Sie sind auf Seite 1von 11

review

Inflammatory networks underlying


colorectal cancer
Audrey Lasry1,2, Adar Zinger1,2 & Yinon Ben-Neriah1

Inflammation is emerging as one of the hallmarks of cancer, yet its role in most tumors remains unclear. Whereas a minority
of solid tumors are associated with overt inflammation, long-term treatment with non-steroidal anti-inflammatory drugs is
remarkably effective in reducing cancer rate and death. This indicates that inflammation might have many as-yet-unrecognized
© 2016 Nature America, Inc. All rights reserved.

facets, among which an indolent course might be far more prevalent than previously appreciated. In this Review, we explore the
various inflammatory processes underlying the development and progression of colorectal cancer and discuss anti-inflammatory
means for its prevention and treatment.

Chronic inflammation is one of the hallmarks of cancer1, and many and its surroundings, which include billions of microorganisms, as
tumors arise following prolonged inflammation or display character- well as numerous organic and inorganic substances, that are poten-
istics of chronic inflammation throughout their progression2,3 (Fig. 1). tially harmful. In the gastrointestinal tract, intestinal epithelial cells
Colorectal cancer (CRC) has long been known as one of the best examples (IECs) acquire a series of properties that enable them to function as a
of a tumor tightly associated with chronic inflammation, which can be selective barrier, orchestrate the complex interaction between the host
present from the earliest stages of tumor onset. CRC, the third most com- and its environment, and maintain tissue homeostasis. Along with
mon form of cancer-related death in the USA, is most often sporadic (with other mucosal and sub-mucosal cells, including cells of the immune
no clear hereditary basis), usually caused by a somatic mutation in a gene system, and the overlying mucus layer, the IECs constitute a pro-
encoding a component of the Wnt signaling pathway4, or is sometimes tective shield that stands between the host and the hazardous
caused by an hereditary mutation, as in patients with Lynch syndrome4 intestinal lumen9,10. Damage to this barrier results in pathological
or familial adenomatous polyposis (FAP)5. Much less often, it arises interactions among epithelial cells, microflora and the immune system.
following prolonged inflammation in the intestine, as in patients with This might lead to a disruption of homeostasis, pathological inflam-
inflammatory bowel disease (IBD), such as Crohn’s disease or ulcera- matory responses and tumorigenesis. Barrier dysfunction has been
tive colitis6. Strikingly, while the hereditary cases are rarely preceded by linked to various pathological conditions11, including complications
npg

overt chronic inflammation, they can be prevented or delayed by treat- of liver cirrhosis12, IBD13,14 and CRC15. Here we will discuss the
ment with anti-inflammatory drugs7,8, which suggests the involvement epithelial immunological networks underlying barrier function in
of inflammatory processes in the tumor onset. Inflammatory reactions health and disease.
are often linked to microbial responses, and the intestinal tract is popu- Epithelial innate immune responses activated by various exoge-
lated by myriad bacterial strains, which commonly live in harmony with nous and endogenous danger signals fulfill a critical role in mucosal
their host, yet any substantial shift in the bacterial population can have a immunity16,17, well beyond classical inflammation, and their impor-
considerable effect on inflammatory responses and contribute to tumor tance in both the maintenance of homeostasis and tumorigenesis is
development. In this Review, we explore the various inflammatory proc- increasingly recognized. Pattern-recognition receptors and sensors
esses underlying the development and progression of CRC and discuss of cytoplasmic nucleic acids are key mediators of innate immunity18.
anti-inflammatory means for the prevention and treatment of CRC. After being activated, those receptors and sensors induce signaling
via type I interferons, the main anti-viral effector arm of the immune
Gut epithelial immunity in CRC system, as well as signaling via the adaptor MyD88 (refs. 19,20). Type I
Epithelial structures, including the skin, respiratory mucosa and interferons and MyD88 can be activated not only by exogenous patho-
intestinal mucosal epithelium, constitute a barrier between the host gens and microbe-associated molecular patterns but also in response
to intrinsic cellular stressors, such as danger-associated molecular
patterns (DAMPs)21. An example of a DAMP is the nucleic acid frag-
1The Lautenberg Center for Immunology and Cancer Research, Institute
for Medical Research Israel-Canada, Hebrew University–Hadassah Medical
ments produced as part of the DNA-repair process after damage to
School, Jerusalem, Israel. 2These authors contributed equally to this work. DNA. Damage caused by ultraviolet irradiation, for example, results in
Correspondence should be addressed to Y.B.-N. (yinonb@ekmd.huji.ac.il). the release of a 30–base pair nucleotide-excision product that includes
Received 19 October 2015; accepted 17 December 2015; published online the damage site22. Such fragments, protected by association with a
16 February 2016; doi:10.1038/ni.3384 repair protein, could ignite various signaling pathways, including

