Download Inflammation and colorectal cancer - Tel

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
Transcript
COPHAR-709; NO OF PAGES 6
Available online at www.sciencedirect.com
Inflammation and colorectal cancer
Sarah Kraus1 and Nadir Arber1,2
Patients with long-standing inflammatory bowel disease (IBD)
have an increased risk of developing colorectal cancer (CRC).
However, the underlying mechanisms are not entirely clear. A
genetic basis for the increased risk of CRC in IBD patients is
only a partial explanation. It is possible that high levels of
inflammatory mediators that are produced in this setting may
contribute to the development and progression of CRC.
Growing evidence supports a role for various cytokines,
released by epithelial and immune cells, in the pathogenesis of
IBD-associated neoplasia. Two key genes in the inflammatory
process, cyclooxygenase-2 (COX-2) and nuclear factor
kappaB (NF-kB), provide a mechanistic link between
inflammation and cancer while other factors such as, TNF-a
and IL-6-induced signaling have been recently shown to
promote tumor growth in experimental models of colitisassociated cancer. This article reviews the pathogenesis of
IBD-related CRC and summarizes the molecular mechanisms
underlying the development of intestinal neoplasia in the setting
of chronic inflammation.
Addresses
1
Integrated Cancer Prevention Center, Tel Aviv Souraski Medical
Center, 6 Weizmann Street, Tel Aviv 64239, Israel
2
Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
Corresponding author: Arber, Nadir ([email protected])
Current Opinion in Pharmacology 2009, 9:1–6
This review comes from a themed issue on
Cancer
Edited by Bharat B Aggarwal
1471-4892/$ – see front matter
Published by Elsevier Ltd.
DOI 10.1016/j.coph.2009.06.006
Introduction
The functional relationship between inflammation and
cancer has long been recognized and was already established two centuries ago. This link was made on the basis
of several observations (reviewed in [1]):
1. Tumors arise at sites of chronic inflammation.
2. Inflammatory cells, chemokines and cytokines are
present in tumors.
3. Overexpression of cytokines and chemokines can
induce cancer.
4. The same molecular targets and similar pathways are
activated or shut down in inflammation, as well as in
the carcinogenesis process.
www.sciencedirect.com
5. Epidemiological studies have shown that chronic
inflammatory states increase the risk of numerous
cancers.
6. Non-steroidal anti-inflammatory drugs (NSAIDs)
reduce the incidence and the mortality of several
cancers.
The role of the immune system during cancer development is complex, involving extensive reciprocal interactions between genetically altered cells, adaptive and
innate immune cells, their soluble mediators and structural components present in the tumor microenvironment
[2].
In this review we discuss the model of colorectal cancer
(CRC) and inflammation as the proof of concept linking
between these two processes.
CRC remains a major health concern in the Western
world. There is a body of literature suggesting a link
between chronic inflammation and CRC, in which gastrointestinal (GI) inflammation may contribute to the promotion of CRC development. Anti-inflammatory
treatment is known to reduce GI neoplasia, while CRC
incidence is increased in inflammatory bowel disease
(IBD) [3].
Human CRC can be classified by etiology as: (1) inherited, including hereditary non-polyposis colorectal cancer
(Lynch Syndrome/HNPCC) due to genetic instability,
and familial adenomatous polyposis coli (FAP) due to a
mutation in the adenomatous polyposis coli gene, APC;
(2) inflammatory, including Crohn’s disease (CD) and
ulcerative colitis (UC); or (3) sporadic, accounting for
80% of CRCs but with poorly defined etiology.
Patients with long-standing IBD have an increased risk of
developing CRC [4]. In fact, although patients with IBD
represent only a small fraction of CRC cases (1–2%), these
patients are among those at greatest risk of CRC in the
general population. Moreover, in patients with prolonged
(>30 years) and extensive colitis, the risk of CRC is much
higher (18%). Risk factors for CRC include extent and
duration of UC, primary sclerosing cholangitis, a family
history of sporadic CRC, severity of bowel inflammation
and early onset of the disease [3].
