Inflamatie Si Cancer

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Biochem. J. (2008) 410, 225–235 (Printed in Great Britain) doi:10.

1042/BJ20071341 225

REVIEW ARTICLE
Genetic aspects of inflammation and cancer
Georgina L. HOLD and Emad M. EL-OMAR1
Department of Medicine and Therapeutics, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, U.K.

Chronic inflammation is involved in the pathogenesis of most the molecular and cellular inflammatory mechanisms involved is
common cancers. The aetiology of the inflammation is varied and vital for developing preventive and therapeutic strategies against
includes microbial, chemical and physical agents. The chronically cancer. The purpose of the present review is to evaluate the
inflamed milieu is awash with pro-inflammatory cytokines mechanistic pathways that underlie chronic inflammation and
and is characterized by the activation of signalling pathways cancer with particular emphasis on the role of host genetic factors
that cross-talk between inflammation and carcinogenesis. Many that increase the risk of carcinogenesis.
of the factors involved in chronic inflammation play a dual
role in the process, promoting neoplastic progression but also Key words: cancer, chronic infection, genetic polymorphism,
facilitating cancer prevention. A comprehensive understanding of inflammation.

INTRODUCTION matory cell involvement. Mast cells especially play an important


role in releasing preformed stored inflammatory mediators that
There is a significant body of epidemiological evidence showing attract migratory inflammatory cells to the site. Next, monocytes
a clear association between chronic inflammatory diseases migrate to the area, differentiate into macrophages and become
and subsequent malignant transformation. Microbially induced activated in response to local chemokine and cytokine interac-
inflammation is estimated to be associated with approx. 15 % tions. Macrophages in turn secrete a number of potent bioactive
of the global cancer burden [1]. Moreover, strong evidence is inflammatory mediators which promote tissue healing at the site of
also available to support long-term use of non-steroidal anti- injury. When considering the inflammatory environment within a
inflammatory drugs as a means of reducing risk of several cancers neoplastic site, TAMs (tumour-associated macrophages) make up
[2]. Attempts aimed at defining the role of inflammation in the largest proportion of the inflammatory cell infiltrate and their
carcinogenesis initially identified that inflammation can activate presence has a profound impact on the local micro-environment
and induce a variety of oxidant-generating enzymes including [4,5]. However, the downside is that persistent macrophage
NADPH oxidase and iNOS (inducible nitric oxide synthase), activation can result in continued tissue damage through the
which can cause DNA damage and ultimately result in tumour stimulation of local tissue remodelling, cellular proliferation and
initiation. More recently, however, it has been recognized that angiogenesis, which helps to potentiate neoplastic progression [6].
cancer development on the back of inflammation may be driven
by inflammatory cells and a variety of mediators which together
establish an inflammatory micro-environment.
ROS (REACTIVE OXYGEN SPECIES)
It is well-established that free-radical-mediated DNA damage
ACUTE OR CHRONIC INFLAMMATION WITHIN THE TUMOUR
occurs in cancer tissue, with overexpression of iNOS, an enzyme
ENVIRONMENT
catalysing nitric oxide production, found in several cancers with
Chronic inflammation may progress from acute inflammation if known inflammatory involvement [7–11]. Leucocytes including
the injurious agent persists, but more often than not, the inflam- neutrophils, dendritic cells, mast cells and lymphocytes are all
matory response is chronic from the outset. The most compelling capable of producing a variety of ROS including nitric oxide,
examples of chronic-inflammation-induced carcinogenesis are superoxide anions and hydrogen peroxide. In addition to ROS,
seen within the gastrointestinal tract including inflammatory- RNS (reactive nitrogen species), such as peroxynitrites and
bowel-disease-induced colon carcinogenesis and also Helicobac- nitrogen oxides, are also formed and can react to generate
ter-pylori-induced gastric cancer [3]. However, there are numer- mutagenic adducts [12–15]. To date, more than 100 products have
ous other examples throughout the body (Table 1). In contrast been identified which cause oxidation of DNA. Nitric oxide and
with the largely vascular changes of acute inflammation, chronic its products may exert oncogenic effects via several mechanisms
inflammation is characterized by infiltration of damaged tissue by including direct DNA (single- or double-stranded DNA breaks,
leucocytes. Initially neutrophils and tissue mast cells are recruited deoxynucleotide or deoxyribose modifications and DNA cross-
as part of a multifactorial mechanism which co-ordinates inflam- links) and also protein damage. This can result in altered

Abbreviations used: CI, confidence interval; COX, cyclo-oxygenase; ECM, extracellular matrix; HCV, hepatitis C virus; HIF-1, hypoxia-inducible factor-1;
IFN, interferon; IκB, inhibitor of nuclear factor κB; IL, interleukin; iNOS, inducible nitric oxide synthase; LPS, lipopolysaccharide; MCP, macrophage
chemotactic protein; MIF, migration inhibitory factor; MMP, matrix metalloproteinase; NF-κB, nuclear factor κB; OR, odds ratio; PGE2 , prostaglandin E2 ;
PPAR, peroxisome-proliferator-activated receptor; PRR, pattern-recognition receptor; ROS, reactive oxygen species; SNP, single nucleotide polymorphism;
Th-1, T helper-1; TLR, Toll-like receptor; TNF-α, tumour necrosis factor α; VEGF, vascular endothelial growth factor.
1
To whom correspondence should be addressed (email e.el-omar@abdn.ac.uk).


c The Authors Journal compilation 
c 2008 Biochemical Society
226 G. L. Hold and E. M. El-Omar

Table 1 Cancers associated with infectious organisms ROS, tumour survival and metastasis
It has been suggested that when subjected to oxidative stress
Infectious organism Inflammation/cancer link
such as the effects of ROS, certain cancer cells exhibit decreased
Associations with identified organisms
attachment to the basal lamina meaning that they are more
Viral likely to detach and possibly metastasize. ROS can regulate
Hepatitis B (HBV) Hepatitis/hepatocellular carcinoma cellular adhesion by modulating integrin expression, they have
Hepatitis C (HCV) Hepatitis/hepatocellular carcinoma also been shown to suppress anoikis [apoptosis which is induced
Epstein–Barr virus (EBV) Nasopharyngeal carcinoma by anchorage-dependent cells detaching from their surrounding
Malignant lymphoma ECM (extracellular matrix)]. The degradation of the ECM
Human herpes 8 (HH8) Kaposi’s sarcoma
Squamous cell carcinoma
involves MMPs (matrix metalloproteinases), with ROS including
Non-Hodgkin’s lymphoma hydrogen peroxide, nitric oxide and iNOS implicated in the
Human immunodeficiency virus (HIV) Kaposi’s sarcoma increased expression of several MMPs and also inactivation of
Squamous cell carcinoma tissue inhibitors of metalloproteinases [27–30]. It is possible
Non-Hodgkin’s lymphoma that this effect on MMP expression may be due to activation
Human T-cell leukaemia virus type-1 (HTLV-1) T-cell lymphoma of Ras, or direct action by MAPK (mitogen-activated protein
Leukaemia
Human papillomavirus (HPV) Penile carcinoma
kinase) family members including ERK1/2 (extracellular-signal-
Anogenital carcinoma regulated kinase 1/2), p38 or JNK (c-Jun N-terminal kinase). In
Cervical carcinoma vitro experiments have shown that induction of various MMPs
Ovarian carcinoma occurs in response to expression of the Ras oncogene [31]. Ras
Cytomegalovirus (CMV) Gastric carcinoma can be activated by ROS via oxidative modification, which leads to
Bacterial the inhibition of GDP/GTP exchange [32]. Other redox-sensitive
Helicobacter pylori Gastritis/gastric carcinoma
Chlamydia trachomatis Cervical carcinoma
transcription factors such as NF-κB (nuclear factor κB) have also
Parasitic been implicated in MMP gene regulation [27].
Chinese liver fluke infestation Cholangiocarcinoma
Schistosomiasis Chronic cystitis/bladder carcinoma HIF-1 (HYPOXIA-INDUCIBLE FACTOR-1)
Liver fluke (Opisthorchis viverrini ) Cholangiosarcoma/colon carcinoma
Associations with unidentified infectious organisms Aside from elevated ROS, the reduction in physiological tissue
Inflammatory bowel disease/colorectal oxygen tension (hypoxia), which occurs during tumour initiation,
carcinoma
can also regulate cancer cell proliferation. Since angiogenesis
Chronic cholecystitis/gall
bladder carcinoma within a tumour can be both erratic and also sub-optimal
Osteomyelitis/skin carcinoma in once established, tumours often become hypoxic. HIF-1, a
draining sinuses heterodimeric transcription factor comprising HIF-1α and HIF-
1β, is a mediator of oxygen homoeostasis which, under hypoxic
conditions, binds to the promoter of hypoxia-regulated genes
transcription or signal transduction induction, genomic instability and activates a range of hypoxia-responsive molecules including
and also replication errors [16,17]. Other mechanisms include iNOS and VEGF (vascular endothelial growth factor), a potent
inhibition of apoptosis, mutation of DNA and cellular repair angiogenic factor that is important in tumour growth and
functions such as p53 and also via promotion of angiogenesis metastasis [14,33,34]. As such it is considered that the presence
[9,11]. There is also evidence of mitochondrial oxidative DNA of HIF-1α plays an important role, in particular in early
damage within the carcinogenic process, with mutations and tumorigenesis in endocrine cancers [35]. Hypoxia-induced NF-
altered expression in mitochondrial genes encoding complexes κB activation is dependent on the presence of HIF-1α, the oxygen-
I, III, IV and V having been identified in several human cancers sensitive component of the HIF-1 molecule [36,37]. HIF-1α is
[17a]. also activated by pro-inflammatory cytokines, including TNF-α
Most of the DNA-damaging effects of ROS are non-specific; (tumour necrosis factor α) and IL (interleukin)-1β, in an NF-
however, studies have revealed that, in addition to inducing κB dependent manner [38,39]. COX-2 (cyclo-oxygenase-2) also
these actions, ROS can specifically activate certain intracellular mediates IL-1β-induced HIF-1α expression through production
signalling cascades and thus contribute to tumour development of PGE2 (prostaglandin E2 ) [38]. Cancer cells have adapted these
and metastasis through the regulation of cellular functions such as pathways effectively, allowing tumours to survive and even grow
proliferation, death and motility. Growing evidence suggests that under adverse hypoxic conditions. This adaptation of tumour
ROS production is tightly regulated and their downstream targets cells to hypoxia contributes to the malignant phenotype and
are quite specific [18]. It is also known that different cellular to aggressive tumour progression, with the presence of tumour
responses are affected by specific ROS. ROS are divided into hypoxia resulting in poorer prognosis at diagnosis [40,41].
free oxygen radicals and non-radical ROS. Free radicals contain
one or more unpaired electrons and include superoxide and nitric
COX-2
oxide. Non-radical ROS include hydrogen peroxide. Hydrogen
peroxide, which is generated by numerous intracellular reactions, Another inducible enzyme with carcinogenic properties that is
exerts most of its mutagenic effects via hydroxyl ions which active within inflamed and malignant tissues is COX-2. COX-2
can activate oncogenes including K-ras [19]. In contrast, nitric is overexpressed in various cancers and, as COX-2 protein
oxide production is more specifically involved with tumour-cell accumulates, it catalyses the formation of prostaglandins (most
killing and activation of the proto-oncogene p21 and the tumour- notably PGE2 within the tumour environment) and reactive by-
suppressor gene p53. These effects are thought to be caused as a products which in turn accelerate neoplastic progression [42].
consequence of overproduction of nitric oxide [20]. Nitric oxide is This includes inhibition of apoptosis, modulation of cellular
also associated with the generation of carcinogenic p53 mutations adhesion and motility, promotion of angiogenesis and metastasis,
which have been shown to occur in approx. 50 % of cancers [21– and immunosuppression [43–47]. Other by-products of the
26]. COX-2 pathway, such as malondialdehyde production, are known