230 VOLUME 17  NUMBER 3  MARCH 2016  nature immunology


review

Figure 1  The inflammatory spectrum a Chronic inflammation


Inflammatory cytokines
underlying the development of CRC. (a) Chronic IL-6, TNF, IL-1β, IL-17
inflammation, such as IBD, can be caused Invading bacteria DNA
damage
by barrier dysfunction, Paneth cell defects or
defective innate immune responses, which lead IBD Inflamed epithelial
to enhanced susceptibility to bacterial invasion. cells and
breached barrier
The inflamed environment acts as a fertile
ground for the accumulation of mutations, Normal epithelial tissue
and intact barrier
which can result in tumor development. (b) The
formation of ectopic lymphoid structures (ELSs)
by cells of the immune system in the intestine
b Focal inflammation and ELSs

acts as a tumor-suppressor mechanism for the


eliminatation of transformed cells. In the liver,
however, such structures act as a niche for
Immune Cytotoxic activity
tumor progenitor cells until they mature, acquire of ELSs
cells
self-sufficiency and egress to form tumors.
(c) DNA damage in the intestinal epithelium

Marina Corral Spence/Nature Publishing Group


can activate reactions of the innate immune
system via sensors of double-stranded Tumor
cells ELSs as a niche
DNA. Prolonged DNA damage can result in
for tumor cells
parainflammation, which renders the cells
more sensitive to transformation after
mutation of the gene encoding p53 (TP53).
© 2016 Nature America, Inc. All rights reserved.

signaling by the innate immune system via c Parainflammation


TP53
sensors of nucleic acids (Fig. 2). STING is Chronic DNA damage mutation
a receptor for cytoplasmic nucleotides and
serves as a mediator of signaling by other
Normal epithelial tissue Parainflamed epithelial tissue Tumor
innate sensors23. In a mouse model of defi-
ciency in the DNA-damage sensor ATM
(‘ataxia-telangiectasia mutated’), loss of ATM leads to inactivation partly as mediators between intracellular processes and the tumor
of DNA-repair pathways and constitutive DNA damage, followed microenvironment (Box 1).
by release of DNA to the cytoplasm, which results in a type I inter- Inflammasomes are cytoplasmic protein complexes that sense
feron response via activation of the STING pathway24. When treated diverse inflammatory stimuli, many of which are DAMPs, and serve
with the carcinogen AOM, followed by induction of intestinal inflam- as intracellular signaling platforms downstream of those stimuli28–30.
mation by dextran sulfate sodium (DSS), STING-deficient mice The inflammasome components include, among others, the sensor
display more phosphorylation of the transcription factor STAT3 and NAIP, the scaffold proteins NLRC4, NLRP3, NLRP6 and AIM2,
activation of the transcription factor NF-κB, followed by a greater tumor and the adaptor ASC31. Once the complex is assembled, it activates
load, than that of their wild-type counterparts; this suggests an anti- caspase-1, which in turn leads to the cleavage of precursors and
tumorigenic role for STING in colitis-associated CRC (CAC)25. release of the proinflammatory cytokines interleukin 1β (IL-1β)
Indeed, cytoplasmic sensors are dysregulated in cancerous lesions26,27 and IL-18. Whereas macrophages are considered the main source of
and have been recognized as important participants in tumorigenesis, IL-1β32, both IL-1β and IL-18 are also produced by IECs and regulate
npg

Figure 2  DNA damage and the innate immune


Bacteria UV
response. Cytoplasmic nucleic acid fragments (A)
act as danger signals that modulate various
inflammatory and tumorigenic pathways. ROS Oligonucleotide-repair
Bacterial toxin protein complex (A)
These can be from an exogenous source (A),
such as microbial nucleic acids, bacterial Innate sensors
toxins, ultraviolet irradiation (UV) or ROS, or (B)
endogenous signals (B), a result of continuous STING AIM2
Inflammasome
DNA damage due to impaired mismatch-repair
Mismatch-
mechanisms, as in Lynch syndrome. It has repair defect
Marina Corral Spence/Nature Publishing Group

been suggested that oligonucleotides that


Type I interferon Parainflammation
are excised from the genome in the process
of repairing damage caused by ultraviolet IL-18 IL-1β
irradiation might remain in a cytoplasmic
complex with repair proteins and act as a
signaling factor. These danger signals induce
inflammasome activation, type I interferon
responses and parainflammation. The Intestinal barrier
inflammasome-processed cytokine IL-1β causes
barrier dysfunction, dysbiosis and recruitment
of inflammatory cells, whereas the other Inflammatory
inflammasome-derived cytokine, IL-18, acts to cells
reinforce the intestinal barrier.