Many of the molecular alterations responsible for sporadic
CRC, namely chromosomal and microsatellite instability,
and hypermethylation also play a role in colitis-associated
CRC carcinogenesis. This article provides an overview on
the pathogenesis of IBD-related colorectal neoplasia and
Current Opinion in Pharmacology 2009, 9:1–6
Please cite this article in press as: Kraus S, Arber N. Inflammation and colorectal cancer, Curr Opin Pharmacol (2009), doi:10.1016/j.coph.2009.06.006
COPHAR-709; NO OF PAGES 6
2 Cancer
some of the molecular mechanisms underlying malignant
transformation in the mucosa of IBD patients.
signaling. A role for Notch signaling in cancer has extensively been reported (reviewed in [13]).
Chronic inflammation and tumor development
Many human cancers exhibit elevated prostaglandin (PG)
levels owing to upregulation of COX-2, a key enzyme in
eicosanoid biosynthesis. PGE(2) is a major downstream
mediator of COX-2 that promotes cellular proliferation
and angiogenesis, inhibits apoptosis, enhances invasiveness, and modulates immunosuppression. COX-2 is significantly overexpressed in a variety of tumors, including
colorectal neoplasms, making it an attractive therapeutic
target [14]. Although COX-2 is regularly expressed at low
levels in colonic mucosa, its activity increases dramatically following mutation of the APC gene, suggesting that
the APC-b-catenin/TCF signaling activity may regulate
COX-2 gene expression.
Chronic inflammation is characterized by a continued
active inflammatory response and tissue destruction.
Many of the immune cells contributing to the pathology
of chronic inflammation (e.g. macrophages, neutrophils
and eosinophils) are involved directly or by producing
inflammatory cytokines that may influence the carcinogenesis process [5].
Chronic inflammation promotes carcinogenesis by inducing gene mutations, inhibiting apoptosis, or stimulating
angiogenesis and cell proliferation [6]. Inflammation also
induces epigenetic alterations that are associated with
cancer development. Two key genes in the inflammatory
process, cyclooxygenase-2 (COX-2) and nuclear factor
kappaB (NF-kB), provide a mechanistic link between
inflammation and cancer and are targets for chemoprevention and particularly, in CRC [7].
NF-kB transcription factors have a key role in many
physiological processes and disease conditions. Given
its important role in mediating inflammatory signals, a
lot of attention has been focused on the role of NF-kB and
its upstream activator, IkB kinase (IKK), and their involvement in inflammation and cancer [8].
NF-kB is not a single gene but represents a family of
closely related transcription factors that includes five
genes NF-kB1 (p50/p105), NF-kB2 (p52/p100), RelA
(p65), c-Rel and RelB. NF-kB regulates the expression
of genes, many of which play important roles in the
regulation of inflammation and apoptosis. Although RelA,
RelB and NF-kB1 genetic alterations are rare in human
cancer, these proteins are constitutively activated in a
wide variety of human tumors and have been associated
with tumor progression [9]. Constitutive NF-kB activation has been recently evaluated in CRC [10]. Thus,
constitutively active NF-kB was found in 40% of CRC
tissues and 67% of cell lines, and shown to promote tumor
growth.
The upstream IkB kinases are also constitutively active in
some types of cancer. Defective IKKa was found in
several solid tumors such as breast, colon, ovarian, pancreatic, bladder, prostate carcinomas and melanoma [11].
A recent study has shown that IKKa is activated in
colorectal tumors and associates to the chromatin of
specific Notch targets, leading to the release of the corepressor SMRT (silencing mediator of retinoic acid and
thyroid hormone receptor) [12]. The Notch pathway
plays an extensive role in the immune system and operates at various levels by acting in conjunction with
defined immunological signals such as cytokines, T cell
antigen receptor and co-stimulatory receptor-mediated
Selective inhibitors of COX-2 (coxibs) have established
efficacy in the treatment of pain and inflammation comparable to that of non-selective NSAIDs but with superior
GI safety. Subsequently, additional pharmacologic activities have emerged outside of coxibs’ analgesic activity.
Celecoxib (Celebrex1, Pfizer, USA) is unique among the
coxibs and traditional NSAIDs, because this particular
drug displays the greatest potency to induce apoptotic
cell death and has clinically relevant anti-neoplastic
applications, arising from additional activities that may
exert anti-tumor functions independent of the COX-2
inhibitory activity [15].