c The Authors Journal compilation 
c 2008 Biochemical Society
Genetic aspects of inflammation and cancer 227

Table 2 Human TLRs, their specific ligands and cell expression patterns
DC, dendritic cell; gp96, heat-shock protein gp96; Hsp, heat-shock protein; HMGB1, high mobility group box protein-1; NK, natural killer; PMN, polymorphonuclear; RSV, respiratory syncytial virus;
–, currently unknown. Endogenous ligands present during tissue injury including chronic inflammation are shown in bold-type.

Receptor Exogenous ligand Endogenous ligand Cell type

TLR1 Triacylated lipoprotein – Ubiquitous


TLR2 Peptidoglycans, triacylated/diacylated lipoprotein Hsp60, Hsp70, Gp96, HMGB1 PMN leucocytes, DCs and monocytes
TLR3 Double-stranded RNA mRNA DCs and NK cells
TLR4 Bacterial LPS, viral fusion protein (RSV), envelope Hsp60, Hsp70, Gp96, fibrinogen, fibronectin, Variety of cell types including monocytes,
protein, paclitaxel, mannan heparan sulfate, soluble hyaluronan, macrophages, endothelial cells and DCs
β-defensin 2, HMGB1
TLR5 Flagellin – Monocytes, immature DCs, epithelial cells, NK cells
and T-cells
TLR6 Diacylated lipoprotein – B-cells, monocytes and NK cells
TLR7 Viral single-stranded RNA mRNA B-cells and plasmacytoid precursor DCs
TLR8 Viral single-stranded RNA mRNA B-cells, monocytes and NK cells
TLR9 DNA with non-methylated CpG islands Autoimmune–IgG complex Plasmacytoid precursor DCs, B-cells, macrophages,
NK cells and microglial cells
TLR10 – – B-cells and plasmacytoid precursor DCs

to form DNA adducts which result in mutations that also inflammation through activation of receptors that recognize
play a role in neoplastic initiation [47]. Upon binding to cell- PAMPs (pathogen-associated molecular patterns), which include
surface receptors, PGE2 not only activates downstream G-coupled LPS, peptidoglycan and also viruses and nucleic acids. The
proteins but also indirectly activates other pathways which best described of these PRRs (pattern-recognition receptors)
are becoming increasingly implicated in cancer progression, are TLRs (Toll-like receptors). TLRs play a key role in the
including the Wnt and PPAR (peroxisome-proliferator-activated inflammatory response against invading micro-organisms. TLRs
receptor) signalling pathways [47–49]. In particular it has been are membrane-bound receptors that are expressed on a large
demonstrated that PGE2 increases phosphorylation of glycogen number of cell types and recognize highly conserved components
synthase kinase-3β which prevents β-catenin degradation and of bacteria, viruses and parasites (Table 2). TLRs also recognize
results in increased β-catenin/T-cell-factor-mediated transcription various endogenous ligands, several of which are present within
which activates various Wnt target genes involved in early the inflammatory environment (Table 2). The engagement of
neoplasia including c-myc, c-jun, cyclin D1 and PPARδ [50]. PRRs activates numerous signal transduction pathways that target
There is strong epidemiological evidence implicating COX-2 several transcription factors which regulate innate and adaptive
in the pathogenesis of a number of epithelial malignancies. immune responses [61,62]. These responses in turn activate other
Inhibitors of the enzyme are associated with a reduction in risk receptors that hone and amplify the inflammatory response, with
of several cancers including gastric, cervical, lung, colon and the central role in this process being played by the NF- κB pathway
prostate [51–53], with a reduction of up to 50 % in the morbidity (Figure 1). However, depending on the cell type in which it acts,
and mortality of colorectal cancer [2,44,54–56]. Among the most NF-κB can either facilitate or inhibit cancer progression [63].
potent inducers of COX-2 are the key pro-inflammatory cytokines The NF-κB transcription factor family was discovered in
IL-1α, IL-1β and TNF-α, as well as LPS (lipopolysaccharide), 1986 by Baltimore and co-workers and has since been shown
growth factors and oncogenes [57]. It has been reported that to be ubiquitously expressed in all human cell types [64,65].
human gastric tumours with p53 missense mutations exhibit NF-κB is a hetero- or homo-dimer consisting of five subunits
higher levels of COX-2 expression compared with tumours of the Rel family of polypeptides comprising RelA (p65), C-
without p53 mutations suggesting that p53 mutations may down- Rel, RelB, p50/p105/NF-κB1 and p52/p100/NF-κB2; however,
regulate COX-2 expression [58]. NF-κB mainly exists in the form of the heterodimer p65/p50.
Until recently the role of COX in tumorigenesis has focused Prior to activation, most NF-κB molecules are retained in the
almost exclusively on the inducible form COX-2. However, cytoplasm, bound to one of the IκB (inhibitor of NF-κB) proteins.
there is now emerging evidence that COX-1, the constitutively Upon stimulation, the IKK (IκB kinase) complex is activated
expressed form of COX, is also important, with increased COX-1 which phosphorylates NF-κB-bound IκB and targets them for
expression shown in cancers of the female reproductive tract polyubiquitination and ultimately degradation. This allows NF-
including ovarian and cervical cancers. Up-regulation of COX-1, κB to enter the nucleus and bind to κB-regulatory elements
but not COX-2, has been demonstrated in ovarian cancer, and co-ordinate the transcriptional activation of many immune
with COX-1-derived PGE2 promoting angiogenesis through response genes [66–68]. This is known as the classical NF-
production of VEGF [59,60]. κB signalling pathway and is triggered in response to pro-
inflammatory cytokines and micro-organisms. Once activated,
NF-κB regulates expression of over 200 genes, including genes
INFECTION, INFLAMMATION AND CANCER: ROLE OF NF-κB
encoding cell adhesion molecules and immune response genes in-
Chronic inflammation caused by persistent infection with a micro- cluding cytokines, and cell proliferation. NF-κB also regulates
organism is a major driving force in tumour development. Classic expression of NF-κB members and also the IκB proteins.
examples among others include H.-pylori-associated gastritis and There are other pathways of NF-κB activation which are less
gastric cancer, inflammatory bowel disease-associated colorectal well described. These ‘alternative’ pathways are not directly
cancer, Epstein–Barr virus and nasopharyngeal carcinoma activated as part of an innate immune response, and are triggered
(Table 1). Unless micro-organisms carry their own oncogenes, by only a limited number of stimuli. These other pathways are
toxins or growth factors, they affect the host and trigger involved in generation of secondary lymphoid organs as well as


c The Authors Journal compilation 
c 2008 Biochemical Society
228 G. L. Hold and E. M. El-Omar

Figure 1 Role of NF-κB in the inflammatory process


Ap-1, activator protein 1; ICAM-1, intercellular adhesion molecule 1; IKK, IκB kinase; IL-1R, IL-1 receptor; MAPK, mitogen-activated protein kinase; NIK, Nck-interacting kinase; TNFR, TNF receptor;
TRAF, TNFR-associated factor.

