [DOI] [PubMed] [Google Scholar]
  • 4.DiSario JA, Burt RW, Vargas H, McWhorter WP. Small bowel cancer: epidemiological and clinical characteristics from a population-based registry. Am J Gastroenterol. 1994;89:699–701. [PubMed] [Google Scholar]
  • 5.Neugut AI, Santos J. The association between cancers of the small and large bowel. Cancer Epidemiol Biomarkers Prev. 1993;2:551–553. [PubMed] [Google Scholar]
  • 6.Scelo G, Boffetta P, Hemminki K, Pukkala E, Olsen JH, Andersen A, et al. Associations between small intestine cancer and other primary cancers: an international population-based study. Int J Cancer. 2006;118:189–196. doi: 10.1002/ijc.21284. [DOI] [PubMed] [Google Scholar]
  • 7.Curtis RE, Freedman DM, Ron E. New malignancies among cancer survivors: SEER cancer registries, 1973–2000. National Cancer Institute; Bethesda (MD): 2006. NIH Publication No. 05-5302. Ref Type: Generic. [Google Scholar]
  • 8.Evans HS, Moller H, Robinson D, Lewis CM, Bell CM, Hodgson SV. The risk of subsequent primary cancers after colorectal cancer in southeast England. Gut. 2002;50:647–652. doi: 10.1136/gut.50.5.647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Babovic-Vuksanovic D, Constantinou CL, Rubin J, Rowland CM, Schaid DJ, Karnes PS. Familial occurrence of carcinoid tumors and association with other malignant neoplasms. Cancer Epidemiol Biomarkers Prev. 1999;8:715–719. [PubMed] [Google Scholar]
  • 10.Chen CC, Neugut AI, Rotterdam H. Risk factors for adenocarcinomas and malignant carcinoids of the small intestine: preliminary findings. Cancer Epidemiol Biomarkers Prev. 1994;3:205–207. [PubMed] [Google Scholar]
  • 11.Tichansky DS, Cagir B, Borrazzo E, Topham A, Palazzo J, Weaver EJ, et al. Risk of second cancers in patients with colorectal carcinoids. Dis Colon Rectum. 2002;45:91–97. doi: 10.1007/s10350-004-6119-y. [DOI] [PubMed] [Google Scholar]
  • 12.Grossmann J, Walther K, Artinger M, Rummele P, Woenckhaus M, Scholmerich J. Induction of apoptosis before shedding of human intestinal epithelial cells. Am J Gastroenterol. 2002;97:1421–1428. doi: 10.1111/j.1572-0241.2002.05787.x. [DOI] [PubMed] [Google Scholar]
  • 13.Playford RJ. Tales from the human crypt—intestinal stem cell repertoire and the origins of human cancer. J Pathol. 1998;185:119–122. doi: 10.1002/(SICI)1096-9896(199806)185:2<119::AID-PATH83>3.0.CO;2-3. [DOI] [PubMed] [Google Scholar]
  • 14.Bullen TF, Forrest S, Campbell F, Dodson AR, Hershman MJ, Pritchard DM, et al. Characterization of epithelial cell shedding from human small intestine. Lab Invest. 2006;86:1052–1063. doi: 10.1038/labinvest.3700464. [DOI] [PubMed] [Google Scholar]
  • 15.Kloppel G, Perren A, Heitz PU. The gastroenteropancreatic neuroendocrine cell system and its tumors: the WHO classification. Ann N Y Acad Sci. 2004;1014:13–27. doi: 10.1196/annals.1294.002. [DOI] [PubMed] [Google Scholar]
  • 16.Modlin IM, Lye KD, Kidd M. Carcinoid tumors of the stomach. Surg Oncol. 2003;12:153–172. doi: 10.1016/s0960-7404(03)00034-3. [DOI] [PubMed] [Google Scholar]
  • 17.Rindi G, Leiter AB, Kopin AS, Bordi C, Solcia E. The "normal" endocrine cell of the gut: changing concepts and new evidences. Ann N Y Acad Sci. 2004;1014:1–12. doi: 10.1196/annals.1294.001. [DOI] [PubMed] [Google Scholar]
  • 18.Modlin IM, Oberg K, Chung DC, Jensen RT, de Herder WW, Thakker RV, et al. Gastroenteropancreatic neuroendocrine tumors. Lancet Oncol. 2008;9:61–72. doi: 10.1016/S1470-2045(07)70410-2. [DOI] [PubMed] [Google Scholar]
  • 19.Maggard MA, O'Connell JB, Ko CY. Updated population-based review of carcinoid tumors. Ann Surg. 2004;240:117–122. doi: 10.1097/01.sla.0000129342.67174.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Arnold CN, Sosnowski A, Blum HE. Analysis of molecular pathways in neuroendocrine cancers of the gastroenteropancreatic system. Ann N Y Acad Sci. 2004;1014:218–219. doi: 10.1196/annals.1294.023. [DOI] [PubMed] [Google Scholar]
  • 21.Perren A, Komminoth P, Heitz PU. Molecular genetics of gastroenteropancreatic endocrine tumors. Ann N Y Acad Sci. 2004;1014:199–208. doi: 10.1196/annals.1294.021. [DOI] [PubMed] [Google Scholar]
