+ All Categories
Home > Documents > Colorectal cancer in inflammatory bowel disease: molecular and clinical features

Colorectal cancer in inflammatory bowel disease: molecular and clinical features

Date post: 30-Dec-2016
Category:
Upload: harris
View: 216 times
Download: 4 times
Share this document with a friend
13
Colorectal cancer in inflammatory bowel disease: molecular and clinical features Sreekant Murthy, PhD * , Anne Flanigan, PhD, Harris Clearfield, MD Division of Gastroenterolgy and Hepatology, MCP Hahnemann University, Mail Stop 444, Suite 2105 NCB, 245 N 15th Street, Philadelphia, PA 19102-1192, USA The two forms of inflammatory bowel disease (IBD), Crohn’s disease and ulcerative colitis (UC), are characterized by chronic and relapsing inflamma- tion of the intestines. Initiating events, though unclear, presumably occur well before patients are symptomatic. Evidence gathered over the past decade from both IBD patients and animal models of intestinal inflammation have confirmed that IBD represents complex heterogenic forms of diseases, influenced by a combination of environmental, genetic, and immunologic factors working in con- cert [1] to produce exaggerated immune responses, resulting in chronic and re- mitting inflammation. There is familial occurrence of IBD and concordance of disease in mono- zygotic twins. There is a large degree of heterogeneity due to the polygenic nature of the diseases. Chromosome 16 has been specifically linked to Crohn’s disease in non-Jewish families (IBD-1). Overall susceptibility to IBD in certain loci of chromosomes 12, 7, and 3 has been clearly established, in decreasing order [2]. Certain HLA regions on chromosome 2 and 6 are linked to UC [2]. Abnormal mucosal barrier function, aberrant penetration, and recognition of intestinal bacterial and dietary byproducts as foreign antigens cause hyperstimu- lation of the mucosal immune system. This dysregulated immune function involves overactivation of TH1 (Crohn’s disease) and TH2 (UC) helper T-cell responses, resulting in the excessive production of proinflammatory cytokines, causing an imbalance with anti-inflammatory cytokines [3]. In the lamina propria, there is excessive infiltration of neutrophils, macrophages, and T lymphocytes, leading to an increased production of arachidonic acid metabolites, oxidative 0889-8588/03/$ – see front matter D 2003, Elsevier Science (USA). All rights reserved. doi:10.1016/S0889-8588(03)00016-9 Reprinted with permission from Gastroenterology Clinics of North America 2002;31(2):551 – 64. * Corresponding author. E-mail address: [email protected] (S. Murthy). Hematol Oncol Clin N Am 17 (2003) 525 – 537
Transcript

Colorectal cancer in inflammatory bowel

disease: molecular and clinical features

Sreekant Murthy, PhD*, Anne Flanigan, PhD,Harris Clearfield, MD

Division of Gastroenterolgy and Hepatology, MCP Hahnemann University, Mail Stop 444,

Suite 2105 NCB, 245 N 15th Street, Philadelphia, PA 19102-1192, USA

The two forms of inflammatory bowel disease (IBD), Crohn’s disease and

ulcerative colitis (UC), are characterized by chronic and relapsing inflamma-

tion of the intestines. Initiating events, though unclear, presumably occur well

before patients are symptomatic. Evidence gathered over the past decade from

both IBD patients and animal models of intestinal inflammation have confirmed

that IBD represents complex heterogenic forms of diseases, influenced by a

combination of environmental, genetic, and immunologic factors working in con-

cert [1] to produce exaggerated immune responses, resulting in chronic and re-

mitting inflammation.

There is familial occurrence of IBD and concordance of disease in mono-

zygotic twins. There is a large degree of heterogeneity due to the polygenic

nature of the diseases. Chromosome 16 has been specifically linked to Crohn’s

disease in non-Jewish families (IBD-1). Overall susceptibility to IBD in certain

loci of chromosomes 12, 7, and 3 has been clearly established, in decreasing

order [2]. Certain HLA regions on chromosome 2 and 6 are linked to UC [2].

Abnormal mucosal barrier function, aberrant penetration, and recognition of

intestinal bacterial and dietary byproducts as foreign antigens cause hyperstimu-

lation of the mucosal immune system. This dysregulated immune function

involves overactivation of TH1 (Crohn’s disease) and TH2 (UC) helper T-cell

responses, resulting in the excessive production of proinflammatory cytokines,

causing an imbalance with anti-inflammatory cytokines [3]. In the lamina propria,

there is excessive infiltration of neutrophils, macrophages, and T lymphocytes,

leading to an increased production of arachidonic acid metabolites, oxidative

0889-8588/03/$ – see front matter D 2003, Elsevier Science (USA). All rights reserved.

doi:10.1016/S0889-8588(03)00016-9

Reprinted with permission from Gastroenterology Clinics of North America 2002;31(2):551–64.

* Corresponding author.

E-mail address: [email protected] (S. Murthy).

Hematol Oncol Clin N Am

17 (2003) 525–537

stress, and additional proinflammatory cytokines such as interleukin 12 and

interferon-g, further amplifying chronic intestinal inflammation [3].