B-cell maturation and survival and are thought to be important cells which is the basis for resistance to anti-cancer chemotherapy
in B-cell lymphoma and skin carcinogenesis [69–71]. [92].
Evidence supporting the role of NF-κB in the initiation and
promotion of malignancy comes from the fact that constitutive
expression of NF-κB has been identified in a number of AUTOPHAGY AND CANCER
malignancies including hepatocellular carcinoma and breast and
colorectal cancer [72–85]. NF-κB activation in at least two Defects in apoptosis which promote tumour growth and treatment
cell types is crucial for cancer development and progression. resistance are common during tumorigenesis and have been
First, cells that are recruited to the tumour environment and well defined over the years. However, the importance of other
produce cytokines, growth factors and proteases which support forms of cell death such as autophagy and necrosis has been
cancer development and progression. Production of several less well-understood and has only recently been linked with the
pro-inflammatory cytokines, including TNF-α, IL-1 and IL-6, carcinogenic process. Various cancers including breast, ovarian
chemokines such as IL-8, growth factors including VEGF and and prostate cancers have all been shown to display allelic loss
GRO-α (growth-related oncogene α), MMPs, adhesion molecules of an essential autophagy gene beclin 1, which suggests that
and also anti-apoptotic proteins including c-FLIP [cellular FLICE autophagy may well play a role in tumour suppression [93–95].
(Fas-associated death domain-like IL-1β-converting enzyme) Autophagy, or type II programmed cell death is a highly regulated
inhibitory protein], Bcl-2 and p53, by these recruited cells are catabolic process in which organelles and cytoplasm are engulfed
known to be dependent on NF-κB activation via the classical and targeted to lysosomes for degradation [96,97]. This is in
pathway [57,86,87]. However, once activated, these cells can contrast with apoptosis, which is termed type I programmed cell
also promote ROS production which induces DNA damage in a death. Autophagy is an adaptive response to nutrient deprivation,
second cell type, cells that are programmed to undergo malignant allowing cells to persist for prolonged periods under suboptimal
transformation [88]. NF-κB also activates COX-2 expression, nutrient conditions including during carcinogenesis where tumour
which is responsible for the induction of HIF-1α, which ultimately growth frequently outstrips nutrient and oxygen supply, with
controls VEGF gene expression [57,89,90]. The up-regulated autophagy known to localize to hypoxic tumour regions [97,98].
expression of cytokines and growth factors promotes cancer cell However, there is also evidence that autophagy, when allowed
proliferation both directly and indirectly by increasing NF-κB- to proceed to completion, is a means of achieving cell death
mediated angiogenesis, tumour invasion and metastasis, with anti- [98,99]. Autophagy also plays a crucial role in removal of
apoptotic proteins protecting against apoptosis and immune attack damaged or surplus organelles. This removal limits exposure
[91]. NF-κB activation also leads to a suppression of autophagy of cellular DNA to free radical damage which ultimately decreases
which is an alternative cell death pathway that is called in to play basal DNA mutation rates, inflammation and suppresses
when apoptosis is inactivated. The role of autophagy in cancer oncogenesis [100,101]. These two mechanisms of the role of
will be discussed below. Furthermore, NF-κB contributes to drug autophagy in carcinogenic development would appear to be
resistance through multi-drug resistance-1 expression in cancer paradoxical. However, it is thought that during carcinogenesis,


c The Authors Journal compilation 
c 2008 Biochemical Society
Genetic aspects of inflammation and cancer 229

Figure 2 Role of cell death pathways in tumorigenesis

where normal apoptotic function is decreased, abrogation signalling pathways including induction of iNOS and also COX-2.
of autophagy creates reduced metabolic stress tolerance and Cytokines also induce pro-cancerous pathways and directly
cell viability which ultimately activates necrotic cell death influence tumour-suppressor function and oncogene induction.
and increases the inflammatory load of the tumour [96] Therefore cytokine signalling is thought to contribute to the
(Figure 2). Moreover several oncoproteins including c-Myc, tumour environment via two mechanisms: first, stimulation of cell
PI3K (phosphoinositide 3-kinase) and Ras are potent inhibitors growth and differentiation, and, secondly, inhibition of apoptosis
of autophagy, whereas certain tumour suppressors including of damaged cells [111]. Key pro-inflammatory cytokines include
p53 actually activate autophagy [102–105]. Consistent with the IL-1, -6, -12 and -18, TNF-α and macrophage MIF (migration in-
observation that autophagy is reduced in tumour progression; it hibitory factor) [23]. Anti-inflammatory cytokines include IL-4
has been clearly demonstrated to be induced by several anti-cancer and -10, IFN (interferon)-α and -β. MIF and IL-6 are both
drugs and is therefore a target for cancer therapy. The growing known to ameliorate p53 function which favours cell survival.
evidence identifying the role of autophagy within carcinogenic IL-6 also induces other anti-apoptotic genes including Bcl-2 and
progression cannot now be ignored; however, several important Bcl-XL , with the role of IL-6 becoming increasingly apparent
questions remain to be answered. These include defining what within colon carcinoma progression [112–115]. Cytokines also
benefits a tumour derives from suppressing autophagy and also affect cell death and cell cycle pathways, with IL-2 and TNF-α
identifying the mechanisms by which cancer treatments induce able to induce apoptosis in colon cancer cells [116]. IL-10 is
autophagy. secreted by tumour cells as well as macrophages, and among other
effects, it inhibits cytotoxic T-cells and thus aids in suppressing
the immune response against the tumour [5]. Quite often it is the
CYTOKINES
profile of cytokines existing at an inflammatory site which is
It is known that cancer cells are capable of attracting different pivotal to defining outcome. For example, TNF-α, which is pro-
cell types into the tumour micro-environment through secretion duced mainly by macrophages but also by tumour cells, is
of extracellular proteases, pro-angiogenic factors and cytokines. associated with tissue destruction and plays a role in destroying
Cytokines are small molecules that can either inhibit or propagate tumour blood supply. However, when chronically produced, it can
inflammation and activate or deactivate cancer genes and their act as a tumour promoter by contributing to tissue remodelling
pathways. Several examples exist to show how important and stromal development [4,117].
cytokines are in the inflammatory neoplastic environment, with Chemokines, which comprise the largest family of cytokines,
various animal knockout models showing predisposition of are characterized by their ability to induce migration and
knockout animals to cancer development and also a growing activation of leucocytes to specific sites. This includes tumour
body of evidence showing that numerous cytokine polymorphisms stroma and the CC chemokine MCP (macrophage chemotactic
are associated with increased risk of inflammatory diseases and protein)-1, which has been shown to be a major determinant of
cancer [106–110]. Production of cytokines is induced via the monocyte/macrophage infiltration in tumours. Tumour epithelial
classical NF-κB pathway; with many cytokines directly affecting areas have also been found to express MCP-1, whereas additional


c The Authors Journal compilation 
c 2008 Biochemical Society
230 G. L. Hold and E. M. El-Omar

chemokines such as MIP (macrophage inflammatory protein)- dominant one with onset of gastric atrophy (gastric cancer
1β and RANTES (regulated upon activation, normal T-cell phenotype) [121]. Thus it was clear that an endogenous agent
expressed and secreted) may be detected in the stroma and regu- that was up-regulated in the presence of H. pylori, has a profound
late the infiltrate of other inflammatory cells including T-cells. pro-inflammatory effect and was also an acid inhibitor would be
Furthermore, chemokines may stimulate cells to release the most relevant host genetic factor to be studied. IL-1β fitted
proteolytic enzymes, aiding the digestion of the ECM and this profile perfectly, for not only is it one of the earliest and most
providing a path for further inflammatory cell migration, tumour important pro-inflammatory cytokines in the context of H. pylori
growth and metastasis. However, it is important to appreciate that infection, but also it is the most powerful acid inhibitor known
in many preneoplastic conditions an inflammatory cell infiltrate [122]. We have shown that pro-inflammatory IL-1 gene cluster
is already well-established and drives pro-tumour effects. polymorphisms (IL-1B encoding IL-1β and IL-1RN encoding
its naturally occurring receptor antagonist) increase the risk of
gastric cancer and its precursors in the presence of H. pylori
THE ABILITY OF HOST GENETICS TO MODIFY CANCER RISK
[108]. Individuals with the IL-1B − 31*C (rs1143627) or − 511*T
Genetic polymorphisms have emerged in recent years as important (rs16944) and IL-1RN*2/*2 genotypes are at increased risk of
determinants of disease susceptibility and severity [118]. The developing hypochlorhydria and gastric atrophy in response to H.
completion of the human genome project has opened up the op- pylori infection. This risk also extends to gastric cancer itself with
portunity to dissect complex human diseases including cancers a 2–3-fold increased risk of malignancy compared with subjects
and to identify and understand the role of genetic polymorphisms. who have the less pro-inflammatory genotypes [108,109].
Although linkage analysis studies are suitable for pursuing rare Furthermore, the pro-inflammatory IL-1 genotypes increased
high-risk alleles in conditions that have a hereditary basis, the risk of both intestinal and diffuse types of gastric cancer
population-based association studies are much more useful for but the risk was restricted to the non-cardia subsite. Indeed,
examining genes with a role in more common multifactorial the IL-1 markers had no effect on the risk of cardia gastric
diseases that have a strong environmental component [119]. adenocarcinoma, oesophageal adenocarcinoma or oesophageal
These association studies often estimate the risk of developing a squamous cell carcinoma [109]. The latter findings are entirely
certain disease in carriers and non-carriers of a particular genetic in keeping with the proposed mechanism for the effect of these
polymorphism. The overwhelming majority of polymorphisms polymorphisms in gastric cancer, namely reduction of gastric acid
studied are SNPs (single nucleotide polymorphisms) that occur secretion. Thus a high IL-1β genotype increases the risk of non-
with a frequency of >1 % in the normal population (in con- cardia gastric cancer, a disease characterized by hypochlorhydria,
trast with mutations that occur with a frequency of <1 %). It whereas it has no effect on cancers associated with high acid
is estimated that up to 10 million SNPs are probably present in exposure such as oesophageal adenocarcinoma and some cardia
the human genome although not all have thus far been identified. cancers.
Naturally, most of these SNPs do not occur in coding sequences The association between IL-1 gene cluster polymorphisms
and even those that do are not associated with any alteration and gastric cancer and its precursors has been confirmed
in the amino acid sequence and are therefore of no functional independently by other groups covering Caucasian, Asian and
consequence. A more useful definition of the variability of these Hispanic populations [123–129]. Machado et al. [125] were the
SNPs involves haplotype analysis. Haplotypes are a combination first to confirm the association between IL-1 markers and gastric
of alleles at different markers along the same chromosome that cancer in Caucasians and reported similar ORs (odds ratios)
are inherited as a unit. The HapMap project was set up to to those reported by our group [108]. Furthermore, the same
address this (http://www.hapmap.org) and is a valuable tool that group subsequently reported on the combined effects of pro-
will facilitate the study of genetic polymorphisms relevant to inflammatory IL-1 genotypes and H. pylori bacterial virulence
human health and disease. Other types of genetic variation include factors (cagA positive, VacA s1 and VacA m1) [123]. They showed
deletion and insertion polymorphisms and microsatellite-repeat that for each combination of bacterial/host genotype, the odds of
polymorphisms. having gastric carcinoma were greatest in those with both bacterial
Genetic polymorphisms directly influence inter-individual and host high-risk genotypes. This highlights the important
variation in the magnitude of an inflammatory response, and interaction between host and bacterium in the pathogenesis of
this clearly contributes to the ultimate clinical outcome of an gastric cancer. The role of IL-1 polymorphisms has also been
individual. An excellent example of this from our own work investigated in other cancers, with associations shown with
[120] is the role of genetic polymorphisms in the pathogenesis hepatitis-C-induced hepatocellular carcinoma, colorectal and also
of H.-pylori-induced gastric cancer. H. pylori infection elicits breast cancer [130–133].
a powerful humoral and cell-mediated immune response that Soon after the IL-1 gene cluster polymorphisms were identified
causes inflammation in the gastric mucosa. The mediators of as risk factors for gastric cancer, the pro-inflammatory genotypes
this inflammatory response belong to the two main arms of the of TNF-A and IL-10 were reported as independent additional risk
immune system, namely adaptive and innate immunity. We factors for non-cardia gastric cancer [109]. TNF-α is another
speculated that the most relevant candidate genes would be powerful pro-inflammatory cytokine that is produced in the
ones whose products were involved in handling the H. pylori gastric mucosa in response to H. pylori infection. Like IL-1β,
attack and ones that mediated the resulting inflammation. it has an acid inhibitory effect, albeit much weaker [134]. The
Because such a list of candidate genes would be prohibitively TNF-A − 308 G>A (rs1800629) polymorphism is known to be
extensive, our initial search focused on genes that were most involved in a number of inflammatory conditions. Carriage of
relevant to gastric physiology and, in particular, gastric acid the pro-inflammatory A allele increased the OR for non-cardia
secretion. H.-pylori-induced gastritis is associated with three gastric cancer to 2.2 [95 % CI (confidence intervals), 1.4–3.7].
main phenotypes that correlate closely with clinical outcome: The role of the TNF-A − 308 G>A polymorphism in gastric
duodenal ulcer phenotype, benign phenotype and gastric cancer cancer was independently confirmed by a study from Machado
phenotype [120]. Studies have shown that inhibition of gastric et al. [124], with its role in other cancers also now confirmed
acid pharmacologically can lead to a shift from an antrum- [135,136]. IL-10 is an anti-inflammatory cytokine that down-
predominant pattern (duodenal ulcer phenotype) to a corpus-pre- regulates IL-1β, TNF-α, IFN-γ and other pro-inflammatory