Colitis-associated colorectal cancer
The association of IBD with CRC was first described by
Burrill Crohn in 1925 [16]. More than two decades
elapsed before the first description of a cancer of the
colon complicating ‘regional enteritis’ [17]. Later on,
additional case reports were published describing CRC
in patients with CD [18]. However, no statistically significant increase in cancer risk among these patients was
found, mainly because the absolute numbers of CDassociated cancer cases were usually small. Therefore,
CRC in CD has been less well studied and most of our
knowledge on the pathobiology of CRC is based on
studies of patients with UC.
In the early 1980s, a first attempt to calculate direct
comparisons between cancer incidences in CD and UC
was described. Studies conducted at The Mount Sinai
Hospital noted a parity of similar extent between the
relative risks of colon cancer in CD and in left sided
UC. Thus, it was suggested that ‘when cases of ulcerative and Crohn’s colitis of similar anatomic extent are
followed for similar durations of time, the two diseases
may ultimately prove to have similar increases in risk
for CRC [19]. Generally, both diseases are characterized
by chronic, relapsing inflammation of the intestines. It
is assumed that chronic inflammation is what causes
cancer.
Current Opinion in Pharmacology 2009, 9:1–6
Please cite this article in press as: Kraus S, Arber N. Inflammation and colorectal cancer, Curr Opin Pharmacol (2009), doi:10.1016/j.coph.2009.06.006
www.sciencedirect.com
COPHAR-709; NO OF PAGES 6
Inflammation and colorectal cancer Kraus and Arber 3
The neoplastic transformation in IBD is thought to be
similar to the adenoma–carcinoma sequence in sporadic
CRC (reviewed in [20]). However, unlike sporadic CRC
where dysplastic lesions arise in one or two focal areas of
the colon, in colitic mucosa, the dysplasia is usually
multifocal, reflecting a broader ‘field effect’.
The transition from normal epithelium to adenoma to
carcinoma is associated with acquired molecular events
involving mutations in oncogenes and tumor suppressor
genes, abnormal gene expression and genetic defects in a
variety of genes. This tumor progression model was
deduced by Fearon and Vogelstein from comparison of
genetic alterations seen in normal colon epithelium,
adenomas of progressively larger size, and malignancies
[21]. Among the earliest events in CRC carcinogenesis is
loss of the APC gene, which appears to be consistent with
its important role in predisposing carriers of germline APC
mutations to colorectal tumors. These studies showed
that the molecular steps that occur after the activation of
the APC/b-catenin/TCF pathway involve a non-linear
accumulation of specific genetic changes, which include
detectable losses at the molecular level of portions of
chromosome 5q, chromosome 18q, and chromosome 17p,
changes in methylation patterns and mutation of the Kras oncogene. The important genes involved in these
chromosome losses are APC(5q), DCC/MADH2/
MADH4(18q), and p53(17p).
Many of the molecular alterations responsible for sporadic
CRC development also play a role in colitis-associated
colon carcinogenesis. However, there are several differences in the sequence of molecular events leading from
dysplasia to invasive adenocarcinoma in IBD as compared
with sporadic CRC. For example, APC loss of function,
considered to be a common early event in sporadic CRC,
is much less frequent and usually occurs late in the colitisassociated dysplasia–carcinoma sequence. On the contrary, p53 mutations in sporadic neoplasia usually occur
late in the adenoma–carcinoma sequence, whereas in
patients with colitis, p53 mutations occur early and are
often detected in non-dysplastic mucosa [20].
Microsatellite instability and hypermethylation
Chromosomal instability, microsatellite instability (MSI),
and hypermethylation are assuming increasing importance as mechanisms contributing to the genetic alterations in IBD. MSI due to mismatch repair deficiency has
been reported to occur at variable frequencies in IBDassociated intestinal neoplasias [22]. Moreover, the
mutational events in target genes for instability in these
cancers are similar to those in sporadic and hereditary
colorectal cancers, indicating a colon-related repertoire of
target gene alterations.