nature immunology  VOLUME 17  NUMBER 3  MARCH 2016 231


review

Box 1  Atypical inflammatory responses in CRC


In addition to the well-established role of chronic inflammation in the initiation of CRC, there is also accumulating evidence for the
involvement of atypical inflammatory responses, which might have a substantial effect on tumor development and progression.
  Ectopic lymphoid structures are often found in inflamed or cancerous tissues and exhibit all of the characteristics of lymph nodes
associated with activation of the adaptive immune response161,162. In CRC, these structures have been identified as a positive
prognostic marker163. The mechanism for their formation in tumors has yet to be elucidated; they differ in size and composition in
different patients161, which suggests that their role might also vary. Interestingly, they have a tumor-promoting role in hepatocellular
carcinoma, in which they can act as a niche for the developing tumor cells164 (Fig. 1).
  Another atypical inflammatory role can be attributed to the activation of innate nucleotide sensors in hereditary CRC syndromes.
The two most common familial syndromes are FAP and hereditary non-polyposis CRC (HNPCC; Lynch syndrome). FAP is characterized
by mutation of the gene encoding APC, a negative regulator of Wnt signaling that also regulates cell polarity and chromosome segrega-
tion during mitosis165. Patients with FAP have a truncated APC protein, which leads to the activation of Wnt and chromosomal instability
and as a result, they develop thousands of polyps in the intestine. HNPCC is caused by specific mutations in genes encoding factors
involved in DNA-mismatch repair, which leads to microsatellite instability and eventually results in mutation of the gene encoding
APC166. Whereas both FAP and HNPCC are tightly linked to specific mutations in genes encoding products in signaling pathways
related to DNA repair, they are generally not associated with inflammatory syndromes. However, several studies have indicated that
the treatment of patients with FAP or HNPCC with anti-inflammatory drugs has a beneficial effect and can delay tumor onset7,8, which
suggests that inflammation contributes to tumor formation in these patients. Innate DNA sensors might thus provide the link between
chronic DNA damage and inflammation; several sensors of DNA damage have also been linked to the sensing of cytosolic DNA and the
activation of an immune response via the adaptor STING167,168. It is possible that in patients with impaired DNA-repair pathways,
© 2016 Nature America, Inc. All rights reserved.

the sensors are constantly engaged, which would generate a state of chronic inflammation. Constant activation of DNA sensors by
persistent DNA damage might also be linked to parainflammation. High rates of mutation might also favor loss of functional p53,
which, when combined with parainflammation, creates a potent tumor-promoting mechanism73 (Fig. 3).

the epithelial barrier function32–34 and activity of effector and in colonic myofibroblasts within the epithelial stem cell niche, with
regulatory CD4+ T cells in intestinal mucosa35 (Fig. 2). Accumulating an important role in the regulation of epithelial proliferation and
evidence has shown substantial expression of inflammasome pro- wound healing42.
teins in IECs, which highlights their role in mucosal immunity and
the maintenance of gut homeostasis36. In the AOM-DSS model, IBD and CRC
caspase-1 has a protective effect against tumorigenesis, probably when Patients with IBD, most commonly Crohn’s disease or ulcerative
activated downstream of the NLRC4 inflammasome. This effect is colitis, are at a higher risk of developing CRC, and CAC is responsible
not achieved through a reduction in colonic inflammation but is for approximately 15% of deaths due to IBD6. IBD is characterized
instead a consequence of direct regulation of the proliferation and by a prolonged inflammatory response to luminal bacteria or to
apoptosis of IECs37. sustained mucosal danger signals (DAMPs), yet the genetic factors
AIM2 is a sensor of double-stranded DNA that serves as one of the underlying IBD have only just begun to be delineated, with approxi-
mediators of inflammasome assembly. The contribution of AIM2 to mately 100 different risk loci for IBD identified via genome-wide
tumorigenesis has gained interest over the past few years. Analysis of tis- association studies, 30 of which are common to Crohn’s disease
npg