The multistage process of tumorigenesis in UC involving invasion, progres-

sion, and promotion is illustrated in Fig. 1. Fundamentally, more aggressive

behavior of several inflammatory cascades invokes abnormal epithelial prolif-

eration and differentiation responses, thereby increasing the risk of developing

cancer in IBD compared with sporadic cancer. Effective suppression of inflam-

mation reduces the risk of cancer, suggesting the involvement of these cascades

in the malignant transformation of the colonic mucosa [4].

Molecular pathways of dysplasia and cancer in IBD

Colorectal cancer (CRC) is known to be associated with chronic colitis, more

often in UC than in Crohn’s disease, although carcinoma in chronic Crohn’s

colitis is increasingly recognized. Crohn’s-associated small intestinal inflam-

mation has been shown to be associated with a low incidence of small intestinal

cancers [5]. UC patients have a 20 to 30 fold higher risk of developing CRC than

the general population [6]. Epidemiologic studies have shown that the risk of

developing cancer or dysplasia in UC-associated CRC increases with the age of

onset, duration, and extent of disease [7–9]. In UC, CRC develops via dysplasia

[10–12], rather than the classic adenoma–CRC sequence [13,14].

Determining the molecular pathways of UC-associated CRC is hindered by

the complexity of underlying colitis and the presence of reactive regenerative

epithelium, which is difficult to distinguish from true dysplasia, thus the need to

Fig. 1. Multistage inflammation-mediated tumorigenesis in IBD.

S. Murthy et al / Hematol Oncol Clin N Am 17 (2003) 525–537526

defer surveillance biopsies until acute inflammatory mucosal changes have

subsided. Furthermore, multiple inflammatory cascades may participate in the

initiation, induction, and progression of malignancy into metastatic tumors

(Fig. 1). Despite inflammation, not all patients with UC nor all animals with

experimental colitis develop dysplasia or cancer.

IBD-associated cancers do not arise de novo, but do arise depending on

genetic make-up and proceed through a series of genetic alterations. This genetic

instability, in some respects, is dissimilar to sporadic CRC (SCRC) and heritable

CRC (HCRC). They could, however, still be manifested by allelic loss, deletions,

rearrangements, germ-line or somatic mutations of tumor proto-oncogenes or

tumor suppressor genes such as p53, Ki-ras, deleted in colon cancer, adenom-

atous polyposis coli (APC), and polymorphisms in DNA mismatch repair genes

(MSH2) [14]. There could also be altered expression of many key proteins such

as p16, p27, transforming growth factor (TGF)-b1 type II receptor, cadherins,

and catenins. All of these factors play a major role in cellular differentiation

and proliferation.

There has been significant attention focused on how reactive oxygen species

cause cellular damage and neoplasia. Inflammation through free radical pathways

damages DNA and protein. This damage, in the background of increased

proliferation, can fix and propagate mutations of many genes resulting in the

development of dysplasia and invasive cancers.

The temporal sequences of molecular pathways that govern neoplastic

processes in UC-associated CRC do not appear to parallel SCRC. Morpholog-

ically, most SCRCs in Western countries arise through an adenoma–carcinoma

sequence. In UC, however, they tend to develop in association with dysplasia

[15–17] and from either flat mucosa or a dysplasia-associated lesion or mass

(DALM) [11]. Identification of accurate and sensitive molecular and biochemical

markers in these stages of malignant transformation in UC-associated CRC are

just beginning to emerge (Table 1), and the suggested pathways appear to be

different from SCRC.

Nuclear expression of the tumor suppressor gene, p53, has been observed in

dysplasia and cancers of UC patients [18–20]. Immunohistochemical and

molecular studies show that nuclear expression of p53 mutations, and loss of

heterozygosity are early events that may precede dysplasia in UC-associated

CRC [21]. In SCRC, however, mutations and loss of heterozygosity are late

events [21]. Recently, Hussain et al [22] suggested that detection of mutant p53

might facilitate selecting patients for endoscopic surveillance. The detection

assay they recommended provides a sensitive measure to quantitate mutated p53

alleles in predysplastic lesions; however, the applications of this assay for

selecting subjects for endoscopic surveillance needs further evaluation by using

well-designed prospective studies. Hudson et al [23] showed that a macrophage

inhibitory factor, a proinflammatory cytokine, inhibits functional transcriptional

activities of p53, suggesting that exaggerated production of macrophage inhib-

itory factor in chronic inflammatory conditions promotes tumorigenesis. Thus,

this pivotal in vitro study carves a pathway for the distinct association between

S. Murthy et al / Hematol Oncol Clin N Am 17 (2003) 525–537 527

Table 1

Molecular and genetic markers of sporadic and UC associated colon cancer

Molecular or genetic marker and

(chromosome location) Gene/protein function Involvement in SCRC or HCRC Involvement in UC

Adenomatous polyposis coli

(chromosome 5q21)