About 12–15% of sporadic CRCs display defective DNA
mismatch repair, manifested as high levels of MSI. In this
www.sciencedirect.com
group of cancers, promoter hypermethylation of the mismatch repair gene hMLH1 is strongly associated with
MSI. Silencing the hMLH1 gene expression by its promoter methylation stops the formation of MLH1 protein,
and prevents the normal activation of the DNA repair
gene. This is an important cause for microsatellite
instability and cell proliferation to the point of colorectal
cancer formation [23]. In IBD neoplasia, hMLH1 promoter hypermethylation is a frequent event. Furthermore, hMLH1 hypermethylation and MSI are strongly
associated with reduced hMLH1 protein expression in
IBD-related neoplasia, suggesting that its hypermethylation causes defective DNA mismatch repair in at least a
subset of IBD cancers [24].
CpG island hypermethylation is a mechanism of gene
silencing that can be used by neoplastic cells to inactivate
undesirable genes. Methylation of CpG islands in several
genes seems to precede dysplasia and is more widespread
in UC patients [25]. Schulmann et al. have determined the
frequency of high-level MSI and the mutational and
methylation profile of IBD-related neoplasia, and found
that the profiles of coding microsatellite mutations (instabilotypes) differ significantly between IBD cancers and
sporadic CRCs [26]. Specifically, TGFBR2 and ACVR2
mutations are significantly rarer in IBD. Furthermore,
HPP1 methylation occurs early, in non-dysplastic and
dysplastic mucosa, whereas RAB32 methylation occurs
at the transition to invasive growth, being rarer in dysplasias. Therefore, it is reasonable to assume that these
mutations evolve through different pathways, leading to
differentially expressed instabilotypes.
Tumor necrosis factor-alpha
Growing evidence supports a role for various cytokines,
released by epithelial and immune cells, in the pathogenesis of colitis-associated cancer. Cytokines are essential mediators of the interactions between activated
immune cells and non-immune cells, including epithelial
and mesenchymal cells. Tumor necrosis factor-alpha
(TNF-a) is a potent pro-inflammatory cytokine thought
to be involved in the pathogenesis of IBD. Recent data
indicate that TNF-a promotes tumor development in
models of experimental colitis. Popivanova et al. have
recently shown that blockade of TNF-a reduces the
formation of colorectal tumors in mice lacking the
TNF receptor, p55 (TNF-Rp55 knockout) [27]. Combined treatment with azoxymethane and dextran sodium
sulfate (AOM/DSS), which causes severe colonic inflammation and the subsequent development of multiple
tumors, induced the intracolonic expression of TNF-a,
which in turn regulated the trafficking of inflammatory
cells, a major source of COX-2. These findings are of
particular interest because they suggest that blocking
of TNF signaling can reverse tumorigenesis even
when CRC is already present, probably by reducing
the infiltration of inflammatory cells and, consequently,
Current Opinion in Pharmacology 2009, 9:1–6
Please cite this article in press as: Kraus S, Arber N. Inflammation and colorectal cancer, Curr Opin Pharmacol (2009), doi:10.1016/j.coph.2009.06.006
COPHAR-709; NO OF PAGES 6
4 Cancer
the circulating levels of COX-2. Therefore, strategies
targeting TNF-a could potentially provide effective
anti-tumor therapies.
It should be noted that the regulation of TNF-a is partly
genetic. Single nucleotide polymorphisms (SNPs) in the
promoter of TNF-a were found associated with an
increased risk for IBD and may genetically predispose
towards developing colitis-associated CRC [28].
Nuclear factor kappaB
Although the molecular mechanisms linking IBD with
CRC are not well understood, recent studies in preclinical models point to NF-kB as a central player. NFkB regulates the expression of various cytokines and
modulates the inflammatory processes in IBD. On the
contrary, NF-kB stimulates the proliferation of tumor
cells and enhances their survival through the regulation
of anti-apoptotic genes.
Pro-inflammatory cytokines have been suggested to
regulate pre-neoplastic growth during colitis-associated
tumorigenesis. Interleukin 6 (IL-6) is a multifunctional
NF-kB-regulated cytokine that acts on epithelial and
immune cells. The importance of the IL-6 family of
pro-inflammatory cytokines and their downstream effector STAT3 in colitis-associated CRC has been recently
described (reviewed in [29]). Human patients with CRC
have been reported to have elevated levels of IL-6 and
increased levels of IL-6 have also been shown in murine
experimental models of colitis-associated cancer induced
by AOM/DSS [30].