  • 22.Tonnies H, Toliat MR, Ramel C, Pape UF, Neitzel H, Berger W, et al. Analysis of sporadic neuroendocrine tumors of the enteropancreatic system by comparative genomic hybridization. Gut. 2001;48:536–541. doi: 10.1136/gut.48.4.536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.D'adda T, Pizzi S, Azzoni C, Bottarelli L, Crafa P, Pasquali C, et al. Different patterns of 11q allelic losses in digestive endocrine tumors. Hum Pathol. 2002;33:322–329. doi: 10.1053/hupa.2002.32219. [DOI] [PubMed] [Google Scholar]
  • 24.Kidd M, Modlin IM, Pfragner R, Eick GN, Champaneria MC, Chan AK, et al. Small bowel carcinoid (enterochromaffin cell) neoplasia exhibits transforming growth factor-betal-mediated regulatory abnormalities including up-regulation of C-Myc and MTA1. Cancer. 2007;109:2420–2431. doi: 10.1002/cncr.22725. [DOI] [PubMed] [Google Scholar]
  • 25.Kytola S, Hoog A, Nord B, Cedermark B, Frisk T, Larsson C, et al. Comparative genomic hybridization identifies loss of 18q22-qter as an early and specific event in tumorigenesis of midgut carcinoids. Am J Pathol. 2001;158:1803–1808. doi: 10.1016/S0002-9440(10)64136-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Das A, Neugut AI, Cooper GS, Chak A. Association of ampullary and colorectal malignancies. Cancer. 2004;100:524–530. doi: 10.1002/cncr.11943. [DOI] [PubMed] [Google Scholar]
  • 27.Stolte M, Pscherer C. Adenoma-carcinoma sequence in the papilla of Vater. Scand J Gastroenterol. 1996;31:376–382. doi: 10.3109/00365529609006414. [DOI] [PubMed] [Google Scholar]
  • 28.Severson RK, Schenk M, Gurney JG, Weiss LK, Demers RY. Increasing incidence of adenocarcinomas and carcinoid tumors of the small intestine in adults. Cancer Epidemiol Biomarkers Prev. 1996;5:81–84. [PubMed] [Google Scholar]
  • 29.Levine JS, Ahnen DJ. Clinical practice. Adenomatous polyps of the colon. N Engl J Med. 2006;355:2551–2557. doi: 10.1056/NEJMcp063038. [DOI] [PubMed] [Google Scholar]
  • 30.Sellner F. Investigations on the significance of the adenoma-carcinoma sequence in the small bowel. Cancer. 1990;66:702–715. doi: 10.1002/1097-0142(19900815)66:4<702::aid-cncr2820660419>3.0.co;2-z. [DOI] [PubMed] [Google Scholar]
  • 31.Campbell WJ, Spence RA, Parks TG. Familial adenomatous polyposis. Br J Surg. 1994;81:1722–1733. doi: 10.1002/bjs.1800811207. [DOI] [PubMed] [Google Scholar]
  • 32.Offerhaus GJ, Giardiello FM, Krush AJ, Booker SV, Tersmette AC, Kelley NC, et al. The risk of upper gastrointestinal cancer in familial adenomatous polyposis. Gastroenterology. 1992;102:1980–1982. doi: 10.1016/0016-5085(92)90322-p. [DOI] [PubMed] [Google Scholar]
  • 33.Jass JR. Gastrointestinal polyposes: clinical, pathological and molecular features. Gastroenterol Clin North Am. 2007;36:927–46. doi: 10.1016/j.gtc.2007.08.009. [DOI] [PubMed] [Google Scholar]
  • 34.Arber N, Hibshoosh H, Yasui W, Neuget AI, Hibshoosh A, Yao Y, et al. Abnormalities in the expression of cell cycle-related proteins in tumors of the small bowel. Cancer Epidemiol Biomarkers Prev. 1999;8:1101–1105. [PubMed] [Google Scholar]
  • 35.Murata M, Iwao K, Miyoshi Y, Nagasawa Y, Yabu M, Himeno S, et al. Activation of the beta-catenin gene by interstitial deletions involving exon 3 as an early event in colorectal tumorigenesis. Cancer Lett. 2000;159:73–78. doi: 10.1016/s0304-3835(00)00533-4. [DOI] [PubMed] [Google Scholar]
  • 36.Rashid A, Hamilton SR. Genetic alterations in sporadic and Cohn's associated adenocarcinomas of the small intestine. Gastroenterology. 1997;113:127–135. doi: 10.1016/s0016-5085(97)70087-8. [DOI] [PubMed] [Google Scholar]
  • 37.Younes N, Fulton N, Tanaka R, Wayne J, Straus FH, Kaplan EL. The presence of K-12 ras mutations in duodenal adenocarcinomas and the absence of ras mutations in other small bowel adenocarcinomas and carcinoid tumors. Cancer. 1997;79:1804–1808. [PubMed] [Google Scholar]
  • 38.Zhao B, Kimura W, Futakawa N, Muto T, Kubota K, Harihara Y, et al. p53 and p21/Waf1 protein expression and K-ras codon 12 mutation in carcinoma of the papilla of Vater. Am J Gastroenterol. 1999;94:2128–2134. doi: 10.1111/j.1572-0241.1999.01309.x. [DOI] [PubMed] [Google Scholar]
  • 39.Fearnhead NS, Britton MP, Bodmer WF. The ABC of APC. Hum Mol Genet. 2001;10:721–733. doi: 10.1093/hmg/10.7.721. [DOI] [PubMed] [Google Scholar]