Tumor suppressor gene involved

in cell adhesion

Early event in SCRC

Mutations are present in 30%

of adenomatous lesions or 75%

of SCRC

Present but rare

Appears in same frequency in

polypoid and flat lesions

Ki-ras (chromosome 12p12) Oncogene involved in cell

cycle regulation

Frequently observed in SCRC Late event

Inconsistent frequency of appearance

hMSH2 (chromosome 2p22) Human mismatch repair gene Early event in SCRC and HCRC

Microsatellite instability

High frequency of microsatellite

instability in dysplasia

Deleted in colon cancer

(chromosome 18q21)

(see Ref. [60])

Tumor suppressor gene involved

in cell adhesion

Involved in formation of polyps

in HCRC

Gene function is lost in 70% to

75% of cancers

Limited data available

May be associated with transition

of dysplasia to cancer

p53 (chromosome 17p13) Tumor suppressor gene involved

in cell cycle regulation

and apoptosis

Late event

Gene function is lost in 70% to

80% of cancers

Early event

Precedes dysplasia

p16 (chromosome 9p21) Tumor suppressor gene involved

in cell cycle regulation

Gene function is lost in most

primary tumors

Frequent and early event

Hypermethylation is observed

Hypermethylation is observed

p27 (chromosome 12p13) Tumor suppressor gene involved

in cell cycle regulation

Eliminated in SCRC

(controversial)

Gene function is lost

May cause aggression of UC

associated CRC

Transforming growth factor Transforming growth Factor-b1type II receptor

Overexpressed metastatic

colon cancer

Commonly observed in all

unstable SCRC

Early appearance in certain neoplasms

in combination with other

microsatellite instabilities

Abbreviations: CRC, colorectal cancer; HCRC, heritable CRC; SCRC, sporadic CRC; UC, ulcerative colitis.

S.Murth

yet

al/Hem

atolOncolClin

NAm

17(2003)525–537

528

inflammation and cancer. This observation is important because massive infiltra-

tion and activation of macrophages is commonly observed in IBD.

In SCRC and HCRC, mutation of the APC gene and impairment of its

function are early events [24]. In UC-associated CRC, however, APC gene

mutations are rare. The loss of heterozygosity of APC in both polypoid and flat

lesions in UC cancer patients appear at the same frequency, but is slightly

increased in DALMs in UC [25].

Investigators have studied mutations of the proto-oncogene, Ki-ras, in both

UC-associated CRC and in SCRC [26]. In sporadic cancer, mutations of Ki-ras are

frequently observed. These mutations appear later in the development of CRC

arising from UC. Unfortunately, there have been inconsistencies in the reporting of

Ki-ras mutation frequencies in UC ranging from as low as 3% to as high as 50%

[27]. Carcinomatous lesions and high-grade dysplasia show a generally higher

frequency of Ki-rasmutations [28]. Some studies have recommended screening of

colonic lavages for Ki-ras mutations for the early detection of UC-associated

colon cancer [29], but the validity of this screening remains to be confirmed.

Germ-line mutations of the human MSH2 (a gene whose protein products help

repair mistakes made in DNA replication) and microsatellite D165541 have been

implicated in both HCRC and UC-associated CRC [30,31]. The microsatellite

marker for IBD-1(D16S541) was found in 33% of UC cancer patients, but only

12% of control HCRC. Thirty-two percent of patients with dysplastic UC also

had this genotype, whereas only 8% of patients with nondysplastic UC had the

genotype [31]. The authors of this study [31] suggested that this microsatellite

marker, when combined with other markers, has the potential to be used as a

screening tool for CRC and dysplasia in patients with UC. This recommendation

needs to be confirmed by additional studies.

The roles played by other factors such as TGF-b II receptor, p16, and p27 are

beginning to emerge. Dysregulation of growth factors and cognate receptors may

play an important role in the formation of neoplasms. Recently, a TGF-b 1 type II

receptor (TGF-b 1RII) mutation (via a microsatellite instability) was described in

UC-associated CRC [32]. This study showed that mutations of this gene occur

early in unstable UC neoplasms but are restricted to certain subsets in which there

are microsatellite instabilities at other chromosomal loci. This form of instability

is unrestricted and commonly observed in all unstable SCRCs.

The protein p16 (also known as CDKN2, INK4a, or MTS1), whose loss has

been implicated in cancer, maps to chromosome 9p21 and is an inhibitor for

cyclin-dependent kinase, CDK4, and CDK6. It binds to the phosphorylated

CDK-cyclin complex, resulting in progression from the G1 phase to the S phase

of the cell cycle. Therefore, p16 can negatively regulate the cell cycle and is a

candidate tumor suppressor gene. Loss of p16 is observed in a majority of tumor

cell lines and in most primary tumors. Evidence has shown that transcriptional

silencing as a consequence of hypermethylation is the predominant mechanism of

p16 gene inactivation in SCRC. More recent studies identified the significance of

p16INK4a methylation in the colonic epithelium of patients with long-standing

UC. These results showed that hypermethylation of the p16INK4a promoter

S. Murthy et al / Hematol Oncol Clin N Am 17 (2003) 525–537 529

region is a frequent and early occurring event in UC-associated CRC progres-

sion [33,34].