sues from 414 patients with CRC revealed that 67% of them had reduced and ulcerative colitis43.
expression of AIM2 in tumor tissue compared with that of adjacent Genome-wide association studies have enabled the identification of
normal tissue. Lower expression of AIM2 was linked to higher common pathways affected by mutations in IBD-risk genes, including
mortality rate26. For characterization of the role of AIM2 in colonic intestinal barrier function44,45, defense against microbes46, regulation
tumorigenesis, the AOM-DSS CRC protocol was applied to AIM2- of innate immunity, generation of reactive oxygen species (ROS) 47,
deficient mice. AIM2-deficient mice developed a significantly greater autophagy48–50, endoplasmic reticulum stress51 and adaptive immu-
tumor load, both in number and size, than that of their wild-type nity. In some cases, a mutation in a relevant gene is enough to cause
counterparts. This effect was inflammasome independent and was the disease, while in other cases the mutation by itself will not lead to
attributed to the inhibitory effect of AIM2 on the tumor-initiating disease development, unless it occurs in combination with an as-yet-
intestinal stem cells with high Wnt activity38,39. unknown environmental effect52. Patients with IBD, as well as mouse
NLRP6, a member of the NLR family of receptors, takes part in models mimicking the human disease, often show defects in intes-
inflammasome signaling and has a critical role in host defense against tinal barrier function that result from several causes. These include
infection. Several studies have shown that NLRP6 is an important par- defects in the mucus layer lining the intestine, as well as changes
ticipant in the maintenance of mucosal homeostasis. It is expressed in in intestinal microbiota, which culminate in an increase in muco-
IECs, especially in goblet cells, in which it regulates mucus secretion lytic bacteria. Impaired innate immune responses can occur due to
and barrier integrity via control of the autophagy pathway40. Defects mutations in genes encoding major regulators of the innate immune
in the NLRP6 inflammasome result in mucosal hyperplasia, spontane- system that affect the function of dendritic cells, macrophages and
ous colitis and exacerbation of the inflammatory response after treat- neutrophils, as well as mutations in genes encoding such factors that
ment with DSS. The protective effect of NLRP6 has been attributed regulate IECs. An example of the latter is the NLRP3 inflamma-
to inflammasome-mediated release of IL-18 and modulation of the some, in which single-nucleotide polymorphisms in related genes
intestinal microbiome41, possibly via the secretion of anti-microbial are associated with enhanced susceptibility for Crohn’s disease53.
peptides34. Notably, one study has shown high expression of NLRP6 Mice with deficiency in the NLRP3 inflammasome display defects in

232 VOLUME 17  NUMBER 3  MARCH 2016  nature immunology


review

Figure 3  Inflammatory basis of sporadic CRC a Sporadic CRC


or of CAC. (a) In sporadic CRC, an early event Normal tissue Early adenoma Late adenoma Carcinoma
is mutation in the Wnt pathway, usually in the IL-17, IL-23
Bacteria
gene encoding APC, accompanied by loss of
cell polarity and tight junctions, which enables
the invasion of bacteria and production of the APC KRAS TP53
pro-inflammatory cytokines IL-23 and IL-17.
Mutations in the gene encoding APC are
usually followed by mutations in the oncogene Intestinal crypts
KRAS and the gene encoding p53 (TP53),

Marina Corral Spence/Nature Publishing Group


which enable the progression from adenoma to Activation of innate dsDNA sensors, loss of Modulated inflammatory environment,
cell polarity, tumor-suppressive parainflammation tumor-promoting parainflammation
carcinoma. dsDNA, double-stranded DNA.
(b) In CAC, inflammatory cytokines are constantly
present in the intestinal tissue. Mutation of
the gene encoding p53, an early event in CAC,
b CAC
Inflamed tissue Low-grade dysplasia High-grade dysplasia Carcinoma
results in prolonged activation of NF-κB and
IL-6, TNF
enhanced inflammation. Additionally, barrier
dysfunction promotes the invasion of bacteria,
which further enhances the inflammatory TP53 APC
response. The inflammatory environment
generates ROS, which can drive DNA damage, ROS
finally resulting in mutation of the gene encoding
APC and the development of carcinomas. STAT3 activation, NF-κB activation, barrier dysfunction
© 2016 Nature America, Inc. All rights reserved.