  • 40.Wheeler JM, Warren BF, Mortensen NJ, Kim HC, Biddolph SC, Elia G, et al. An insight into the genetic pathway of adenocarcinoma of the small intestine. Gut. 2002;50:218–223. doi: 10.1136/gut.50.2.218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Bläker H, von Herbay A, Penzel R, Gross S, Otto HF. Genetics of adenocarcinomas of the small intestine: frequent deletions at chromosome 18q and mutations of the SMAD4 gene. Oncogene. 2002;21:158–164. doi: 10.1038/sj.onc.1205041. [DOI] [PubMed] [Google Scholar]
  • 42.Hamilton SR, Arltonen KA. World Health Organization Classification of tumors: pathology and genetics of tumors of the digestive system. IARC Press; Lyon: 2000. pp. 69–91. [Google Scholar]
  • 43.Lynch HT, Smyrk TC, Lynch PM. Adenocarcinoma of the small bowel in Lynch syndrome II. Cancer. 1989;64:2178–2183. doi: 10.1002/1097-0142(19891115)64:10<2178::aid-cncr2820641033>3.0.co;2-0. [DOI] [PubMed] [Google Scholar]
  • 44.Mellemkjaer L, Johansen C, Gridley G, Linet MS, Kjær SK, Olsen J. Cohn's disease and cancer risk (Denmark) Cancer Causes Control. 2000;11:145–150. doi: 10.1023/a:1008988215904. [DOI] [PubMed] [Google Scholar]
  • 45.Rodriguez-Bigas MA, Vasen HF, Lynch HT, Watson P, Myrhoj T, Jarvinen HJ, et al. Characteristics of small bowel carcinoma in hereditary nonpolyposis colorectal carcinoma. International Collaborative Group on HNPCC. Cancer. 1998;83:240–244. doi: 10.1002/(sici)1097-0142(19980715)83:2<240::aid-cncr6>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
  • 46.Schulmann K, Brasch FE, Kunstmann E, Engel C, Pagenstecher C, Vogelsang H, et al. HNPCC-associated small bowel cancer: clinical and molecular characteristics. Gastroenterology. 2005;128:590–599. doi: 10.1053/j.gastro.2004.12.051. [DOI] [PubMed] [Google Scholar]
  • 47.Gruber SB, Entius MM, Petersen GM, Laken SJ, Longo PA, Boyer R, et al. Pathogenesis of adenocarcinoma in Peutz-Jeghers syndrome. Cancer Res. 1998;58:5267–5270. [PubMed] [Google Scholar]
  • 48.Jenne DE, Reimann H, Nezu J, Friedel W, Loff S, Jeschke R, et al. Peutz-Jeghers syndrome is caused by mutations in a novel serine threonine kinase. Nat Genet. 1998;18:38–43. doi: 10.1038/ng0198-38. [DOI] [PubMed] [Google Scholar]
  • 49.Sato N, Rosty C, Jansen M, Fukushima N, Ueki T, Yeo CJ, et al. STK11/LKB1 Peutz-Jeghers gene inactivation in intraductal papillary-mucinous neoplasms of the pancreas. Am J Pathol. 2001;159:2017–2022. doi: 10.1016/S0002-9440(10)63053-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Boardman LA, Thibodeau SN, Shaid DJ, Lindor NM, McDonnell SK, Burgart LJ, et al. Increased risk for cancer in patients with the Peutz-Jeghers syndrome. Ann Intern Med. 1998;128:896–899. doi: 10.7326/0003-4819-128-11-199806010-00004. [DOI] [PubMed] [Google Scholar]
  • 51.Giardello FM, Brensinger JD, Tersmette AC, Goodman SN, Petersen GM, Booker SV, et al. Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology. 2000;119:1447–1453. doi: 10.1053/gast.2000.20228. [DOI] [PubMed] [Google Scholar]
  • 52.Konishi F, Wyse NE, Muto T. Peutz-Jeghers polyposis associated with carcinoma of the digestive organs. Dis Colon Rectum. 1989;30:790–799. doi: 10.1007/BF02554629. [DOI] [PubMed] [Google Scholar]
  • 53.Carbone A. Emerging pathways in the development of AIDS-related lymphomas. Lancet Oncol. 2003;4:22–29. doi: 10.1016/s1470-2045(03)00957-4. [DOI] [PubMed] [Google Scholar]
  • 54.Kuppers R. Mechanisms of B-cell lymphoma pathogenesis. Nat Rev Cancer. 2005;5:251–262. doi: 10.1038/nrc1589. [DOI] [PubMed] [Google Scholar]
  • 55.Landgren O, Engels EA, Pfeiffer RM, Gridley G, Mellemkjaer L, Olsen JH, et al. Autoimmunity and susceptibility to Hodgkin lymphoma: a population-based case-control study in Scandinavia. J Natl Cancer Inst. 2006;98:1321–1330. doi: 10.1093/jnci/djj361. [DOI] [PubMed] [Google Scholar]
  • 56.Amieva MR, El-Omar EM. Host-bacterial interactions in Helicobacter pylori infection. Gastroenterology. 2008;134:306–323. doi: 10.1053/j.gastro.2007.11.009. [DOI] [PubMed] [Google Scholar]
  • 57.Moslehi R, Devesa SS, Schairer C, Fraumeni JF., Jr Rapidly increasing incidence of ocular non-Hodgkin lymphoma. J Natl Cancer Inst. 2006;98:936–939. doi: 10.1093/jnci/djj248. [DOI] [PubMed] [Google Scholar]