c The Authors Journal compilation 
c 2008 Biochemical Society
Genetic aspects of inflammation and cancer 231

cytokines. Relative deficiency of IL-10 may result in a Th- This is due to inadequate production of IL-10-secreting type 1
1 (T helper-1)-driven hyperinflammatory response to H. pylori regulatory cells [150].
with greater damage to the gastric mucosa. We have reported We hypothesized that the TLR4+ 896A>G polymorphism
that homozygosity for the low-IL-10 ATA haplotype [based on would be associated with an exaggerated and destructive
three promoter polymorphisms at positions − 592 (rs1800872), chronic inflammatory phenotype in H. pylori-infected subjects.
− 819 (rs1800871) and − 1082 (rs1800896)] increased the risk This phenotype would be characterized by gastric atrophy
of non-cardia gastric cancer with an OR of 2.5 (95 % CI, 1.1– and hypochlorhydria, the hallmarks of a subsequent increased
5.7), with the importance of the IL-10 haplotype confirmed risk of gastric cancer. We further hypothesized that the same
now in other cancers ([137] and extensively reviewed in [138]). polymorphism might increase the risk of gastric cancer itself.
We have studied the effect of having an increasing number We proceeded to test the effect of this polymorphism on the
of pro-inflammatory genotypes (IL-1B − 511*T, IL-1RN*2*2, H.-pylori-induced gastric phenotype and the risk of developing
TNF-A − 308*A and IL-10 ATA/ATA) on the risk of non-cardia pre-malignant and malignant outcomes. We assessed associ-
gastric cancer [109]. The risk increased progressively so that ations with pre-malignant gastric changes in relatives of gastric
by the time three or four of these polymorphisms were pre- cancer patients, including those with hypochlorhydria and
sent, the OR for gastric cancer was increased to 27-fold [109]. gastric atrophy. We also genotyped two independent Caucasian
The fact that H. pylori is a pre-requisite for the association population-based case-control studies of upper gastrointestinal
of these polymorphisms with malignancy demonstrates that in tract cancer. TLR4+ 896G carriers had a 7.7-fold (95 % CI,
this situation, inflammation is indeed driving carcinogenesis. 1.6–37.6) increased OR for hypochlorhydria; the polymorphism
Another important cytokine that is key in the pathogenesis of was not associated with gastric acid output in the absence of
H.-pylori-induced diseases is IL-8. This chemokine belongs to H. pylori infection. Carriers also had significantly more severe
the CXC family and is a potent chemoattractant for neutrophils gastric atrophy and inflammation [151]. Also 16 % of gastric
and lymphocytes. It also has effects on cell proliferation, cancer patients in the initial study and 15 % of the non-cardia
migration and tumour angiogenesis. The gene has a well- gastric cancer patients in the replication study had one or two
established promoter polymorphism at position − 251 [IL-8 − 251 TLR4 variant alleles compared with 8 % of both control popul-
A>T (rs4073)]. The A allele is associated with increased ations (combined OR, 2.4; 95 % CI, 1.6–3.4) [151]. In contrast,
production of IL-8 in H.-pylori-infected gastric mucosa [139]. the prevalence of TLR4+ 896G was not significantly increased in
It was also found to increase the risk of severe inflammation oesophageal squamous cell (2 %; OR, 0.4) or adenocarcinoma
and precancerous gastric abnormalities in Caucasian and Asian (9 %; OR, 0.8) or gastric cardia cancer (11 %; OR, 1.2).
populations [140]. However, the same polymorphism was The association of the TLR4+ 896A>G polymorphism with
found to increase risk of gastric cancer only in some Asian both gastric cancer and its precursor lesions implies that it is
populations [141–143] with no apparent effect in Caucasians. relevant to the entire multistage process of gastric carcinogenesis,
It is probable that other pro-inflammatory cytokine gene poly- which starts with H. pylori colonization of the gastric mucosa.
morphisms will be relevant to cancer initiation and progression. Subjects with this polymorphism have an increased risk of severe
This exciting field has expanded greatly over the last few years inflammation and, subsequently, development of hypochlorhydria
and the search is now on for the full complement of risk genotypes and gastric atrophy, which are regarded as the most important
that dictate the likelihood of an individual developing cancer. precancerous abnormalities. This severe inflammation is initiated
by H. pylori infection but it is entirely feasible that subsequent
co-colonization of an achlorhydric stomach by a variety of other
ROLE OF POLYMORPHISMS IN INNATE IMMUNE RESPONSE GENES
bacteria may sustain and enhance the microbial inflammatory
Genetic polymorphisms of cytokines of the adaptive immune re- stimulus and continue to drive the carcinogenic process. Evidence
sponse clearly play an important role in the risk of microbially in- supporting this concept comes from the work of Sanduleanu
duced carcinomas. However, micro-organisms are initially et al. [152] who showed that pharmacological inhibition of
handled by the innate immune response and it is conceivable acid secretion was associated with a higher prevalence of non-
that functionally relevant polymorphisms in genes of this arm of H. pylori bacteria. Furthermore, the simultaneous presence
the immune system could affect the magnitude and subsequent of H. pylori and non-H. pylori bacteria was associated with a
direction of the response of the host against the infection. In markedly increased risk of atrophic gastritis, and with higher
the case of H.-pylori-induced gastric carcinoma, the majority circulating levels of IL-1β and IL-8. Supporting evidence also
of H. pylori cells do not invade the gastric mucosa but the comes from animal studies where hypochlorhydria was induced
inflammatory response against it is triggered through attachment in mice either genetically (G− /G− gastrin-deficient mice) or
of H. pylori to the gastric epithelia, with TLR4 playing an pharmacologically (administration of omeprazole). Zavros et al.
important role in recognition [144]. Arbour et al. [145] described [153] found that genetic or chemical hypochlorhydria predisposes
a functional polymorphism in the TLR4 gene (rs4986790). the stomach to bacterial overgrowth resulting in inflammation,
This A>G transition results in replacement of a conserved which was not present in the wild-type mice or those not treated
aspartic acid residue with a glycine residue at amino acid with an acid inhibitor.
299 (D299G) and alteration in the extracellular domain of the The potential mechanism by which the TLR4 polymorphism
TLR4 receptor. This renders carriers hyporesponsive to LPS increases the risk of gastric cancer and its precursors is intriguing
challenge by either disrupting transport of TLR4 to the cell and may lie in the nature of the overall response of the host
membrane or by impairing ligand binding or protein interactions to the H. pylori LPS attack. Failure to handle the invasion by
[145]. The mutation has been associated with a variety of appropriately recognizing and activating the necessary pathways
inflammatory and infectious conditions including atherosclerosis, may lead to an imbalance of pro- and anti-inflammatory
myocardial infarction, inflammatory bowel disease and septic mediators. The findings emphasize, for the first time, the
shock [146–149]. Recent work demonstrates that defective importance of innate immune response gene polymorphisms in
signalling through the TLR4 receptor ultimately leads to an outcome to neoplastic progression.
exaggerated inflammatory response with severe tissue destruction, This field of host genetic polymorphisms has expanded greatly
even though the initial immune response may be blunted [150]. over the last few years, and the search is now on for the full


c The Authors Journal compilation 
c 2008 Biochemical Society
232 G. L. Hold and E. M. El-Omar