the activation of both macrophages and epithelial cells of the colon rheumatoid arthritis69, psoriasis70 and type 2 diabetes71. In CAC
and are thus more susceptible to DSS-induced colitis 54,55. Finally, mouse models, constitutive activation of Wnt induces TNF, which
mutations that affect the function of Paneth cells have also been linked leads to enhanced activation of NF-κB, with de-differentiation of
to IBD51,56–58. Paneth cells reside in the bottom of intestinal crypts IECs and facilitated tumor initiation72. Similar de-differentiation
and protect against pathogenic bacteria59, and they create a niche for processes are observed in patients with ulcerative colitis72, which
the neighboring intestinal stem cells60. The breakdown of defense suggests that de-differentiation might be a part of the wound-healing
mechanisms activated by Paneth cells can lead to prolonged inflam- response that is activated following bouts of intestinal inflammation
mation and can contribute to a noxious environment for stem cell and facilitates tumor initiation. However, other models of constitutive
proliferation, which might account, at least in part, for the increased activation of Wnt have not provided evidence of NF-κB activation73;
risk for CAC development. hence, the overall effect of the inflammatory basis for epithelial
CAC is the most serious complication of IBD, and thus considerable de-differentiation in CAC remains to be established. Constitutive
effort has been invested in understanding the molecular events that gov- activation of NF-κB can induce DNA damage by activation of ROS74,
ern the transition from inflammation to malignancy (Fig. 3). NF-κB, which provides an additional link between chronic inflammation
one of the key transcription factors that regulates immunological and and tumor initiation. Therefore, in addition to promoting cell pro­
inflammatory responses61, has been identified as one of the main liferation, the inflammatory environment creates a noxious envi-
participants in this transition. In mouse models, epithelial insertion ronment for cell proliferation and thus increases the risk for the
of a constitutively active IκB kinase (IKKβ), which phosphorylates occurrence of tumor-initiating cells.
IκB (the inhibitor of NF-κB)62 and enables constitutive activation NF-κB is also activated following loss of the tumor suppressor p53
npg

of NF-κB, lead to the accelerated development of colorectal tumors in IECs in a mouse model of CAC75,76. Mutation of the gene encoding
caused by enhanced DNA damage, with no overt signs of inflamma- p53 is one of the most common events in sporadic CRC, occurring
tion63. Accordingly, epithelial deletion of IKKβ, which prevents the in approximately 50% of human colorectal tumors77, and is usually
activation of NF-κB, leads to a significant reduction in tumor inci- thought to be the final stage in the transition from benign adenoma to
dence due to increased apoptosis of epithelial cells during the early invasive carcinoma (Fig. 3). However, in CAC arising after prolonged
stages of tumorigenesis, and myeloid deletion of this kinase results intestinal inflammation, mutation of the gene encoding p53 is an early
in reduced tumor size64. That last result is due to reduced production event in tumor development78, which suggests that its role in that is
of pro-inflammatory cytokines, which also promote tumor growth; a different from its role in sporadic CRC. Loss of p53 in AOM-DSS–
comprehensive review of cytokines in CRC has been published else- treated mice is reported to cause activation of NF-κB in epithelial cells
where65. One of the main cytokines induced by NF-κB is IL-6 (ref. 64), as well as surrounding stromal cells75 and to enhance the recruitment
which has a crucial role in the initiation and progression of CAC. IL-6 of inflammatory myeloid cells and invasion. This is mediated by loss
originates from cells of the immune system in the lamina propria of barrier function following ablation of p53, which permits the inva-
and can enhance the proliferation of epithelial cells as well as protect sion of bacteria into tumors and leads to the activation of NF-κB.
them from apoptosis. Deletion of IL-6 reduces tumorigenesis in the Along the same lines, mutations in the gene encoding p53 have been
AOM-DSS mouse model. IL-6 signals from the lamina propria activate found to be associated with activation of NF-κB in an experimental
STAT3 in IECs, which can then promote tumorigenesis via facilitating mouse model of colitis and in samples from patients with CAC76, but
their proliferation, inhibition of apoptosis, and other pro-tumorigenic without defects in epithelial barrier function, and the mechanism by
pathways66,67. The IL-6–STAT3 signaling axis can also repress the which mutant p53 leads to the activation of NF-κB remains unknown.
tumor-suppressor microRNA miR-34a and thereby enhance epithelial- Interestingly, loss of p53 can also modulate the inflammatory response
to-mesenchymal transition and promote tumor-cell invasion68. caused by senescence in a mouse model of sporadic CRC73, which
Another major arm of NF-κB signaling is the activation of TNF, suggests that the loss of p53 at a late stage of CRC influences the
a proinflammatory cytokine involved in various diseases, such as inflammatory environment. Overall, much remains to be elucidated