  • 58.Lecuit M, Abachin E, Martin A, Poyart C, Pochart P, Suarez F, et al. Immunoproliferative small intestinal disease associated with Campylobacter jejuni. N Engl J Med. 2004;350:239–248. doi: 10.1056/NEJMoa031887. [DOI] [PubMed] [Google Scholar]
  • 59.Zuckerman E, Zuckerman T, Levine AM, Douer D, Gutekunst K, Mizokami M, et al. Hepatitis C virus infection in patients with B-cell non-Hodgkin lymphoma. Ann Intern Med. 1997;127:423–428. doi: 10.7326/0003-4819-127-6-199709150-00002. [DOI] [PubMed] [Google Scholar]
  • 60.Fine KD, StoneA-heavy chain disease MJ. Mediterranean lymphoma, and immunoproliferative small intestinal disease: a review of clinicopathological features, pathogenesis and differential diagnosis. Am J Gastroenterol. 1999;94:1139–1152. doi: 10.1111/j.1572-0241.1999.01057.x. [DOI] [PubMed] [Google Scholar]
  • 61.Martin IG, Aldoori MI. Immunoproliferative small intestinal disease: Mediterranean lymphoma and alpha heavy chain disease. Br J Surg. 1994;81:20–24. doi: 10.1002/bjs.1800810107. [DOI] [PubMed] [Google Scholar]
  • 62.Anderson LA, McMillan SA, Watson RG, Monaghan P, Gavin AT, Fox C, et al. Malignancy and mortality in a population-based cohort of patients with coeliac disease or "gluten sensitivity. World J Gastroenterol. 2007;13:146–151. doi: 10.3748/wjg.v13.i1.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Catassi C, Fabiani E, Corrao G, Barbato M, De Renzo A, Carella AM, et al. Risk of non-Hodgkin lymphoma in celiac disease. JAMA. 2002;287:1413–1419. doi: 10.1001/jama.287.11.1413. [DOI] [PubMed] [Google Scholar]
  • 64.Green PHR, Fleischauer AT, Bhagat G, Goyal R, Jabri B, Neuget AI. Risk of malignancy in patients with celiac disease. Am J Med. 2003:191–195. doi: 10.1016/s0002-9343(03)00302-4. [DOI] [PubMed] [Google Scholar]
  • 65.Peters U, Askling J, Gridley G, Ekbom A, Linet M. Causes of death in patients with celiac disease in a population-based Swedish cohort. Arch Intern Med. 2003;163:1566–1572. doi: 10.1001/archinte.163.13.1566. [DOI] [PubMed] [Google Scholar]
  • 66.Dube C, Rostom A, Sy R, Cranney A, Saloojee N, Garritty C, et al. The prevalence of celiac disease in average-risk and at-risk Western European populations: a systematic review. Gastroenterology. 2005;128(4 Suppl. 1):S57–67. doi: 10.1053/j.gastro.2005.02.014. [DOI] [PubMed] [Google Scholar]
  • 67.Rewers M. Epidemiology of celiac disease: what are the prevalence, incidence, and progression of celiac disease? Gastroenterology. 2005;128(4 Suppl. 1):S47–51. doi: 10.1053/j.gastro.2005.02.030. [DOI] [PubMed] [Google Scholar]
  • 68.Mowat AM. Coeliac disease—a meeting point for genetics, immunology, and protein chemistry. Lancet. 2003;361:1290–1292. doi: 10.1016/s0140-6736(03)12989-3. [DOI] [PubMed] [Google Scholar]
  • 69.Wolters VM, Wijmenga C. Genetic background of celiac disease and its clinical implications. Am J Gastroenterol. 2008;103:190–195. doi: 10.1111/j.1572-0241.2007.01471.x. [DOI] [PubMed] [Google Scholar]
  • 70.Green PH, Alaedini A, Sander HW, Brannagan TH III, Latov N, Chin RL. Mechanisms underlying celiac disease and its neurologic manifestations. Cell Mol Life Sci. 2005;62:791–799. doi: 10.1007/s00018-004-4109-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Green PHR, Jabri B. Coeliac disease. Lancet. 2003;362:383–391. doi: 10.1016/S0140-6736(03)14027-5. [DOI] [PubMed] [Google Scholar]
  • 72.Askling J, Linet M, Gridley G, Halstensen TS, Ekstrom K, Ekbom A. Cancer incidence in a population-based cohort of individuals hospitalized with celiac disease or dermatitis herpetiformis. Gastroenterology. 2002;123:1428–1435. doi: 10.1053/gast.2002.36585. [DOI] [PubMed] [Google Scholar]
  • 73.Green PH, Jabri B. Celiac disease. Annu Rev Med. 2006;57:207–221. doi: 10.1146/annurev.med.57.051804.122404. [DOI] [PubMed] [Google Scholar]
  • 74.Murray J A. The widening spectrum of celiac disease. Am J Clin Nutr. 1999;69:354–365. doi: 10.1093/ajcn/69.3.354. [DOI] [PubMed] [Google Scholar]
  • 75.Tran T, Davila JA, El-Serag HB. The epidemiology of malignant gastrointestinal stromal tumors: an analysis of 1,458 cases from 1992 to 2000. Am J Gastroenterol. 2005;100:162–168. doi: 10.1111/j.1572-0241.2005.40709.x. [DOI] [PubMed] [Google Scholar]