complement of risk genotypes that dictate the likelihood of an 7 Kawanishi, S., Hiraku, Y., Pinlaor, S. and Ma, N. (2006) Oxidative and nitrative DNA
individual of developing cancer. This approach has now been damage in animals and patients with inflammatory diseases in relation to
adopted for many other cancers as described below. In Japanese inflammation-related carcinogenesis. Biol. Chem. 387, 365–372
8 Lechner, M., Lirk, P. and Rieder, J. (2005) Inducible nitric oxide synthase (iNOS) in
patients with chronic HCV (hepatitis C virus) infection, the IL-
tumor biology: the two sides of the same coin. Semin. Cancer Biol. 15, 277–289
1B − 511 T/T genotype has been associated with an increased 9 Ohshima, H. (2003) Genetic and epigenetic damage induced by reactive nitrogen
risk of progression to hepatocellular carcinoma [131]. Because species: implications in carcinogenesis. Toxicol. Lett. 140–141, 99–104
the T/T pro-inflammatory genotype is related to greater IL-1 10 Ohshima, H., Tazawa, H., Sylla, B. S. and Sawa, T. (2005) Prevention of human cancer by
production, it is feasible that the risk of malignant transformation modulation of chronic inflammatory processes. Mutat. Res. 591, 110–122
is higher. IL-1 leads to the production of PGE2 and hepatocyte 11 Jaiswal, M., LaRusso, N. F. and Gores, G. J. (2001) Nitric oxide in gastrointestinal
growth factor and has angiogenic influence via iNOS and COX-2 epithelial cell carcinogenesis: linking inflammation to oncogenesis. Am. J. Physiol.
expression. Furthermore, the degree of HCV-induced liver Gastrointest. Liver Physiol. 281, G626–G634
inflammation and fibrosis has been correlated with hepatic 12 Tamir, S., Rojas-Walker, T., Wishnok, J. S. and Tannenbaum, S. R. (1996) DNA damage
expression of Th-1 cytokines. At present, there is relatively little and genotoxicity by nitric oxide. Methods Enzymol. 269, 230–243
13 Maeda, H. and Akaike, T. (1998) Nitric oxide and oxygen radicals in infection,
information on the relationship between other gastrointestinal
inflammation, and cancer. Biochemistry 63, 854–865
malignancies and innate immune and cytokine poly- 14 Valko, M., Rhodes, C. J., Moncol, J., Izakovic, M. and Mazur, M. (2006) Free radicals,
morphisms. Some studies have addressed the influence of metals and antioxidants in oxidative stress-induced cancer. Chem. Biol. Interact. 160,
polymorphisms on cancer outcome. Barber et al. [154] found that 1–40
possession of a genotype resulting in increased IL-1 production 15 Sandhu, J. K., Privora, H. F., Wenckebach, G. and Birnboim, H. C. (2000) Neutrophils,
was associated with shortened survival in pancreatic cancer. nitric oxide synthase, and mutations in the mutatect murine tumor model. Am. J. Pathol.
Park et al. [155] investigated TNF-A and -B polymorphisms 156, 509–518
in 136 colorectal cancer patients and 325 healthy controls in 16 Marnett, L. J. (2000) Oxyradicals and DNA damage. Carcinogenesis 21, 361–370
an Asian population. Their results indicated that TNF-B*1/ 17 Cooke, M. S., Evans, M. D., Dizdaroglu, M. and Lunec, J. (2003) Oxidative DNA
damage: mechanisms, mutation, and disease. FASEB J. 17, 1195–1214
TNF-B*1 genotypes showed an increased risk for colorectal
17a Penta, J. S., Johnson, F. M., Wachsman, J. T. and Copeland, W. C. (2001) Mitochondrial
cancer. De Jong et al. [156] performed pooled analyses on 30 DNA in human malignancy. Mutat. Res. 488, 119–133
polymorphisms in 20 low-penetrance genes and identified an 18 Finkel, T. (2003) Oxidant signals and oxidative stress. Curr. Opin. Cell Biol. 15, 247–254
additional three studies investigating TNF-A polymorphisms and 19 Jackson, J. H. (1994) Potential molecular mechanisms of oxidant-induced
colorectal cancer. Associations were detected for the a2, a5 carcinogenesis. Environ. Health Perspect. 102, 155–157
and a13 TNF alleles and colorectal cancer. 20 Lander, H. M., Ogiste, J. S., Teng, K. K. and Novogrodsky, A. (1995) p21ras as a
common signaling target of reactive free radicals and cellular redox stress.
J. Biol. Chem. 270, 21195–21198
CONCLUDING REMARKS 21 Li, C. Q. and Wogan, G. N. (2005) Nitric oxide as a modulator of apoptosis. Cancer Lett.
226, 1–15
There is now substantial evidence that chronic inflammation and 22 Cui, S., Reichner, J. S., Mateo, R. B. and Albina, J. E. (1994) Activated murine
malignant development are causally linked. However, further macrophages induce apoptosis in tumor cells through nitric oxide-dependent or
studies are required to fully understand the complex relationship. -independent mechanisms. Cancer Res. 54, 2462–2467
The situation is further complicated by the recent acceptance of 23 Hofseth, L. J., Saito, S., Hussain, S. P., Espey, M. G., Miranda, K. M., Araki, Y.,
the role of chronic infection and also the contribution of host Jhappan, C., Higashimoto, Y., He, P., Linke, S. P. et al. (2003) Nitric oxide-induced
genetic polymorphisms to the risk of an individual of developing cellular stress and p53 activation in chronic inflammation. Proc. Natl. Acad. Sci. U.S.A.
malignancy. Chronic inflammation, whether caused by microbes, 100, 143–148
chemical or physical trauma, within a pre-malignant environment 24 Hussain, S. P., Amstad, P., He, P., Robles, A., Lupold, S., Kaneko, I., Ichimiya, M.,
favours neoplastic progression. However, once present, cancer Sengupta, S., Mechanic, L., Okamura, S. et al. (2004) p53-induced up-regulation of
MnSOD and GPx but not catalase increases oxidative stress and apoptosis. Cancer Res.
cells can ‘educate’ the immune infiltrate to produce the necessary 64, 2350–2356
cytokine composition to facilitate tumour growth and metastasis 25 Masuda, M., Nishino, H. and Ohshima, H. (2002) Formation of 8-nitroguanosine
as well as acquiring immune tolerance. Understanding this in cellular RNA as a biomarker of exposure to reactive nitrogen species.
feedback loop is fundamental to resolving the complex interplay Chem. Biol. Interact. 139, 187–197
between the causes of chronic inflammation and the genetic 26 Souici, A. C., Mirkovitch, J., Hausel, P., Keefer, L. K. and Felley-Bosco, E. (2000)
disposition of the host to neoplastic progression, which will aid Transition mutation in codon 248 of the p53 tumor suppressor gene induced by reactive
the development of new and more effective anti-inflammatory oxygen species and a nitric oxide-releasing compound. Carcinogenesis 21, 281–287
cancer prevention strategies in the future. 27 Nelson, K. K. and Melendez, J. A. (2004) Mitochondrial redox control of matrix
metalloproteinases. Free Radical Biol. Med. 37, 768–784
28 Kundu, N., Zhang, S. and Fulton, A. M. (1995) Sublethal oxidative stress inhibits tumor
REFERENCES cell adhesion and enhances experimental metastasis of murine mammary carcinoma.
Clin. Exp. Metastasis 13, 16–22
1 Kuper, H., Adami, H. O. and Trichopoulos, D. (2000) Infections as a major preventable 29 Gregg, D., de Carvalho, D. D. and Kovacic, H. (2004) Integrins and coagulation: a role
cause of human cancer. J. Intern. Med. 248, 171–183 for ROS/redox signaling? Antioxid. Redox Signaling 6, 757–764
2 Gupta, R. A. and DuBois, R. N. (2001) Colorectal cancer prevention and treatment by 30 Halliwell, B. (2007) Oxidative stress and cancer: have we moved forward? Biochem. J.
inhibition of cyclooxygenase-2. Nat. Rev. Cancer 1, 11–21 401, 1–11
3 Macarthur, M., Hold, G. L. and El Omar, E. M. (2004) Inflammation and cancer II: role of 31 Thomas, P., Khokha, R., Shepherd, F. A., Feld, R. and Tsao, M. S. (2000) Differential
chronic inflammation and cytokine gene polymorphisms in the pathogenesis of expression of matrix metalloproteinases and their inhibitors in non-small cell lung
gastrointestinal malignancy. Am. J. Physiol. Gastrointest. Liver Physiol. 286, cancer. J. Pathol. 190, 150–156
G515–G520 32 Lander, H. M., Hajjar, D. P., Hempstead, B. L., Mirza, U. A., Chait, B. T., Campbell, S. and
4 Balkwill, F. and Mantovani, A. (2001) Inflammation and cancer: back to Virchow? Lancet Quilliam, L. A. (1997) A molecular redox switch on p21(ras). Structural basis for the
357, 539–545 nitric oxide-p21(ras) interaction. J. Biol. Chem. 272, 4323–4326
5 Coussens, L. M. and Werb, Z. (2002) Inflammation and cancer. Nature 420, 860–867 33 Jain, R. K. (2002) Tumor angiogenesis and accessibility: role of vascular endothelial
6 Schoppmann, S. F., Birner, P., Stockl, J., Kalt, R., Ullrich, R., Caucig, C., Kriehuber, E., growth factor. Semin. Oncol. 29, 3–9
Nagy, K., Alitalo, K. and Kerjaschki, D. (2002) Tumor-associated macrophages express 34 Semenza, G. L. (2003) Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer 3, 721–732
lymphatic endothelial growth factors and are related to peritumoral lymphangiogenesis. 35 Kimbro, K. S. and Simons, J. W. (2006) Hypoxia-inducible factor-1 in human breast and
Am. J. Pathol. 161, 947–956 prostate cancer. Endocr. Relat. Cancer 13, 739–749


c The Authors Journal compilation 
c 2008 Biochemical Society
Genetic aspects of inflammation and cancer 233