The protein p27 is yet another CDK inhibitor that negatively regulates the G1

to S phase of the cell cycle. This protein is known to protect against inflam-

mation-induced injury and helps epithelial cell differentiation. Unlike in sporadic

cancer, p27 is underexpressed in CRC that has been associated with UC [35]. The

low expression of this key protein has been suggested to further contribute to the

aggression of cancer in IBD.

b-catenin, a 92-kDa protein, has a high sequence similarity to the polarity gene

Armadillo in Drosophila [36]. This protein plays a key role in signal transduction

and cell adhesion. Altered expression of b-catenin and its binding partners

E-cadherin and the APC protein are frequent early events in SCRC. It appears

that APC is responsible for regulating its homeostatic control; however, with loss

of APC function, b-catenin accumulates in the cytoplasm and nucleus and activates

Tcf4 and c-myc. b-Catenin is normally expressed in the lateral membranes of the

colonic epithelium. It is overexpressed in both sporadic and UC-associated CRC.

This overexpression is closely linked to E-cadherin alterations in UC-associated

CRC. Abnormal b-catenin expression, however, was more closely linked to APC

alterations in sporadic cancers. In sporadic cancers, b-catenin translocates from the

cell membrane to the nucleus or cytoplasm. On the contrary, Karayiannakis et al

[37] have shown that in human UC and Crohn’s disease, b-catenin is always

localized to the cell membrane. Recently, Mikami et al [38] showed stronger

b-catenin staining in UC-associated neoplasms compared with stronger expression

in the nucleus and cytoplasm of sporadic cancers, suggesting different pathways

of tumorigenesis for UC-associated CRC compared with sporadic cancers.

Taken together, some of these diverse molecular events associated with

UC-associated CRC demonstrate a mechanistic connection between inflamma-

tion and tumorigenesis that is distinct from the pathways observed in both HCRC

and SCRC. These findings highlight opportunities to identify distinct molecular

targets to not only determine potential risks of developing cancer but also for

strategizing therapeutic interventions.

Molecular pathways of dysplasia: lessons from animal models

There are many animal models of IBD but only few of these models are ideal

to study the dysplasia–cancer sequence. Certain cotton top tamarins (Sanguinas

oedipus) develop colitis-associated colon cancer in a captive environment,

approximately 3 to 6 times more than those animals without the familial history

of CRC [39]; however, these cancers arise unassociated with dysplasia, which is

different from human UC patients. Other mouse models such as interleukin-B10

knockout and GaI knockout develop colitis-associated adenocarcinoma, but they

have not been adequately used for cancer studies as they relate to humans.

Many investigators have reported the appearance of dysplasia and cancers in

mice, rats, and hamsters when they are fed dextran sulfate sodium solutions. This

S. Murthy et al / Hematol Oncol Clin N Am 17 (2003) 525–537530

model has provided opportunities to evaluate the spectrum of biochemical and

histopathologic changes that migrate from colitis to dysplasia to cancer. In a

recent study, Cooper et al [40] studied the association of clinical disease severity,

incidence, distribution, multifocality of dysplasia, cancer, and immunoexpression

of p53 and b-catenin in a mouse model of dextran sulfate sodium–induced

colitis. The results of the study showed several similarities with human colitis-

associated colon cancer. Similar to humans, only few animals developed

dysplasia/cancer and there was a lack of association between clinical disease

severity and carcinogenesis.

Inflammation scores were higher in those animals with flat cancerous lesions

and invasive cancers, suggesting an association between inflammation and tu-

morigenesis. In contrast to humans, p53 mutations were uncommon in this model.

An interesting observation was the differential overexpression of b-catenin in twotypes of lesions observed in this model. Immunohistochemical studies showed that

DALMs showed complete translocation of membrane b-catenin to the cytoplasm

and nucleus; however, flat lesions exclusively expressed b-catenin on the cell

membrane. This interesting observation suggests different molecular pathways

for different cancerous lesions. Some lesions may have loss of APC function

(DALM) and others may not. Mice expressing a dominant negative N-cadherin

have been shown to develop adenomas [41]. This mutant (NCAD Delta) dem-

onstrated abnormal proliferation, migration, and crypt loss, which eventually

lead to small intestinal adenomas. This unique model could yield an opportunity

to study the role of adhesion molecules and the low risk of small intestinal can-

cers in Crohn’s disease.

In another study, Cooper et al [42] also showed that colitis acts as a promoter

of cancer in mice with multiple intestinal neoplasia, which has a single germ line

mutation of APC. In this model, dextran sulfate sodium-associated colon cancer

occurred though the loss of APC via loss of heterozygosity, as in classic multiple

intestinal neoplasia.

These studies suggest that animal models could serve as valuable tools to

understand the molecular neoplastic pathways of colitis-associated cancer.

Clinical features of cancer in IBD patients

The increase in overall cancer risk in patients with UC results from the

increased incidence of both CRC and hepatobiliary cancer associated with

primary sclerosing cholangitis. An increased risk of CRC is increasingly

recognized in Crohn’s disease as well. Friedman et al [43] screened 259 patients

with Crohn’s colitis every 2 years and concluded that the probability of detecting

dysplasia or carcinoma was 22% by the fourth surveillance. Surveillance

colonoscopy is therefore advised for patients with chronic and extensive Crohn’s

colitis, as well as for CUC. The challenge is to identify risk factors for

malignancy in IBD patients and to develop screening and surveillance strategies

to permit earlier detection and, therefore, more effective therapy.