STAT3, a nuclear transcription factor downstream of
gp130, is necessary for the growth of colitis-associated
CRC in mice [31,32]. It appears to function through
increased epithelial proliferation and protection against
AOM/DSS-induced epithelial cell apoptosis. Interestingly, DSS-induced mucosal inflammation or colitis was
markedly increased in both IL-6-deficient mice and in
mice lacking STAT3 in intestinal epithelial cells, which
correlated with an increase in the expression of proinflammatory cytokines within the colonic mucosa.
Oxidative stress
The exact mechanism by which chronic inflammation
results in carcinogenesis is still unclear. Persistent inflammation is believed to result in increased cell proliferation
as well as oxidative stress, leading to the development of
dysplasia [33]. Oxidative stress develops particularly in
inflammatory reactions because the inflammatory cells,
activated neutrophils, and macrophages produce large
amounts of reactive oxygen and nitrogen species (RONs).
DNA damage caused by oxidative stress in the characteristic damage-regeneration cycle is a major contributor
to CRC development in UC patients. Thus, oxidative
stress-induced cellular damage may provide a mechan-
istic basis for many of the events thought to drive UCassociated colon carcinogenesis in humans and animal
models, including specific gene alterations, genetic
instability and aberrant methylation. The available evidence suggests that DNA damage caused by oxidative
stress in the characteristic damage-regeneration cycle is a
major contributor to CRC development in UC patients.
RONs produced by inflammatory cells can interact with
key genes involved in carcinogenic pathways such as p53,
DNA mismatch repair genes, and others. Factors such as
NF-kB and COX-2 may also contribute.
Studies in animal models of UC have provided support as
to the involvement of oxidative stress in inflammationdriven CRC. Seril et al. have recently examined the role of
nitric oxide (NO) in UC-associated colorectal carcinogenesis using the DSS-induced and iron-enhanced model of
chronic UC in inducible nitric oxide synthase (iNOS)
knockout mice. These results showed that there is no
difference in UC-associated cancer development in
iNOS+/+ and iNOS / mice, suggesting that in the
absence of iNOS, other factors, such as eNOS, may play
a role in nitrosative stress and UC-associated carcinogenesis in this model system [34].
Notably, UC patients frequently experience iron
deficiency anemia owing to chronic disease and colonic
blood loss, and anemia is corrected in these individuals by
iron supplementation. It has been suggested that iron
over nutrition may contribute to the carcinogenesis process by augmenting oxidative damage and inflammationcaused epithelial proliferation. However, further clinical
studies are needed to clarify this issue [35].
Transforming growth factor-beta and other factors
Despite a primary tumor suppressor role, there is increasing evidence suggesting that transforming growth factorbeta (TGF-b) can promote tumor growth, invasion and
metastasis in advanced stages of CRC. TGF-b has been
shown to attenuate an anti-tumor immune response
through the induction of regulatory T cells in spontaneous and inflammation-associated cancer. However,
IL-6 signaling promotes tumor growth in experimental
colitis-associated cancer, and this signaling has been
shown to be inhibited by TGF-b [36]. Therefore, the
role of TGF-b in this setting is not entirely clear.
Members of the IL-12 family, p35/p40 play an important
role in T helper (Th) cell polarization and Th1 T cell
differentiation. Recent findings have shown that both p35
and p40 can form other cytokines by heterodimerizing
with different proteins (IL-23: p19/p40; IL-35: p35/
EBI3). Furthermore, the cytokine IL-27 (EBI3/p28)
was recently identified as a member of the IL-12 family.
These cytokines have been implicated in the pathogenesis of colitis and IL-23, which plays a key role in the
induction and maintenance of gut inflammation during
Current Opinion in Pharmacology 2009, 9:1–6
Please cite this article in press as: Kraus S, Arber N. Inflammation and colorectal cancer, Curr Opin Pharmacol (2009), doi:10.1016/j.coph.2009.06.006
www.sciencedirect.com
COPHAR-709; NO OF PAGES 6
Inflammation and colorectal cancer Kraus and Arber 5
IBD, seems to be involved in inflammation-associated
carcinogenesis (reviewed in [37]).