  • 76.Katz SC, DeMatteo RP. Gastrointestinal stromal tumors and leiomyosarcomas. J Surg Oncol. 2008;97:350–359. doi: 10.1002/jso.20970. [DOI] [PubMed] [Google Scholar]
  • 77.Rossi CR, Mocellin S, Mencarelli R, Foletto M, Pilati P, Nitti D, et al. Gastrointestinal stromal tumors: from a surgical to a molecular approach. Int J Cancer. 2003;107:171–176. doi: 10.1002/ijc.11374. [DOI] [PubMed] [Google Scholar]
  • 78.Corless CL, Heinrich MC. Molecular pathobiology of gastrointestinal stromal sarcomas. Annu Rev Pathol. 2008;3:557–586. doi: 10.1146/annurev.pathmechdis.3.121806.151538. [DOI] [PubMed] [Google Scholar]
  • 79.Hoeben A, Schöffski P, Debiec-Rychter M. Clinical implications of mutational analysis in gastrointestinal stromal tumors. Br J Cancer. 2008;98:684–688. doi: 10.1038/sj.bjc.6604217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Maertens O, Prenen H, Debiec-Rychter M, Wozniak A, Sciot R, Pauwels P, et al. Molecular pathogenesis of multiple gastrointestinal stromal tumors in NF1 patients. Hum Mol Genet. 2006;15:1015–1023. doi: 10.1093/hmg/ddl016. [DOI] [PubMed] [Google Scholar]
  • 81.Botteri E, Iodice S, Raimondi S, Maisonneuve P, Lowenfels AB. Cigarette smoking and adenomatous polyps: a meta-analysis. Gastroenterology. 2008;134:388–395. doi: 10.1053/j.gastro.2007.11.007. [DOI] [PubMed] [Google Scholar]
  • 82.Terry MB, Neugut AI. Cigarette smoking and the colorectal adenomacarcinoma sequence: a hypothesis to explain the paradox. Am J Epidemiol. 1998;147:903–910. doi: 10.1093/oxfordjournals.aje.a009379. [DOI] [PubMed] [Google Scholar]
  • 83.Kune GA, Vitetta L. Alcohol consumption and the etiology of colorectal cancer: a review of the scientific evidence from 1957 to 1991. Nutr Cancer. 1992;18:97–111. doi: 10.1080/01635589209514210. [DOI] [PubMed] [Google Scholar]
  • 84.Moghaddam AA, Woodward M, Huxley R. Obesity and risk of colorectal cancer: a meta-analysis of 31 studies with 70,000 events. Cancer Epidemiol Biomarkers Prev. 2007;16:2533–2547. doi: 10.1158/1055-9965.EPI-07-0708. [DOI] [PubMed] [Google Scholar]
  • 85.Pischon T, Lahmann PH, Boeing H, Friedenreich C, Norat T, Tjonneland A, et al. Body size and risk of colon and rectal cancer in the European Prospective Investigation Into Cancer and Nutrition (EPIC) J Natl Cancer Inst. 2006;98:920–931. doi: 10.1093/jnci/djj246. [DOI] [PubMed] [Google Scholar]
  • 86.Ma J, Stampfer MJ, Giovannucci E, Artigas C, Hunter DJ, Fuchs C, et al. Methylenetetrahydrofolate reductase polymorphism, dietary interactions, and risk of colorectal cancer. Cancer Res. 1997;57:1098–1102. [PubMed] [Google Scholar]
  • 87.Michels KB, Edward G, Joshipura KJ, Rosner BA, Stampfer MJ, Fuchs CS, et al. Prospective study of fruit and vegetable consumption and incidence of colon and rectal cancers. J Natl Cancer Inst. 2000;92:1740–1752. doi: 10.1093/jnci/92.21.1740. [DOI] [PubMed] [Google Scholar]
  • 88.Trock B, Lanza E, Greenwald P. Dietary fiber, vegetables, and colon cancer: critical review and meta-analyses of the epidemiologic evidence. J Natl Cancer Inst. 1990;82:650–661. doi: 10.1093/jnci/82.8.650. [DOI] [PubMed] [Google Scholar]
  • 89.Neugut AI, Jacobson JS, Suh S, Mukherjee R, Arber N. The epidemiology of cancer of the small bowel. Cancer Epidemiol Biomarkers Prev. 1998;7:243–251. [PubMed] [Google Scholar]
  • 90.Chow W- H, Linet MS, McLaughlin JK. Risk factors for small intestine cancer. Cancer Causes Control. 1993;4:163–169. doi: 10.1007/BF00053158. [DOI] [PubMed] [Google Scholar]
  • 91.Kaerlev L, Teglbjaerg PS, Sabroe S, Kolstad HA, Ahrens W, Eriksson M, et al. The importance of smoking and medical history for development of small bowel carcinoid tumor: a European population-based case-control study. Cancer Causes Control. 2002;13:27–34. doi: 10.1023/a:1013922226614. [DOI] [PubMed] [Google Scholar]
  • 92.Kaerlev L, Teglbjaerg PS, Sabroe S, Kolstad HA, Ahrens W, Eriksson M, et al. Is there an association between alcohol intake or smoking and small bowel adenocarcinoma? Results from a European multi-center case-control study. Cancer Causes Control. 2000;11:791–797. doi: 10.1023/a:1008920502888. [DOI] [PubMed] [Google Scholar]