36 Salceda, S. and Caro, J. (1997) Hypoxia-inducible factor 1α (HIF-1α) protein is rapidly 65 Singh, H., Sen, R., Baltimore, D. and Sharp, P. A. (1986) A nuclear factor that binds to a
degraded by the ubiquitin-proteasome system under normoxic conditions. Its conserved sequence motif in transcriptional control elements of immunoglobulin genes.
stabilization by hypoxia depends on redox-induced changes. J. Biol. Chem. 272, Nature 319, 154–158
22642–22647 66 Karin, M. (2006) Nuclear factor-κB in cancer development and progression. Nature
37 Semenza, G. L. (1999) Regulation of mammalian O2 homeostasis by hypoxia-inducible 441, 431–436
factor 1. Annu. Rev. Cell Dev. Biol. 15, 551–578 67 Karin, M. and Greten, F. R. (2005) NF-κB: linking inflammation and immunity to cancer
38 Jung, Y., Isaacs, J. S., Lee, S., Trepel, J., Liu, Z. G. and Neckers, L. (2003) development and progression. Nat. Rev. Immunol. 10, 749–759
Hypoxia-inducible factor induction by tumour necrosis factor in normoxic cells requires 68 Chun, K. S. and Surh, Y. J. (2004) Signal transduction pathways regulating
receptor-interacting protein-dependent nuclear factor κB activation. Biochem. J. 370, cyclooxygenase-2 expression: potential molecular targets for chemoprevention.
3–7 Biochem. Pharmacol. 68, 1089–1100
39 Jung, Y. J., Isaacs, J. S., Lee, S., Trepel, J. and Neckers, L. (2003) IL-1β-mediated 69 Bonizzi, G. and Karin, M. (2004) The two NF-κB activation pathways and their role in
up-regulation of HIF-1α via an NFκB/COX-2 pathway identifies HIF-1 as a critical link innate and adaptive immunity. Trends Immunol. 25, 280–288
between inflammation and oncogenesis. FASEB J. 17, 2115–2117 70 Neri, A., Chang, C. C., Lombardi, L., Salina, M., Corradini, P., Maiolo, A. T., Chaganti,
40 Harris, A. L. (2002) Hypoxia: a key regulatory factor in tumour growth. Nat. Rev. Cancer R. S. and Dalla-Favera, R. (1991) B cell lymphoma-associated chromosomal
2, 38–47 translocation involves candidate oncogene lyt-10, homologous to NF-κB p50. Cell 67,
41 Vleugel, M. M., Greijer, A. E., Shvarts, A., van der Groep, P., van Berkel, M., Aarbodem, 1075–1087
Y., van Tinteren, H., Harris, A. L., van Diest, P. J. and van der Wall, E. (2005) Differential 71 Kato, Jr, T., Delhase, M., Hoffmann, A. and Karin, M. (2003) CK2 is a C-terminal IκB
prognostic impact of hypoxia induced and diffuse HIF-1α expression in invasive breast kinase responsible for NF-κB activation during the UV response. Mol. Cell 12, 829–839
cancer. J. Clin. Pathol. 58, 172–177 72 Shishodia, S. and Aggarwal, B. B. (2004) Nuclear factor-κB: a friend or a foe in cancer?
42 Fitzpatrick, F. A. (2001) Inflammation, carcinogenesis and cancer. Int. Biochem. Pharmacol. 68, 1071–1080
Immunopharmacol. 1, 1651–1667 73 Luo, J. L., Kamata, H. and Karin, M. (2005) IKK/NF-κB signaling: balancing life and
43 Tsujii, M., Kawano, S. and DuBois, R. N. (1997) Cyclooxygenase-2 expression in human death: a new approach to cancer therapy. J. Clin. Invest. 115, 2625–2632
colon cancer cells increases metastatic potential. Proc. Natl. Acad. Sci. U.S.A. 94, 74 Aggarwal, B. B., Shishodia, S., Sandur, S. K., Pandey, M. K. and Sethi, G. (2006)
3336–3340 Inflammation and cancer: how hot is the link? Biochem. Pharmacol. 72, 1605–1621
44 Marnett, L. J. and DuBois, R. N. (2002) COX-2: a target for colon cancer prevention. 75 Okamoto, T., Sanda, T. and Asamitsu, K. (2007) NF-κB signaling and carcinogenesis.
Annu. Rev. Pharmacol. Toxicol. 42, 55–80 Curr. Pharm. Des. 13, 447–462
45 Prescott, S. M. and Fitzpatrick, F. A. (2000) Cyclooxygenase-2 and carcinogenesis. 76 Herrmann, J. L., Beham, A. W., Sarkiss, M., Chiao, P. J., Rands, M. T., Bruckheimer,
Biochim. Biophys. Acta 1470, M69–M78
E. M., Brisbay, S. and McDonnell, T. J. (1997) Bcl-2 suppresses apoptosis resulting
46 Williams, C. S., Mann, M. and DuBois, R. N. (1999) The role of cyclooxygenases in
from disruption of the NF-κB survival pathway. Exp. Cell Res. 237, 101–109
inflammation, cancer, and development. Oncogene 18, 7908–7916
77 Palayoor, S. T., Youmell, M. Y., Calderwood, S. K., Coleman, C. N. and Price, B. D.
47 Cha, Y. I. and DuBois, R. N. (2007) NSAIDs and cancer prevention: targets downstream
(1999) Constitutive activation of IκB kinase α and NF-κB in prostate cancer cells is
of COX-2. Annu. Rev. Med. 58, 239–252
inhibited by ibuprofen. Oncogene 18, 7389–7394
48 Yang, W. L. and Frucht, H. (2001) Activation of the PPAR pathway induces apoptosis and
78 Wang, W., Abbruzzese, J. L., Evans, D. B., Larry, L., Cleary, K. R. and Chiao, P. J. (1999)
COX-2 inhibition in HT-29 human colon cancer cells. Carcinogenesis 22, 1379–1383
The nuclear factor-κB RelA transcription factor is constitutively activated in human
49 Castellone, M. D., Teramoto, H., Williams, B. O., Druey, K. M. and Gutkind, J. S. (2005)
pancreatic adenocarcinoma cells. Clin. Cancer Res. 5, 119–127
Prostaglandin E2 promotes colon cancer cell growth through a Gs-axin-β-catenin
79 Dejardin, E., Deregowski, V., Chapelier, M., Jacobs, N., Gielen, J., Merville, M. P. and
signaling axis. Science 310, 1504–1510
Bours, V. (1999) Regulation of NF-κB activity by IκB-related proteins in
50 Gregorieff, A. and Clevers, H. (2005) Wnt signaling in the intestinal epithelium: from
adenocarcinoma cells. Oncogene 18, 2567–2577
endoderm to cancer. Genes Dev. 19, 877–890
80 Pikarsky, E., Porat, R. M., Stein, I., Abramovitch, R., Amit, S., Kasem, S.,
51 Dai, Y. and Wang, W. H. (2006) Non-steroidal anti-inflammatory drugs in prevention of
Gutkovich-Pyest, E., Urieli-Shoval, S., Galun, E. and Ben-Neriah, Y. (2004) NF-κB
gastric cancer. World J. Gastroenterol. 12, 2884–2889
functions as a tumour promoter in inflammation-associated cancer. Nature 431,
52 Schreinemachers, D. M. and Everson, R. B. (1994) Aspirin use and lung, colon, and
461–466
breast cancer incidence in a prospective study. Epidemiology 5, 138–146
53 Funkhouser, E. M. and Sharp, G. B. (1995) Aspirin and reduced risk of esophageal 81 Greten, F. R., Eckmann, L., Greten, T. F., Park, J. M., Li, Z. W., Egan, L. J., Kagnoff, M. F.
carcinoma. Cancer 76, 1116–1119 and Karin, M. (2004) IKKβ links inflammation and tumorigenesis in a mouse model of
54 Chan, A. T., Ogino, S. and Fuchs, C. S. (2007) Aspirin and the risk of colorectal cancer colitis-associated cancer. Cell 118, 285–296
in relation to the expression of COX-2. N. Eng. J. Med. 356, 2131–2142 82 Krappmann, D., Emmerich, F., Kordes, U., Scharschmidt, E., Dorken, B. and
55 Wang, D. and DuBois, R. N. (2007) Inflammatory mediators and nuclear receptor Scheidereit, C. (1999) Molecular mechanisms of constitutive NF-κB/Rel activation
signaling in colorectal cancer. Cell Cycle 6, 682–685 in Hodgkin/Reed-Sternberg cells. Oncogene 18, 943–953
56 Bertagnolli, M. M. (2007) Chemoprevention of colorectal cancer with cyclooxygenase-2 83 Mori, N., Fujii, M., Ikeda, S., Yamada, Y., Tomonaga, M., Ballard, D. W. and Yamamoto,
inhibitors: two steps forward, one step back. Lancet Oncol. 8, 439–443 N. (1999) Constitutive activation of NF-κB in primary adult T-cell leukemia cells. Blood
57 Karin, M., Cao, Y., Greten, F. R. and Li, Z. W. (2002) NF-κB in cancer: from innocent 93, 2360–2368
bystander to major culprit. Nat. Rev. Cancer 2, 301–310 84 Bharti, A. C., Shishodia, S., Reuben, J. M., Weber, D., Alexanian, R., Raj-Vadhan, S.,
58 Leung, W. K., To, K. F., Ng, Y. P., Lee, T. L., Lau, J. Y., Chan, F. K., Ng, E., Chung, S. C. Estrov, Z., Talpaz, M. and Aggarwal, B. B. (2004) Nuclear factor-κB and STAT3 are
and Sung, J. J. (2001) Association between cyclo-oxygenase-2 overexpression and constitutively active in CD138+ cells derived from multiple myeloma patients, and
missense p53 mutations in gastric cancer. Br. J. Cancer 84, 335–339 suppression of these transcription factors leads to apoptosis. Blood 103, 3175–3184
59 Sales, K. J., Katz, A. A., Howard, B., Soeters, R. P., Millar, R. P. and Jabbour, H. N. (2002) 85 Sanda, T., Iida, S., Ogura, H., Asamitsu, K., Murata, T., Bacon, K. B., Ueda, R. and
Cyclooxygenase-1 is up-regulated in cervical carcinomas: autocrine/paracrine Okamoto, T. (2005) Growth inhibition of multiple myeloma cells by a novel IκB kinase
regulation of cyclooxygenase-2, prostaglandin E receptors, and angiogenic factors by inhibitor. Clin. Cancer Res. 11, 1974–1982
cyclooxygenase-1. Cancer Res. 62, 424–432 86 Balkwill, F. (2004) Cancer and the chemokine network. Nat. Rev. Cancer 4, 540–550
60 Gupta, R. A., Tejada, L. V., Tong, B. J., Das, S. K., Morrow, J. D., Dey, S. K. and DuBois, 87 Li, Q., Withoff, S. and Verma, I. M. (2005) Inflammation-associated cancer: NF-κB is the
R. N. (2003) Cyclooxygenase-1 is overexpressed and promotes angiogenic growth lynchpin. Trends Immunol. 26, 318–325
factor production in ovarian cancer. Cancer Res. 63, 906–911 88 Inoue, J., Gohda, J., Akiyama, T. and Semba, K. (2007) NF-κB activation in development
61 Medzhitov, R. (2001) Toll-like receptors and innate immunity. Nat. Rev. Immunol. 1, and progression of cancer. Cancer Sci. 98, 268–274
135–145 89 Marx, J. (2004) Cancer research. Inflammation and cancer: the link grows stronger.
62 Akira, S., Takeda, K. and Kaisho, T. (2001) Toll-like receptors: critical proteins linking Science 306, 966–968
innate and acquired immunity. Nat. Immunol. 2, 675–680 90 Nakanishi, C. and Toi, M. (2005) Nuclear factor-κB inhibitors as sensitizers to
63 Karin, M. (2006) NF-κB and cancer: mechanisms and targets. Mol. Carcinogenesis 45, anticancer drugs. Nat. Rev. Cancer 5, 297–309
355–361 91 Chen, R., Alvero, A. B., Silasi, D. A. and Mor, G. (2007) Inflammation, cancer and
64 Sen, R. and Baltimore, D. (1986) Multiple nuclear factors interact with the chemoresistance: taking advantage of the toll-like receptor signaling pathway. Am. J.
immunoglobulin enhancer sequences. Cell 46, 705–716 Reprod. Immunol. 57, 93–107