S. Murthy et al / Hematol Oncol Clin N Am 17 (2003) 525–537 531

Risk factors in IBD patients

Duration and extent of disease

The absolute risk of CRC in UC is 5% to 10% after 20 years of disease and

approximately 30% after 35 years [44]. The risk of CRC is related to the duration

of disease and the extent, with pancolitis patients having a 14.8 relative risk, 2.8

for left-sided colitis, and 1.7 for proctitis [44]. The risk of dysplasia and cancer in

IBD patients is more directly related to the duration and extent of colonic disease

than the age of onset [45,46]. IBD carcinoma occurs at a younger age than in

patients with non-IBD cancer. UC patients have a similar cancer distribution to

those with non-IBD colon malignancies, but patients with Crohn’s disease have a

higher percentage of right-sided lesions, perhaps reflecting the tendency for right-

sided Crohn’s colitis in patients with ileocolitis [47].

Strictures in UC and Crohn’s disease

Dysplasia and cancer are more likely to occur in UC strictures. Gumaste et al

[48] reported a 5% incidence of strictures in their UC patients, but 24% of the

strictures were malignant. Strictures occurring after 20 years of disease were

more likely to be malignant in their study. Strictures occurring in the left colon

and rectum were more often benign. Therefore, those strictures occurring after

20 years of disease, located proximal to the splenic flexure, and presenting with

obstructing symptoms favor malignancy. Although strictures in UC may reveal

no evidence of dysplasia or malignancy by biopsy, several UC stricture cases

were reported that had previously been negative by biopsy but were later found to

have advanced-stage carcinoma, thus raising the question as to whether UC

strictures require colectomy or whether surveillance strategies are appropriate in

such patients [49]. If strictures cannot be adequately evaluated by colonoscopy,

then colectomy should be considered. Strictures in the presence of colonic

Crohn’s disease have been reported to have a 6.8% frequency of malignancy

compared with a 0.7% cancer incidence in similar patients without strictures [50].

Perianal fistulas and malignancy

Rectal carcinoma may present as an acute perianal fistula. One of the authors

(H.C.) encountered a patient who presented in this fashion and the malignant

nature of the fistula was not apparent until a surgical biopsy was performed. In

other instances, the fistulas may be chronic and secondarily develop malignancy.

Carcinoma should be considered when perianal fistulas result in progressive pain,

fail to heal with medical therapy, or rectal stricturing occurs.

Influence of treatment on IBD cancer

The possibility that long-term immunosuppressive therapy such as aza-

thioprine or 6-mercapatopurine could lead to an increased frequency of neo-

S. Murthy et al / Hematol Oncol Clin N Am 17 (2003) 525–537532

plasms is a concern to all who manage patients with IBD. No increased risk of

carcinoma or non-Hodgkin’s lymphoma was found in one series of 775 IBD

patients [51]. Eighty-six patients had been given 2 mg/kg of azathioprine for a

median of 12.5 months. The follow-up was for a median of 9 years. There was no

cancer in either the patients treated with azathioprine or those who did not receive

the therapy; however, many IBD patients are receiving immunosupressive

therapy for considerably longer periods than 1 year, raising the possibility that

the incidence of carcinoma could increase over time. Lymphomas occur with

greater frequency in IBD patients, although the increased incidence appears to be

relatively small. The influence of azathioprine and 6-MP on the development of

lymphoma appears to be little, if any, whereas there is insufficient evidence at

present (because of the relatively short follow-up) to determine whether inflix-

imab promotes malignancies.

Primary sclerosing cholangitis

Marchesa et al [52] compared 27 patients with primary sclerosing cholangitis

with a UC cohort of 1185 patients. Dysplasia during cancer surveillance biopsies

prompted colectomy in 16 patients (59.5%) compared with 11.5% in control

patients. A population-based cohort of 125 primary sclerosing cholangitis

patients from the Swedish Cancer Registry reported a cumulative cancer risk

of 25% after 10 years of UC [53].

Positive family history of sporadic cancer

Patients with UC and Crohn’s disease who have a family history of CRC have

a more than twofold increase in their cancer risk [54]. The risk appears to be

higher if the family member developed cancer prior to age 50 (which would also

increase the cancer risk in non-IBD patients).

Cancer surveillance in UC

Surveillance colonoscopy is predicated on the supposition that detection of

dysplasia will identify those at greatest risk of proceeding to cancer and thus

prompt prophylactic colectomy. The most cost-effective frequency of colonos-

copic surveillance has not been established. There is a consensus that surveil-

lance colonoscopy should be initiated after 8 to 10 years of pancolitis, and started

somewhat later for patients with a history of left-sided colitis. Between 10 and

20 years of disease, the frequency could be every 2 to 3 years but yearly after

20 years of disease. Two biopsies are obtained at 10-cm intervals and placed in

four fixative jars: right, transverse, left, and recto-sigmoid. The procedure should

not be performed during active disease because the regeneration process could

simulate low-grade dysplasia.