While some cytokines promote tumor growth, others such
as IL-10, provide an anti-tumor effect. IL-10-deficient
mice develop colitis and colitis-associated cancer within
two to three weeks after birth. The IL-10 knockout
mouse model results in a disease similar to human IBD
and therefore, has been proven useful as an experimental
model for developing new and effective therapies for
active IBD and CD, in particular. However, a previous
report aiming to identify possible mutations of oncogenes
and tumor suppressor genes involved in colorectal tumorigenesis in these mice, showed no alterations in K-ras,
p53, APC and MSH genes, suggesting that other genes are
involved in the development of these tumors [38]. Therefore, despite the high similarities between the histopathological pattern of CRCs in IL-10-deficient mice and IBDassociated carcinomas, this model is apparently not appropriate for investigating IBD-associated carcinogenesis.
Since most of the above mentioned cytokines are
involved in both, inflammation and carcinogenesis, it is
difficult to assess their contribution to the individual steps
during pathogenesis of colitis-associated cancer.
Conclusion
Patients with IBD have an increased risk of developing
CRC. This risk appears to be closely associated to the
cumulative effect of chronic inflammation and is in
direct correlation with the severity of the inflammation
and the extent and duration of the disease. The exact
mechanism as to how chronic inflammation results in
carcinogenesis is not resolved, although it is becoming
clear that chronic inflammation and IBD in particular, is
an important driving mechanism that promotes the development of cancer in the colon. Further studies are
needed in order to elucidate the pathogenesis of colitisassociated CRC.
References and recommended reading
Papers of particular interest published within the period of review have
been highlighted as:
of special interest
of outstanding interest
1. Mantovani A, Allavena P, Sica A, Balkwill F: Cancer-related
inflammation. Nature 2008, 454:436-444.
This article reviews the current knowledge of the molecular and cellular
pathways that link inflammation and cancer and describes how these
pathways suppress anti-tumor immunity during cancer progression.
2.
de Visser KE, Eichten A, Coussens LM: Paradoxical roles of the
immune system during cancer development. Nat Rev Cancer
2006, 6:24-37.
3.
Lakatos PL, Lakatos L: Risk for colorectal cancer in ulcerative
colitis: changes, causes and management strategies. World J
Gastroenterol 2008, 14:3937-3947.
This review discusses recent epidemiological trends and risk factors for
CRC in patients with ulcerative colitis. The authors also describe the
contribution of genetic factors to the pathogenesis of CRC in IBD.
www.sciencedirect.com
4.
Munkholm P: Review article: the incidence and prevalence of
colorectal cancer in inflammatory bowel disease. Aliment
Pharmacol Ther 2003, 18(Suppl. 2):1-5.
5.
Seruga B, Zhang H, Bernstein LJ, Tannock IF: Cytokines and
their relationship to the symptoms and outcome of cancer.
Nat Rev Cancer 2008, 8:887-899.
This is a perspective review that discusses the role of cytokines in causing
symptoms in patients with cancer and their influence in the development
and progression of cancer.
6.
Kundu JK, Surh YJ: Inflammation: gearing the journey to
cancer. Mutat Res 2008, 659:15-30.
7.
Burstein E, Fearon ER: Colitis and cancer: a tale of inflammatory
cells and their cytokines. J Clin Invest 2008, 118:464-467.
8.
Karin M: The IkappaB kinase—a bridge between inflammation
and cancer. Cell Res 2008, 18:334-342.
9.
Dolcet X, Llobet D, Pallares J, Matias-Guiu X: NF-kB in
development and progression of human cancer. Virchows Arch
2005, 446:475-482.
10. Sakamoto K, Maeda S, Hikiba Y, Nakagawa H, Hayakawa Y,
Shibata W, Yanai A, Ogura K, Omata M: Constitutive NF{kappa}B activation in colorectal carcinoma plays a key role in
angiogenesis, promoting tumor growth. Clin Can Res 2009,
15:2248-2258 Epub ahead of print.
This study evaluates the role of constitutive NF-kB activation in CRC cells
and shows that this activation plays a role in angiogenesis and antiapoptosis. These results suggest that NF-kB blockade may be a reasonable therapeutic approach for IBD and colitis-associated cancer.
11. Affara NI, Coussens LM: IKKalpha at the crossroads of
inflammation and metastasis. Cell 2007, 129:25-26.