c The Authors Journal compilation 
c 2008 Biochemical Society
234 G. L. Hold and E. M. El-Omar

92 Bentires-Alj, M., Barbu, V., Fillet, M., Chariot, A., Relic, B., Jacobs, N., Grielen, J., 120 Smith, M. G., Hold, G. L., Tahara, E. and El-Omar, E. M. (2006) Cellular and molecular
Merville, M. P. and Bours, V. (2003) NF-κB transcription factor induces drug resistance aspects of gastric cancer. World J. Gastroenterol. 12, 2979–2990
through MDR1 expression in cancer cells. Oncogene 22, 90–97 121 Kuipers, E. J., Lee, A., Klinkenberg-Knol, E. C. and Meuwissen, S. G. (1995) The
93 Botti, J., Djavaheri-Mergny, M., Pilatte, Y. and Codogno, P. (2006) Autophagy signaling development of atrophic gastritis: Helicobacter pylori and the effects of acid suppressive
and the cogwheels of cancer. Autophagy 2, 67–73 therapy. Aliment. Pharmacol. Ther. 9, 331–340
94 Aita, V. M., Liang, X. H., Murty, V. V., Pincus, D. L., Yu, W., Cayanis, E., Kalachikov, S., 122 El Omar, E. M. (2001) The importance of interleukin 1β in Helicobacter pylori associated
Gilliam, T. C. and Levine, B. (1999) Cloning and genomic organization of beclin 1, a disease. Gut 48, 743–747
candidate tumor suppressor gene on chromosome 17q21. Genomics 59, 59–65 123 Figueiredo, C., Machado, J. C., Pharoah, P., Seruca, R., Sousa, S., Carvalho, R.,
95 Liang, X. H., Jackson, S., Seaman, M., Brown, K., Kempkes, B., Hibshoosh, H. and capelinha, A. F., Quint, W., Caldas, C., van Doorn, L. J. et al. (2002) Helicobacter pylori
Levine, B. (1999) Induction of autophagy and inhibition of tumorigenesis by beclin 1. and interleukin 1 genotyping: an opportunity to identify high-risk individuals for gastric
Nature 402, 672–676 carcinoma. J. Natl. Cancer Inst. 94, 1680–1687
96 Jin, S. and White, E. (2007) Role of autophagy in cancer: management of metabolic 124 Machado, J. C., Figueiredo, C., Canedo, P., Pharoah, P., Carvalho, R., Nabais, S.,
stress. Autophagy 3, 28–31 Castro Alves, C., Campos, M. L., van Doorn, L. J., Caldas, C. et al. (2003) A
97 Degenhardt, K., Mathew, R., Beaudoin, B., Bray, K., Anderson, D., Chen, G., Mukherjee, proinflammatory genetic profile increases the risk for chronic atrophic gastritis and
C., Shi, Y., Gélinas, C., Fan, Y. et al. (2006) Autophagy promotes tumor cell survival and gastric carcinoma. Gastroenterology 125, 364–371
125 Machado, J. C., Pharoah, P., Sousa, S., Carvalho, R., Oliveira, C., Figueiredo, C.,
restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 10, 51–64
Amorim, A., Seruca, R., Caldas, C., Carniero, F. and Sobrinho-Simoes, M. (2001)
98 Edinger, A. L. and Thompson, C. B. (2003) Defective autophagy leads to cancer. Cancer
Interleukin 1B and interleukin 1RN polymorphisms are associated with increased risk of
Cell 4, 422–424
gastric carcinoma. Gastroenterology 121, 823–829
99 Levine, B. (2005) Eating oneself and uninvited guests: autophagy-related pathways in
126 Palli, D., Saieva, C., Luzzi, I., Masala, G., Topa, S., Sera, F., Gemma, S., Zanna, I.,
cellular defense. Cell 120, 159–162
D’Errico, M., Zini, E. et al. (2005) Interleukin-1 gene polymorphisms and gastric cancer
100 Jin, S. (2006) Autophagy, mitochondrial quality control, and oncogenesis. Autophagy 2,
risk in a high-risk Italian population. Am. J. Gastroenterol. 100, 1941–1948
80–84 127 Furuta, T., El-Omar, E. M., Xiao, F., Shirai, N., Takashima, M. and Sugimura, H. (2002)
101 Zhang, Y., Qi, H., Taylor, R., Xu, W., Liu, L. F. and Jin, S. (2007) The role of autophagy in Interleukin 1β polymorphisms increase risk of hypochlorhydria and atrophic gastritis
mitochondria maintenance: characterization of mitochondrial functions in and reduce risk of duodenal ulcer recurrence in Japan. Gastroenterology 123, 92–105
autophagy-deficient S. cerevisiae strains. Autophagy 3, 337–346 128 Zeng, Z. R., Hu, P. J., Hu, S., Pang, R. P., Chen, M. H., Ng, M. and Sung, J. J. (2003)
102 Gozuacik, D. and Kimchi, A. (2004) Autophagy as a cell death and tumor suppressor Association of interleukin 1B gene polymorphism and gastric cancers in high and low
mechanism. Oncogene 23, 2891–2906 prevalence regions in China. Gut 52, 1684–1689
103 Codogno, P. and Meijer, A. J. (2005) Autophagy and signaling: their role in cell survival 129 Garza-Gonzalez, E., Bosques-Padilla, F. J., El-Omar, E., Hold, G., Tijerina-Menchaca, R.,
and cell death. Cell Death Differ. 12, 1509–1518 Maldonado-Garza, H. J. and Perez-Perez, G. I. (2005) Role of the polymorphic IL-1B,
104 Reef, S., Zalckvar, E., Shifman, O., Bialik, S., Sabanay, H., Oren, M. and Kimchi, A. IL-1RN and TNF-A genes in distal gastric cancer in Mexico. Int. J. Cancer 114, 237–241
(2006) A short mitochondrial form of p19ARF induces autophagy and 130 Wang, Y., Kato, N., Hoshida, Y., Yoshida, H., Taniguchi, H., Goto, T., Moriyama, M.,
caspase-independent cell death. Mol. Cell 22, 463–475 Otsuka, M., Shiina, S., Shiratori, Y. et al. (2003) Interleukin-1β gene polymorphisms
105 Crighton, D., Wilkinson, S., O’Prey, J., Syed, N., Smith, P., Harrison, P. R., Gasco, M., associated with hepatocellular carcinoma in hepatitis C virus infection. Hepatology 37,
Garrone, O., Crook, T. and Ryan, K. M. (2006) DRAM, a p53-induced modulator of 65–71
autophagy, is critical for apoptosis. Cell 126, 121–134 131 Tanaka, Y., Furuta, T., Suzuki, S., Orito, E., Yeo, A. E., Hirashima, N., Sugauchi, F.,
106 Kuhn, R., Lohler, J., Rennick, D., Rajewsky, K. and Muller, W. (1993) Ueda, R. and Mizokami, M. (2003) Impact of interleukin-1β genetic polymorphisms on
Interleukin-10-deficient mice develop chronic enterocolitis. Cell 75, 263–274 the development of hepatitis C virus-related hepatocellular carcinoma in Japan.
107 Sutton, P., Kolesnikow, T., Danon, S., Wilson, J. and Lee, A. (2000) Dominant J. Infect. Dis. 187, 1822–1825
nonresponsiveness to Helicobacter pylori infection is associated with production of 132 Ito, H., Kaneko, K., Makino, R., Konishi, K., Kurahashi, T., Yamamoto, T., Katagiri, A.,
interleukin 10 but not gamma interferon. Infect. Immun. 68, 4802–4804 Kumekawa, Y., Kubota, Y., Muramoto, T. et al. (2007) Interleukin-1β gene in esophageal,
108 El Omar, E. M., Carrington, M., Chow, W. H., McColl, K. E., Bream, J. H., Young, H. A., gastric and colorectal carcinomas. Oncol. Rep. 18, 473–481
Herrera, J., Lissowska, J., Yuan, C. C., Rothman, N. et al. Interleukin-1 polymorphisms 133 Liu, J., Zhai, X., Jin, G., Hu, Z., Wang, S., Wang, X., Qin, J., Gao, J., Ma, H., Wang, X.
associated with increased risk of gastric cancer. Nature 404, 398–402 et al. (2006) Functional variants in the promoter of interleukin-1β are associated with an
109 El Omar, E. M., Rabkin, C. S., Gammon, M. D., Vaughan, T. L., Risch, H. A., Schoenberg, increased risk of breast cancer: a case-control analysis in a Chinese population.
J. B., Stanford, J. L., Mayne, S. T., Goedert, J., Blot, W. J. et al. (2003) Increased risk of Int. J. Cancer 118, 2554–2558
noncardia gastric cancer associated with proinflammatory cytokine gene 134 Beales, I. L. and Calam, J. (1998) Interleukin 1β and tumour necrosis factor α inhibit
polymorphisms. Gastroenterology 124, 1193–1201 acid secretion in cultured rabbit parietal cells by multiple pathways. Gut 42, 227–234
110 Dranoff, G. (2004) Cytokines in cancer pathogenesis and cancer therapy. 135 Kohaar, I., Thakur, N., Salhan, S., Batra, S., Singh, V., Sharma, A., Sodhani, P., Das,
Nat. Rev. Cancer 4, 11–22 B. C., Sarkar, D. P. and Bharadwaj, M. (2007) TNFα − 308G/A polymorphism as a risk
111 Pollard, J. W. (2004) Tumour-educated macrophages promote tumour progression and factor for HPV associated cervical cancer in Indian population. Cell. Oncol. 29, 249–256
136 Duell, E. J., Casella, D. P., Burk, R. D., Kelsey, K. T. and Holly, E. A. (2006) Inflammation,
metastasis. Nat. Rev. Cancer 4, 71–78
genetic polymorphisms in proinflammatory genes TNF-A, RANTES, and CCR5, and risk
112 Miyamoto, Y., Hosotani, R., Doi, R., Wada, M., Ida, J., Tsuji, . S., Kawaguchi, M.,
of pancreatic adenocarcinoma. Cancer Epidemiol. Biomarkers Prev. 15, 726–731
Nakajima, S., Kobayashi, H., Masui, T. and Imamura, M. (2001) Interleukin-6 inhibits
137 Wei, Y. S., Kuang, X. H., Zhu, Y. H., Liang, W. B., Yang, Z. H., Tai, S. H., Zhao, Y. and
radiation induced apoptosis in pancreatic cancer cells. Anticancer Res. 21, 2449–2456
Zhang, L. (2007) Interleukin-10 gene promoter polymorphisms and the risk of
113 Becker, C., Fantini, M.C., Schramm, C., Lehr, H.A., Wirtz, S., Nikolaev, A., Burg, J.,
nasopharyngeal carcinoma. Tissue Antigens 70, 12–17
Strand, S., Kiesslich, R., Huber, S. et al. (2004) TGF-β suppresses tumor progression in
138 Howell, W. M. and Rose-Zerilli, M,J. (2007) Cytokine gene polymorphisms, cancer
colon cancer by inhibition of IL-6 trans-signaling. Immunity 21, 491–501
susceptibility, and prognosis. J. Nutr. 137, S199
114 Chung, Y. C. and Chang, Y. F. (2003) Serum interleukin-6 levels reflect the disease 139 Smith, M. G., Hold, G. L., Rabkin, C. S., Chow, W. H., McColl, K. E. L., Perez-Perez,
status of colorectal cancer. J. Surg. Oncol. 83, 222–226 G. I., Mowat, A. and El-Omar, E. M. (2004) The IL-8-251 promoter polymorphism is
115 Schneider, M. R., Hoeflich, A., Fischer, J. R., Wolf, E., Sordat, B. and Lahm, H. (2000) associated with high IL-8 production, severe inflammation and increased risk of
Interleukin-6 stimulates clonogenic growth of primary and metastatic human colon pre-malignant changes in H-pylori positive subjects. Gastroenterology 126, A23
carcinoma cells. Cancer Lett. 151, 31–38 140 Taguchi, A., Ohmiya, N., Shirai, K., Mabuchi, N., Itoh, A., Hirooka, Y., Niwa, Y. and
116 Vaculova, A., Hofmanova, J., Soucek, K., Kovarikova, M. and Kozubik, A. (2002) Tumor Goto, H. (2005) Interleukin-8 promoter polymorphism increases the risk of atrophic
necrosis factor-α induces apoptosis associated with poly(ADP-ribose) polymerase gastritis and gastric cancer in Japan. Cancer Epidemiol. Biomarkers Prev. 14, t-93
cleavage in HT-29 colon cancer cells. Anticancer Res. 22, 1635–1639 141 Lu, W., Pan, K., Zhang, L., Lin, D., Miao, X. and You, W. (2005) Genetic polymorphisms
117 Lu, H., Ouyang, W. and Huang, C. (2006) Inflammation, a key event in cancer of interleukin (IL)-1B, IL-1RN, IL-8, IL-10 and tumor necrosis factor α and risk of gastric
development. Mol. Cancer Res. 4, 221–233 cancer in a Chinese population. Carcinogenesis 26, 631–636
118 El-Omar, E. M. (2006) Role of host genes in sporadic gastric cancer. Best Pract. Res. 142 Ohyauchi, M., Imatani, A., Yonechi, M., Asano, N., Miura, A., Iijima, K., Koike, T.,
Clin. Gastroenterol. 20, 675–686 Sekine, H., Ohara, S. and Shimosegawa, T. (2005) The polymorphism interleukin 8 -251
119 Risch, N. J. (2000) Searching for genetic determinants in the new millennium. Nature A/T influences the susceptibility of Helicobacter pylori related gastric diseases in the
405, 847–856 Japanese population. Gut 54, 330–335