Dysplasia is a neoplastic epithelial change without infiltration into the lamina

propria. Many physicians mistakenly believe that dysplasia is a preneoplastic

S. Murthy et al / Hematol Oncol Clin N Am 17 (2003) 525–537 533

disorder, and in one study [55], only 19% correctly defined the term. Further-

more, the designation ‘‘low-grade dysplasia’’ is somewhat unfortunate because it

suggests a benign variant of dysplasia and may thus result in a false sense of

security or inaction as regards colectomy [56]. The average time for progression

of low-grade dysplasia to carcinoma has been reported to be 6.3 years. The

detection of high-grade dysplasia by surveillance colonoscopy not only predicts

future CRC but may also be associated with existing malignancy. Although 69%

of physicians would recommend colectomy for high-grade dysplasia (the

remainder favored continued surveillance), most physicians, unfortunately, would

pursue surveillance for patients with low-grade dysplasia [55]. It is clear that

many physicians do not follow the colectomy recommendations for either low-

grade or high-grade confirmed dysplasia. A diagnosis of low-grade or high-grade

dysplasia should be confirmed by a second pathologist with IBD expertise.

CRC occurs in approximately 40% of patients with a DALM [57]. The finding

of a DALM should prompt colectomy. There has been some question, however,

about the management of patients with chronic UC who are found to have a

colonic adenoma. These lesions could be a subtype of DALM, which may not be

associated with the high predictability for CRC as noted in chronic UC patients.

The distinction between a sporadic adenoma and a DALM is important. If the

lesion lies outside the chronic UC distribution (right sided, for instance, in

patients with left-sided UC) a sporadic adenoma is likely and colectomy need not

be advised. If an adenoma that endoscopically and histologically is compatible

with a sporadic adenoma lies within the distribution of the chronic UC and is

completely removed, multiple biopsies of the ‘‘flat’’ mucosa should be obtained.

If these are negative for dysplasia, surveillance strategies, rather than colectomy,

can be considered [58,59] (Fig. 2).

Fig. 2. Adenoma in the presence of chronic ulcerative colitis. Colectomy should be considered if: (1)

patient is younger than the ‘‘polyp’’ age (less than 40 years of age); (2) polyp cannot be completely

removed or recurs; (3) dysplasia is found in any flat mucosal biopsies; or (4) the patient is not

compliant or a colonoscopy is difficult to perform.

S. Murthy et al / Hematol Oncol Clin N Am 17 (2003) 525–537534

It often is difficult to convince asymptomatic IBD patients about the

importance of surveillance colonoscopies for the early detection of dysplasia or

cancer. Patient education about their potential risks is the most useful motivating

strategy. Perhaps the future availability of more sensitive and reliable predictors

of IBD-associated CRC will permit more specific targeting of patients at risk, and

reduce the frequency of examinations for the remaining patients.

References

[1] Blumberg RS, Strober W. Prospects for research in inflammatory bowel disease. JAMA 2001;

285:643–7.

[2] Van Heel DA, Satsangi J, Carey AH, Jewell DP. Inflammatory bowel disease: progress toward a

gene. Can J Gastroenterol 2000;14:207–18.

[3] Fuss IJ, Neurath M, Boirivant M, et al. Disparate CD4+ lamina propria (LP) lymphokine

secretion profiles in inflammatory bowel disease: Crohn’s disease LP cells manifest increased

secretion of IFN-gamma whereas ulcerative colitis LP cells manifest increased secretion of IL-5.

J Immunol 1996;157:1261–70.

[4] Eaden J, Abrams K, Ekbom A, Jackson E, Mayberry J. Colorectal cancer prevention in ulcerative

colitis: a case-control study. Aliment Pharmacol Ther 2000;14:145–53.

[5] Fireman Z, Grossman A, Lilos P, et al. Intestinal cancer in patients with Crohn’s disease:

a population study in central Israel. Scand J Gastroenterol 1989;24:346–50.

[6] Levin B. Inflammatory bowel disease and colon cancer. Cancer 1992;70:1313–6.

[7] Ekbom A, Helnick C, Zach M, Adani HU. Ulcerative colitis and colorectal cancer. N Eng J Med

1990;323:1228–33.

[8] Brostrom O, Lofberg R, Nordenvall B, et al. The risk of colorectal cancer in ulcerative colitis.

An epidemiologic study. Scand J Gastroenterol 1987;221:1193–4.

[9] Greenstein A, Sachar DB, Smith H, et al. Cancer in universal and left-sided ulcerative colitis.

Gastroenterology 1979;77:290–4.

[10] Bitt JH, Morrison B. Dysplasia and cancer in inflammatory bowel disease [editorial]. Gastro-

enterology 1981;80:865–8.

[11] Blackstone MO, Riddell RH, Rogers BHG, et al. Dysplasia-associated lesions or mass (DALM)

detected by colonoscopy in longstanding ulcerative colitis. An indication for colectomy. Gastro-

enterology 1981;80:366–74.

[12] Rubio CA, Johansson C, Slezak P, et al. Villous dysplasia: an ominous histologic sign in colitis

patients. Dis Colon Rectum 1984;27:283–7.