  • 93.Wu AH, Yu MC, Mack TM. Smoking, alcohol use, dietary factors and risk of small intestinal adenocarcinoma. Int J Cancer. 1997;70:512–517. doi: 10.1002/(sici)1097-0215(19970304)70:5<512::aid-ijc4>3.0.co;2-0. [DOI] [PubMed] [Google Scholar]
  • 94.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. doi: 10.1002/ijc.23529. [DOI] [PubMed] [Google Scholar]
  • 95.Boffetta P, Hashibe M. Alcohol and cancer. Lancet Oncol. 2006;7:149–156. doi: 10.1016/S1470-2045(06)70577-0. [DOI] [PubMed] [Google Scholar]
  • 96.Seitz HK, Stickel F. Molecular mechanisms of alcohol-mediated carcinogenesis. Nat Rev Cancer. 2007;7:599–612. doi: 10.1038/nrc2191. [DOI] [PubMed] [Google Scholar]
  • 97.Negri E, Bosetti C, La Vecchia C, Fioretti F, Conti E, Franceschi S. Risk factors for adenocarcinoma of the small intestine. Int J Cancer. 1999;82:171–174. doi: 10.1002/(sici)1097-0215(19990719)82:2<171::aid-ijc3>3.0.co;2-t. [DOI] [PubMed] [Google Scholar]
  • 98.Samanic C, Gridley G, Chow WH, Lubin J, Hoover RN, Fraumeni JF., Jr Obesity and cancer risk among white and black United States veterans. Cancer Causes Control. 2004;15:35–43. doi: 10.1023/B:CACO.0000016573.79453.ba. [DOI] [PubMed] [Google Scholar]
  • 99.Wolk A, Gridley G, Svensson M, Nyren O, McLaughlin JK, Fraumeni JF, Jr, et al. A prospective study of obesity and cancer risk (Sweden) Cancer Causes Control. 2001;12:13–21. doi: 10.1023/a:1008995217664. [DOI] [PubMed] [Google Scholar]
  • 100.Bjorge T, Tretli S, Engeland A. Height and body mass index in relation to cancer of the small intestine in two million Norwegian men and women. Br J Cancer. 2005;93:807–810. doi: 10.1038/sj.bjc.6602789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Shanahan F. Cohn's disease. Lancet. 2002;359:62–69. doi: 10.1016/S0140-6736(02)07284-7. [DOI] [PubMed] [Google Scholar]
  • 102.Braun J, Wei B. Body traffic: ecology, genetics, and immunity in inflammatory bowel disease. Annu Rev Pathol. 2007;2:401–429. doi: 10.1146/annurev.pathol.1.110304.100128. [DOI] [PubMed] [Google Scholar]
  • 103.Cantor MJ, Nickerson P, Bernstein CN. The role of cytokine gene polymorphisms in determining disease susceptibility and phenotype in inflammatory bowel disease. Am J Gastroenterol. 2005;100:1134–1142. doi: 10.1111/j.1572-0241.2005.40979.x. [DOI] [PubMed] [Google Scholar]
  • 104.Michelassi F, Testa G, Pomidor WJ, Lashner BA, Block GE. Adenocarcinoma complicating Crohn's disease. Dis Colon Rectum. 1993;36:654–661. doi: 10.1007/BF02238592. [DOI] [PubMed] [Google Scholar]
  • 105.Ribeiro MB, Greenstein AJ, Heimann TM, Yamazaki Y, Aufses AH., Jr. Adenocarcinoma of the small intestine in Crohn's disease. Surg Gynecol Obstet. 1991;173:343–349. [PubMed] [Google Scholar]
  • 106.Jess T, Loftus EV, Jr, Velayos FS, Harmsen WS, Zinsmeister AR, Smyrk TC, et al. Risk of intestinal cancer in inflammatory bowel disease: a population-based study from Olmsted County, Minnesota. Gastroenterology. 2006;130:1039–1046. doi: 10.1053/j.gastro.2005.12.037. [DOI] [PubMed] [Google Scholar]
  • 107.Friedman S. Cancer in Crohn's disease. Gastroenterol Clin North Am. 2006;35:621–639. doi: 10.1016/j.gtc.2006.07.008. [DOI] [PubMed] [Google Scholar]
  • 108.Jess T, Gamborg M, Matzen P, Munkholm P, Sorensen TI. Increased risk of intestinal cancer in Crohn's disease: a meta-analysis of population-based cohort studies. Am J Gastroenterol. 2005;100:2724–2729. doi: 10.1111/j.1572-0241.2005.00287.x. [DOI] [PubMed] [Google Scholar]
  • 109.Eaden JA, Abrams KR, Mayberry JF. The risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut. 2001;48:526–535. doi: 10.1136/gut.48.4.526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Isolauri E, Salminen S. Probiotics, gut inflammation and barrier function. Gastroenterol Clin North Am. 2005;34:437–50. doi: 10.1016/j.gtc.2005.05.010. viii. [DOI] [PubMed] [Google Scholar]
  • 111.Wehkamp J, Stange EF. A new look at Crohn's disease: breakdown of the mucosal antibacterial defense. Ann N Y Acad Sci. 2006;1072:321–331. doi: 10.1196/annals.1326.030. [DOI] [PubMed] [Google Scholar]