c The Authors Journal compilation 
c 2008 Biochemical Society
Genetic aspects of inflammation and cancer 235

143 Lee, W. P., Tai, D. I., Lan, K. H., Li, A. F., Hsu, H. C., Lin, E. J., Lin, Y. P., Sheu, M. L., Li, 150 Higgins, S. C., Lavelle, E. C., McCann, C., Keogh, B., McNeela, E., Byrne, P., O’Gorman,
C. P., Chang, F. Y. et al. (2005) The − 251T allele of the interleukin-8 promoter is B., Jarnicki, A., McGuirk, P. and Mills, K. H. (2003) Toll-like receptor 4-mediated innate
associated with increased risk of gastric carcinoma featuring diffuse-type histopathology IL-10 activates antigen-specific regulatory T cells and confers resistance to Bordetella
in Chinese population. Clin. Cancer Res. 11, 6431–6441 pertussis by inhibiting inflammatory pathology. J. Immunol. 171, 3119–3127
144 Segal, E. D., Lange, C., Covacci, A., Tompkins, L. S. and Falkow, S. (1997) Induction of 151 Hold, G. L., Rabkin, C. S., Chow, W. H., Smith, M. G., Gammon, M. D., Risch, H. A.,
host signal transduction pathways by Helicobacter pylori . Proc. Natl. Acad. Sci. U.S.A. Vaughan, T. L., McColl, K. E., Lissowska, J., Zatonski, W. et al. (2007) A functional
94, 7595–7599 polymorphism of toll-like receptor 4 gene increases risk of gastric carcinoma and its
145 Arbour, N. C., Lorenz, E., Schutte, B. C., Zabner, J., Kline, J. N., Jones, M., Frees, K., precursors. Gastroenterology 132, 905–912
Watt, J. L. and Schwartz, D. A. (2000) TLR4 mutations are associated with endotoxin 152 Sanduleanu, S., Jonkers, D., De Bruine, A., Hameeteman, W. and Stockbrugger, R. W.
hyporesponsiveness in humans. Nat. Genet. 25, 187–191 (2001) Double gastric infection with Helicobacter pylori and non-Helicobacter pylori
bacteria during acid-suppressive therapy: increase of pro-inflammatory cytokines and
146 Franchimont, D., Vermeire, S., El Housni, H., Pierik, M., Van Steen, K., Gustot, T.,
development of atrophic gastritis. Aliment. Pharmacol. Ther. 15, 1163–1175
Quertinmont, E., Abramowicz, M., van Gossum, A., Deviere, J. and Rutgeerts, P. (2004)
153 Zavros, Y., Rieder, G., Ferguson, A., Samuelson, L. C. and Merchant, J. L. (2002)
Deficient host-bacteria interactions in inflammatory bowel disease? The toll-like receptor
Genetic or chemical hypochlorhydria is associated with inflammation that modulates
(TLR)-4 Asp299Gly polymorphism is associated with Crohn’s disease and ulcerative
parietal and G-cell populations in mice. Gastroenterology 122, 119–133
colitis. Gut 53, 987–992
154 Barber, M. D., Powell, J. J., Lynch, S. F., Fearon, K. C. and Ross, J. A. (2000) A
147 Kiechl, S., Wiedermann, C. J. and Willeit, J. (2003) Toll-like receptor 4 and polymorphism of the interleukin-1β gene influences survival in pancreatic cancer.
atherogenesis. Ann. Med. 35, 164–171 Br. J. Cancer 83, 1443–1447
148 Kiechl, S., Lorenz, E., Reindl, M., Wiedermann, C. J., Oberhollenzer, F., Bonora, E., 155 Park, K. S., Mok, J. W., Rho, S. A. and Kim, J. C. (1998) Analysis of TNFB and TNFA
Williet, J. and Scwartz, D. A. (2002) Toll-like receptor 4 polymorphisms and NcoI RFLP in colorectal cancer. Mol. Cells 8, 246–249
atherogenesis. N. Engl. J. Med. 347, 185–192 156 de Jong, M. M., Nolte, I. M., te Meerman, G. J., van der Graaf, W. T., de Vries, E. G.,
149 Lorenz, E., Mira, J. P., Frees, K. L. and Schwartz, D. A. (2002) Relevance of mutations in Sijmons, R. H., Hofstra, R. M. and Kleibeuker, J. H. (2002) Low-penetrance genes and
the TLR4 receptor in patients with gram-negative septic shock. Arch. Intern. Med. 162, their involvement in colorectal cancer susceptibility. Cancer Epidemiol. Biomarkers Prev.
1028–1032 11, 1332–1352

Received 1 October 2007/23 November 2007; accepted 30 November 2007


Published on the Internet 12 February 2008, doi:10.1042/BJ20071341


c The Authors Journal compilation 
c 2008 Biochemical Society

You might also like