[13] Day DW, Morrison DC. The adenoma-carcinoma sequence. In: Morrison DC, editor. The patho-

genesis of colorectal cancer. Philadelphia: W.B. Saunders; 1978. p. 81–94.

[14] Cooper HS. Benign intestinal neoplasms. In: Ming SC, Goldman H, editors. Pathology of the

gastrointestinal tract. Philadelphia: W.B. Saunders; 1992. p. 786–815.

[15] Connell WR, Lennard-Jones JE, Williams CB, et al. Factors affecting the outcome of endoscopic

surveillance for cancer and ulcerative colitis. Gastroenterology 1994;107:934–44.

[16] Ranshoff DF, Riddell RH, Levin B. Ulcerative colitis in colonic cancer. Problems in assessing

the diagnostic usefulness of mucosal dysplasia. Dis Colon Rectum 1985;28:383–8.

[17] Taylor BA, Pemberton JH, Carpenter HA, et al. Dysplasia in chronic ulcerative colitis. Impli-

cations for colonoscopic surveillance. Dis Rectum 1992;35:950–6.

[18] Harpaz N, Peck AL, Yin J, et al. p53 protein expression in ulcerative colitis-associated colorectal

dysplasia and carcinoma. Hum Pathol 1994;25:1069–74.

[19] Burmer GC, Crispin DA, Kolli DR, et al. Frequent loss of a p53 allele in carcinomas and their

precursors in ulcerative colitis. Cancer Commun 1991;3:167–72.

[20] Yin J, Harpaz N, Tong Y, et al. p53 point mutations in dysplastic and cancerous ulcerative colitis

lesions. Gastroenterology 1993;104:1633–9.

S. Murthy et al / Hematol Oncol Clin N Am 17 (2003) 525–537 535

[21] Brentnall TA, Crispin DA, Rabinovitch PS, et al. Mutations in the p53 gene: an early marker of

neoplastic progression in ulcerative colitis. Gastroenterology 1994;107:369–78.

[22] Hussain SP, Amstad P, Raja K, et al. Increased p53 mutation load in noncancerous colon tissue

from ulcerative colitis: a cancer-prone chronic inflammatory disease. Cancer Res 2000;60:

3333–7.

[23] Hudson JD, Shoaibi MA, Maestro R, et al. A proinflammatory cytokine inhibits p53 tumor

suppressor activity. J Exp Med 1999;190:1375–82.

[24] Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell 1990;61:759–67.

[25] Odze RD, Brown CA, Hartmann CJ, et al. Genetic alterations in chronic ulcerative colitis-

associated adenoma-like DALMs are similar to non-colitis sporadic adenomas. Am J Surg Pathol

2000;24:1209–16.

[26] Burmer GC, Levine DS, Kulander BG, et al. c-Ki-ras mutations in chronic ulcerative colitis and

sporadic colon carcinoma. Gastroenterology 1990;99:416–20.

[27] Holzmann K, Klump B, Borchard F, et al. Comparative analysis of histology, DNA content p53

and Ki-ras mutations in colectomy specimens with long-standing ulcerative colitis. Int J Cancer

1998;76:1–6.

[28] Chen J, Compton C, Cheng E, et al. c-Ki-ras mutations in dysplastic fields and cancers in

ulcerative colitis. Gastroenterology 1992;102:1983–7.

[29] Lang SM, Stratakis DF, Heinzlmann M, Heldwein W, Wiebecke B, Loeschke K. Molecular

screening of patients with long standing extensive ulcerative colitis: detection of p53 and Ki-ras

mutations by single strand conformation polymorphism analysis and differential hybridisation

in colonic lavage fluid. Gut 1999;44:822–5.

[30] Brentnall TA, Rubin CE, Crispin DA, et al. A germline substitution in the human MSH2 gene is

associated with high grade dysplasia and cancer in ulcerative colitis. Gastroenterology 1995;109:

151–5.

[31] Hunt LE, Eichenberger MR, Petras R, et al. Use of microsatellite marker in predicting dysplasia

in ulcerative colitis. Arch Surg 2000;135:582–5.

[32] Souza RF, Lei J, Yin J, et al. A transforming growth factor beta 1 receptor type II mutation in

ulcerative colitis-associated neoplasms. Gastroenterology 1997;112:40–5.

[33] Hsieh CS, Klump S, Holzmann K, et al. Hypermethylation of the p16INK4a promoter in

colectomy specimens of patients with long-standing and extensive ulcerative colitis. Cancer

Res 1998;58:3942–5.

[34] Jean-Pierre I, Ahuja N, Toyota M, et al. Accelerated age-related CpG island methylation in

ulcerative colitis. Cancer Res 2001;61:3573–7.

[35] Walsh S, Murphy M, Silverman M, et al. p27 expression in inflammatory bowel disease-

associated neoplasia: further evidence of a unique molecular pathogenesis. Am J Pathol 1999;

155:1511–8.

[36] Ilyas M, Thomlinson IPM. The interactions of APC, E-cadherin and b-catenin in tumor develop-

ment and progression. J Pathol 1997;182:128–37.