  • 112.Lievin-Le MV, Servin AL. The front line of enteric host defense against unwelcome intrusion of harmful microorganisms: mucins, antimicrobial peptides, and microbiota. Clin Microbiol Rev. 2006;19:315–337. doi: 10.1128/CMR.19.2.315-337.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Sartor RB. Microbial influences in inflammatory bowel diseases. Gastroenterology. 2008;134:577–594. doi: 10.1053/j.gastro.2007.11.059. [DOI] [PubMed] [Google Scholar]
  • 114.Swidsinski A, Ladhoff A, Pernthaler A, Swidsinski S, Loening-Baucke V, Ortner M, et al. Mucosal flora in inflammatory bowel disease. Gastroenterology. 2002;122:44–54. doi: 10.1053/gast.2002.30294. [DOI] [PubMed] [Google Scholar]
  • 115.Videla S, Vilaseca J, Guarner F, Salas A, Treserra F, Crespo E, et al. Role of intestinal microflora in chronic inflammation and ulceration of the rat colon. Gut. 1994;35:1090–1097. doi: 10.1136/gut.35.8.1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Bamias G, Nyce MR, De La Rue SA, Cominelli F. New concepts in the pathophysiology of inflammatory bowel disease. Ann Intern Med. 2005;143(12):895–904. doi: 10.7326/0003-4819-143-12-200512200-00007. [DOI] [PubMed] [Google Scholar]
  • 117.Braat H, Peppelenbosch MP, Hommes DW. Immunology of Crohn's disease. Ann NY Acad Sci. 2006;1072:135–154. doi: 10.1196/annals.1326.039. [DOI] [PubMed] [Google Scholar]
  • 118.Schreiber S, Rosenstiel P, Albrecht M, Hampe J, Krawczak M. Genetics of Crohn disease, an archetypal inflammatory barrier disease. Nat Rev Genet. 2005;6:376–388. doi: 10.1038/nrg1607. [DOI] [PubMed] [Google Scholar]
  • 119.Gaya DR, Russell RK, Nimmo ER, Satsangi J. New genes in inflammatory bowel disease: lessons for complex diseases? Lancet. 2006;367:1271–1284. doi: 10.1016/S0140-6736(06)68345-1. [DOI] [PubMed] [Google Scholar]
  • 120.Hugot JP. CARD15/NOD2 mutations in Crohn's disease. AnnN Y Acad Sci. 2006;411:9–18. doi: 10.1196/annals.1326.011. [DOI] [PubMed] [Google Scholar]
  • 121.Ahmad T, Armuzzi A, Bunce M, Mulcahy-Hawes K, Marshall SE, et al. The molecular classification of the clinical manifestations of Crohn's disease. Gastroenterology. 2002;122:854–866. doi: 10.1053/gast.2002.32413. [DOI] [PubMed] [Google Scholar]
  • 122.Hugot JP, Chamaillard M, Zouali H. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn's disease. Nature. 2001;411:599–603. doi: 10.1038/35079107. [DOI] [PubMed] [Google Scholar]
  • 123.Cho JH, Abraham C. Inflammatory bowel disease genetics: Nod2. Annu Rev Med. 2007;58:401–416. doi: 10.1146/annurev.med.58.061705.145024. [DOI] [PubMed] [Google Scholar]
  • 124.Lesage S, Zouali H, Cezard JP. CARD15/NOD2 mutational analysis and genotype-phenotype correlation in 612 patients with inflammatory bowel disease. Am J Hum Genet. 2002;70:845–857. doi: 10.1086/339432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Schottenfeld D, Beebe-Dimmer J. Chronic inflammation: a common and important factor in the pathogenesis of neoplasia. CA Cancer J Clin. 2006;56:69–83. doi: 10.3322/canjclin.56.2.69. [DOI] [PubMed] [Google Scholar]
  • 126.Smedby KE, Baecklund E, Askling J. Malignant lymphomas in autoimmunity and inflammation: a review of risks, risk factors, and lymphoma characteristics. Cancer Epidemiol Biomarkers Prev. 2006;15:2069–2077. doi: 10.1158/1055-9965.EPI-06-0300. [DOI] [PubMed] [Google Scholar]
  • 127.O'Shea JJ, Ma A, Lipsky P. Cytokines and autoimmunity. Nat Rev Immunol. 2002;2:37–45. doi: 10.1038/nri702. [DOI] [PubMed] [Google Scholar]
  • 128.Federico A, Morgillo F, Tuccillo C, Ciardiello F, Loguercio C. Chronic inflammation and oxidative stress in human carcinogenesis. Int J Cancer. 2007;121:2381–2386. doi: 10.1002/ijc.23192. [DOI] [PubMed] [Google Scholar]
  • 129.Guyton AC, Hall JE. Textbook of Medical Physiology. 11th Elsevier-Saunders; Philadelphia: 2006. pp. 771–817. [Google Scholar]
  • 130.Mazzone A, Farrugia G. Evolving concepts in the cellular control of gastrointestinal motility: neurogastroenterology and enteric sciences. Gastroenterol Clin North Am. 2007;36:499–513. doi: 10.1016/j.gtc.2007.07.003. vii. [DOI] [PubMed] [Google Scholar]
  • 131.Guarner F, Malagelada JR. Gut flora in health and disease. Lancet. 2003;361:512–519. doi: 10.1016/S0140-6736(03)12489-0. [DOI] [PubMed] [Google Scholar]