12. Fernández-Majada V, Aguilera C, Villanueva A, Vilardell F, Robert
Moreno A, Aytés A, Real FX, Capella G, Mayo MW, Espinosa L,
Bigas A: Nuclear IKK activity leads to dysregulated notchdependent gene expression in colorectal cancer. Proc Natl
Acad Sci USA 2007, 104:276-281.
This study shows that IKK plays an NF-kB independent role in CRC and
suggests a role for IKK in the regulation of NF-kB-independent genes
such as, Notch targets.
13. Leong KG, Karsan A: Recent insights into the role of Notch
signaling in tumorigenesis. Blood 2006, 107:2223-2233.
14. Arber N: Cyclooxygenase-2 inhibitors in colorectal cancer
prevention: point. Cancer Epidemiol Biomarkers Prev 2008,
17:1852-1857.
15. Grösch S, Maier TJ, Schiffmann S, Geisslinger G:
Cyclooxygenase-2 (COX-2)-independent anticarcinogenic
effects of selective COX-2 inhibitors. J Natl Cancer Inst 2006,
98:736-747.
16. Crohn BB, Rosenberg H: The sigmoidoscopic picture
of chronic ulcerative colitis. Am J Med Sci 1925,
170:220-228.
17. Warren S, Sommers SC: Cicatrizing enteritis (regional enteritis)
as a pathologic entity: analysis of one hundred and twenty
cases. Am J Pathol 1948, 24:475-501.
18. Weedon DD, Shorter RG, Ilstrup DM, Huizenga KA, Taylor WR:
Crohn’s disease and cancer. N Engl J Med 1973,
289:1099-1102.
19. Sachar DB: Cancer in Crohn’s disease: dispelling the myths.
Gut 1994, 35:1507-1508.
20. Xie J, Itzkowitz SH: Cancer in inflammatory bowel disease.
World J Gastroenterol 2008, 14:378-389.
This review addresses the clinical and molecular features of colitisassociated CRC and also of other cancers in patients with IBD.
21. Fearon ER, Vogelstein B: A genetic model for colorectal
tumorigenesis. Cell 1990, 61:759-767.
22. Svrcek M, El-Bchiri J, Chalastanis A, Capel E, Dumont S,
Buhard O, Oliveira C, Seruca R, Bossard C, Mosnier JF et al.:
Specific clinical and biological features characterize
inflammatory bowel disease associated colorectal
cancers showing microsatellite instability. J Clin Oncol 2007,
25:4231-4238.
Current Opinion in Pharmacology 2009, 9:1–6
Please cite this article in press as: Kraus S, Arber N. Inflammation and colorectal cancer, Curr Opin Pharmacol (2009), doi:10.1016/j.coph.2009.06.006
COPHAR-709; NO OF PAGES 6
6 Cancer
IBD-associated intestinal neoplasias were screened for microsatellite
instability. The study shows that the mutational events in target genes
for instability were similar in IBD tumors and in CRCs.
23. Niv Y: Microsatellite instability and MLH1 promoter
hypermethylation in colorectal cancer. World J Gastroenterol
2007, 13:1767-1769.
24. Fleisher AS, Esteller M, Harpaz N, Leytin A, Rashid A, Xu Y, Liang J,
Stine OC, Yin J, Zou TT et al.: Microsatellite instability in
inflammatory bowel disease-associated neoplastic lesions is
associated with hypermethylation and diminished expression
of the DNA mismatch repair gene, hMLH1. Cancer Res 2000,
60:4864-4868.
25. Issa JP, Ahuja N, Toyota M, Bronner MP, Brentnall TA:
Accelerated age related CpG island methylation in ulcerative
colitis. Cancer Res 2001, 61:3573-3577.
26. Schulmann K, Mori Y, Croog V, Yin J, Olaru A, Sterian A, Sato F,
Wang S, Xu Y, Deacu E et al.: Molecular phenotype of
inflammatory bowel disease-associated neoplasms with
microsatellite instability. Gastroenterology 2005, 129:74-85.
27. Popivanova BK, Kitamura K, Wu Y, Kondo T, Kagaya T, Kaneko S,
Oshima M, Fujii C, Mukaida N: Blocking TNF-alpha in mice
reduces colorectal carcinogenesis associated with chronic
colitis. J Clin Invest 2008, 118:560-570.