[37] Karayiannakis AJ, Skarayiayrigos KN, Efstathiou J, et al. Expression of catenins in E-cadherins

during epithelial restitution in inflammatory bowel disease. J Pathol 1998;185:413–8.

[38] Mikami T, Mitomi H, Hara A, et al. Decreased expression of CD44, alpha-catenin, and deleted

colon carcinoma and altered expression of beta-catenin in ulcerative colitis-associated dysplasia

and carcinoma, as compared with sporadic colon neoplasms. Cancer 2000;89:733–40.

[39] Bertone ER, Giocannucci EL, King Jr NW, et al. Family history as a risk factor for ulcerative

colitis-associated colon cancer in cotton top tamarin. Gastroenterology 1998;114:669–74.

[40] Cooper HS, Murthy S, Kido K, et al. Dysplasia and cancer in the dextran sulfate sodium

mouse colitis model. Relevance to colitis-associated neoplasia in the human: a study of

histopathology, b-catenin and p53 expression and the role of inflammation. Carcinogenesis

2000;21:757–68.

[41] Hermiston ML, Gordon JI. Inflammatory bowel disease and adenomas in mice expressing a

dominant negative N-cadherin. Science 1995;270:1203–7.

[42] Cooper HS, Everly L, Chang WC, et al. The role of mutant Apc in the development of dysplasia

S. Murthy et al / Hematol Oncol Clin N Am 17 (2003) 525–537536

and cancer in the mouse model of dextran sulfate sodium induced colitis. Gastroenterology 2001;

121(4):1407–14.

[43] Friedman S, Rubin PH, Bodian C, et al. Screening and surveillance colonoscopy in chronic

Crohn’s colitis. Gastroenterology 2001;120:820–6.

[44] Ekbom E, Helmick C, Zack M, et al. Ulcerative colitis and colorectal cancer. New Eng J Med

1990;323:1228–33.

[45] Sugita A, Sachar DB, Bodian C, et al. Colorectal cancer in ulcerative colitis. Influence of

anatomical extent and age at onset on colitis-cancer interval. Gut 1991;32:167–9.

[46] Markowitz J, McKinley M, Kahn E, et al. Endoscopic screening for dysplasia and mucosal

aneuploidy in adolescents and young adults with childhood onset colitis. Am J Gastroenterol

1997;92:2001–6.

[47] Bansal P, Sonnenberg A. Risk factors of colorectal cancer in inflammatory bowel disease. Am J

Gastroenterol 1996;91:44–8.

[48] Gumaste V, Sachar DB, Greenstein AJ. Benign and malignant colorectal strictures in ulcerative

colitis. Gut 1992;33:938–41.

[49] Reiser JR, Waye JD, Janowitz HD, et al. Adenocarcinoma in strictures of ulcerative colitis

without antecedent dysplasia by colonoscopy. Am J Gastroenterol 1994;89:119–22.

[50] Yamazaki Y, Ribeiro MB, Sachar DB, et al. Malignant colorectal strictures in Crohn’s disease.

Am J Gastroenterol 1991;86:882–5.

[51] Connell WR, Kamm MA, Dickson M, et al. Long-term neoplasia risk after azathioprine treat-

ment in inflammatory bowel disease. Lancet 1994;343:1249–52.

[52] Marchesa P, Lashner BA, Lavery IC, et al. The risk of cancer and dysplasia among ulcerative

coilitis patients with primary sclerosing cholangitis. Am J Gastroenterol 1997;92:1285–8.

[53] Kornfeld D, Ekbom A, Ihre T. Is there an excess risk for colorectal cancer in patients with

ulcerative colitis and concomitant primary sclerosing cholangitis? A population based study. Gut

1997;41:522–5.

[54] Askling J, Dickman PW, Karlen P, et al. Family history as a risk factor for colorectal cancer in

inflammatory bowel disease. Gastroenterology 2001;120:1356–62.

[55] Bernsteiln CN, Weinstein WM, Levine DS, et al. Physician’s perceptions of dysplasia and

approaches to surveillance colonoscopy in ulcerative colitis. Am J Gastroenterol 1995;90:

2106–14.

[56] Hanauer SB. Surveying surveillance: are gastroenterologists consistently inconsistent, incon-

sistently consistent, or poorly educated? Gastrointest Endosc 2000;51(2):240–2.

[57] Bernstein CN. Are we telling patients the truth about surveillance colonoscopy in ulcerative

colitis? Lancet 1994;343:71–4.

[58] Engelsgjerd M, Farraye FA, Odze RD. Polypectomy may be adequate treatment for adenoma-

like dysplastic lesions in chronic ulcerative colitis. Gastroenterology 1999;117:1288–94.

[59] Odze RD. Adenomas and adenoma-like DALMs in chronic ulcerative colitis: a clinical, patho-

logical, and molecular review. Am J Gastroenterol 1999;94:1746–50.

[60] Willenbucher RF, Zelman SJ, Ferrell LD, et al. Chromosomal alterations in ulcerative colitis-

related neoplastic progression. Gastroenterology 1997;113:791–801.

S. Murthy et al / Hematol Oncol Clin N Am 17 (2003) 525–537 537


Recommended