  • 132.Lewis SJ, Heaton KW. The metabolic consequences of slow colonic transit. Am J Gastroenterol. 1999;94:2010–2016. doi: 10.1111/j.1572-0241.1999.01271.x. [DOI] [PubMed] [Google Scholar]
  • 133.Mai V, Morris JG., Jr. Colonic bacterial flora: changing understandings in the molecular age. J Nutr. 2004;134:459–464. doi: 10.1093/jn/134.2.459. [DOI] [PubMed] [Google Scholar]
  • 134.Ouwehand AC, Vaughan EE. Gastrointestinal microbiology. Taylor and Francis; New York: 2006. [Google Scholar]
  • 135.Greenstein AJ, Sachar D, Pucillo A, Kreel I, Geller S, Janowitz HD, et al. Cancer in Crohn's disease after diversionary surgery. A report of seven carcinomas occurring in excluded bowel. Am J Surg. 1978;135:86–90. doi: 10.1016/0002-9610(78)90015-6. [DOI] [PubMed] [Google Scholar]
  • 136.Bourlioux P, Koletzko B, Guarner F, Braesco V. The Intelligent Intestine. The intestine and its microflora are partners for the protection of the host: report on the Danone Symposium; Paris. June 14; 2002. Am J Clin Nutr. 2003;78:675-683. [DOI] [PubMed] [Google Scholar]
  • 137.Hooper LV, Gordon JI. Glycans as legislators of host-microbial interactions: spanning the spectrum from symbiosis to pathogenicity. Glycobiology. 2001;11:1R–10R. doi: 10.1093/glycob/11.2.1r. [DOI] [PubMed] [Google Scholar]
  • 138.MacDonald TT, Gordon JN. Bacterial regulation of intestinal immune responses. Gastroenterol Clin North Am. 2005;34:401. doi: 10.1016/j.gtc.2005.05.012. viii. [DOI] [PubMed] [Google Scholar]
  • 139.Laquer GL. Carcinogenic effects of cycad meal and cycasin, methylazoxymethanol glycoside, in rats and effects of cycasin in germfree rats. Fed Proc. 1964;23:1386–1387. [PubMed] [Google Scholar]
  • 140.Kim DH, Jin YH. Intestinal bacterial beta-glucuronidase activity of patients with colon cancer. Arch Pharm Res. 2001;24:564–567. doi: 10.1007/BF02975166. [DOI] [PubMed] [Google Scholar]
  • 141.Reddy BS, Ohmori T. Effect of intestinal microflora and dietary fat on 3,2'-dimethyl-4-aminobiphenyl-induced colon carcinogenesis in F344 rats. Cancer Res. 1981;41:1363–1367. [PubMed] [Google Scholar]
  • 142.Reddy BS, Weisburger JH, Narisawa T, Wynder EL. Colon carcinogenesis in germ-free rats with 1,2-dimethylhydrazine and N-methyl-n'-nitro-N-nitrosoguanidine. Cancer Res. 1974;34:2368–2372. [PubMed] [Google Scholar]
  • 143.Hofmann AF. The enterohepatic circulation of bile acids in man. Clin Gastroenterol. 1977;6:3–24. [PubMed] [Google Scholar]
  • 144.Wexler HM. Bacteroides: the good, the bad, and the nitty-gritty. Clin Microbiol Rev. 2007;20:593–621. doi: 10.1128/CMR.00008-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Hill MJ. Bile flow and colon cancer. Mutat Res. 1990;238:3130–3320. doi: 10.1016/0165-1110(90)90023-5. [DOI] [PubMed] [Google Scholar]
  • 146.Ochsenkuhn T, Bayerdorffer E, Meining A, Schinkel M, Thiede C, Nussler V, et al. Colonic mucosal proliferation is related to serum deoxycholic acid levels. Cancer. 1999;85:1664–1669. [PubMed] [Google Scholar]
  • 147.Ross RK, Hartnett NM, Bernstein L. Epidemiology of adenocarcinomas of the small intestine: Is bile a small bowel carcinogen? Br J Cancer. 1991;63:143–145. doi: 10.1038/bjc.1991.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Imray CH, Radley S, Davis A, Barker G, Hendrickse CW, Donovan IA, et al. Fecal unconjugated bile acids in patients with colorectal cancer or polyps. Gut. 1992;33:1239–1245. doi: 10.1136/gut.33.9.1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Payne CM, Weber C, Crowley-Skillicorn C, Dvorak K, Bernstein H, Bernstein C, et al. Deoxycholate induces mitochondrial oxidative stress and activates NF-kappaB through multiple mechanisms in HCT-116 colon epithelial cells. Carcinogenesis. 2007;28:215–222. doi: 10.1093/carcin/bgl139. [DOI] [PubMed] [Google Scholar]
  • 150.Powolny A, Xu J, Loo G. Deoxycholate induces DNA damage and apoptosis in human colon epithelial cells expressing either mutant or wildtype p53. Int J Biochem Cell Biol. 2001;33:193–203. doi: 10.1016/s1357-2725(00)00080-7. [DOI] [PubMed] [Google Scholar]
  • 151.Lagergren J, Ye W, Ekbom A. Intestinal cancer after cholecystectomy: is bile involved in carcinogenesis? Gastroenterology. 2001;121:542–547. doi: 10.1053/gast.2001.27083. [DOI] [PubMed] [Google Scholar]

RESOURCES