This study demonstrates that TNF-a, through its effects on the immune
system, plays a crucial role in promoting neoplastic transformation in the
setting of chronic inflammation.
28. Garrity-Park MM, Loftus EV Jr, Bryant SC, Sandborn WJ,
Smyrk TC: Tumor necrosis factor-alpha polymorphisms in
ulcerative colitis-associated colorectal cancer.
Am J Gastroenterol 2008, 103:407-415.
Using DNA extracted from formalin-fixed, paraffin-embedded tissues from
ulcerative colitis-associated CRC cases and matched controls, the authors
have demonstrated an association between TNF-a polymorphisms and
UC–CRC. This is a novel finding supporting a genetic predisposition for the
development of colorectal neoplasia in IBD patients.
31. Grivennikov S, Karin E, Terzic J, Mucida D, Yu GY,
Vallabhapurapu S, Scheller J, Rose-John S, Cheroutre H,
Eckmann L, Karin M: IL-6 and Stat3 are required for survival of
intestinal epithelial cells and development of colitisassociated cancer. Cancer Cell 2009, 15:103-113.
This study demonstrates the importance of IL-6 and its downstream
effector STAT3 in colitis-associated colon cancer. It also establishes a
role for IL-6 and STAT3 signaling in IECs during inflammation-associated
colon carcinogenesis. IL-6 was identified as a crucial NF-kB-dependent
pro-tumorigenic cytokine. Both IL-6 and STAT3 are suggested as potential targets for the prevention and treatment of IBD neoplasia.
32. Bollrath J, Phesse TJ, von Burstin VA, Putoczki T, Bennecke M,
Bateman T, Nebelsiek T, Lundgren-May T, Canli O, Schwitalla S
et al.: gp130-mediated Stat3 activation in enterocytes
regulates cell survival and cell-cycle progression during
colitis-associated tumorigenesis. Cancer Cell 2009, 15:91-102.
This study demonstrates that in addition to IKK-dependent NF-kB activation, STAT3 comprises a central checkpoint during inflammation-associated cancer. STAT3 expression and activation was genetically
manipulated in the intestinal epithelium and was found to modulate
colitis-associated tumorigenesis.
33. Roessner A, Kuester D, Malfertheiner P, Schneider-Stock R:
Oxidative stress in ulcerative colitis-associated
carcinogenesis. Pathol Res Pract 2008, 204:511-524.
34. Seril DN, Liao J, Yang GY: Colorectal carcinoma development in
inducible nitric oxide synthase-deficient mice with dextran
sulfate sodium-induced ulcerative colitis. Mol Carcinog 2007,
46:341-353.
35. Seril DN, Liao J, West AB, Yang GY: High-iron diet: foe or feat in
ulcerative colitis and ulcerative colitis-associated
carcinogenesis. J Clin Gastroenterol 2006, 40:391-397.
36. Becker C, Fantini MC, Schramm C, Lehr HA, Wirtz S, Nikolaev A,
Burg J, Strand S, Kiesslich R, Huber S et al.: TGF-beta
suppresses tumor progression in colon cancer by inhibition of
IL-6 trans-signaling. Immunity 2004, 21:491-501.
29. Bromberg J, Wang TC: Inflammation and cancer: IL-6, STAT3
complete the link. Cancer Cell 2009, 15:79-80.
37. Fantini MC, Pallone F: Cytokines: from gut inflammation to
colorectal cancer. Curr Drug Targets 2008, 9:375-380.
30. Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ,
Kagnoff MF, Karin M: IKKbeta links inflammation and
tumorigenesis in a mouse model of colitis-associated cancer.
Cell 2004, 118:285-296.
38. Sturlan S, Oberhuber G, Beinhauer BG, Tichy B, Kappel S, Wang J,
Rogy MA: Interleukin-10-deficient mice and inflammatory
bowel disease associated cancer development.
Carcinogenesis 2001, 22:665-671.
Current Opinion in Pharmacology 2009, 9:1–6
Please cite this article in press as: Kraus S, Arber N. Inflammation and colorectal cancer, Curr Opin Pharmacol (2009), doi:10.1016/j.coph.2009.06.006
www.sciencedirect.com