nature immunology  VOLUME 17  NUMBER 3  MARCH 2016 233


review

Box 2  The intestinal microbiome, inflammation and cancer


Advances in sequencing techniques have allowed considerable progress in elucidating the intestinal microbiome, previously considered
a ‘black box’. Particularly interesting is the integration of the microbiome into the signaling networks that underlie mucosal immunity
and its contribution to intestinal tumorigenesis. CRC is often associated with dysbiosis, a broad change in intestinal microbes and
enrichment for certain microbial strains86,87. While some strains are probably passive bystanders whose overgrowth is a result of the
changing microenvironment, others might have an active role in tumorigenesis. It is well known that microbes produce molecular patterns
that activate innate immunity, yet it appears that commensal bacteria can also modify DNA-repair pathways and induce DAMPs that
trigger epithelial innate immunity. ETBF, via its toxin, upregulates expression of the catabolic enzyme spermin oxydase and induces ROS
formation and thus DNA damage169. Shigella flexneri deregulates expression of the activation of p53 and impairs p53-dependant repair
of DNA170. E. coli strains of the phylogenic group B2 contain a preserved gene cluster (the ‘pks island’) that encodes the genotoxin
colibactin. These E. coli strains induce histone variant γ-H2AX foci in mouse colon as evidence of DNA damage. In vitro studies of
mammalian cell lines have shown that pks+ E. coli strains induce DNA damage, chromosomal aberrations and aneuploidy171.
  In other cases, the microbiota does not have a direct carcinogenic effect in the epithelial cells but serves as a mediator in the
crosstalk between cells of the immune system and the tumor. One example of this is IL-23- and IL-17-dependent tumor growth: the
pro-tumorigenic IL-17 response is triggered by IL-23 secreted from tumor-associated myeloid cells, whose activation is mediated by
tumor-infiltrating microbes88,89. Another example is the inhibitory effect of F. nucleatum on natural killer cells, which allow the tumor
to evade immunosurveillance172.

about the involvement of the loss of p53 or mutation of its gene in the can be eliminated by prolonged antibiotic treatment, which empha-
© 2016 Nature America, Inc. All rights reserved.

development of CAC and of inflammation in CRC. sizes the importance of microbiota in this model.
Innate lymphoid cells are a heterogeneous group of hematopoietic cells.
Microbiota, inflammation and cancer They express various effector cytokines that resemble those expressed by
The association between bacteria and cancer was first described helper T cell populations, but they lack antigen-specific rearranged recep-
decades ago, yet a causal relationship between the two has remained tors, which distinguishes them from classic lymphocytes of the adaptive
debatable. However, accumulating evidence suggests a ‘driver’ role immune system. Innate lymphoid cells are increasingly recognized as
for bacteria in tumorigenesis, rather than a solely passive ‘passenger’ important participants in mucosal immunity, microbe-induced inflam-
role79. A striking, well-known example of this relationship is gas- matory responses, IBD and tumorigenesis90. These cells can promote
tric infection with Helicobacter pylori. Chronic gastritis caused by bacteria-driven colon cancer via IL-23-dependent secretion of IL-22 by
H. pylori is a strong risk factor for gastric adenocarcinoma80,81. inducing phosphorylation of STAT3 and tumor progression91.
Mucosa-associated lymphoid tissue lymphoma is another malignant Studies have suggested the existence of ‘outside-in’ and ‘inside-out’
disease attributed to H. pylori infection82. Strikingly, at an early stage, reciprocal interactions between cell-intrinsic processes and the
this lymphoma can be cured by eradication of H. pylori alone83 and microbiome86. As noted above, AIM2 is a sensor of double-stranded
thus constitutes an example of a malignancy that can be eliminated DNA and an inflammasome protein whose downregulation facilitates
solely by antibiotic treatment. tumorigenic processes and hyper-proliferation of cancer stem cells 38.
The bacterial population that constitutes the gut microbiome Sequencing analysis of 16S rRNA from mouse stools has revealed
exceeds the number of human cells in the body by approximately a markedly distinct microbial population in Aim2-deficient mice
tenfold84, and the microbiota is sometimes referred to as the ‘forgotten relative to that of wild-type mice. This, in turn, modifies the AIM2
npg

organ’ due to its increasingly recognized importance in various pro-tumorigenic phenotype. Strikingly, when Aim2-deficient mice
processes in health and disease85. However, it is not only pathogenic are co-housed with wild-type mice, their tumor load after treatment
bacterial infections that cause overt chronic inflammation that have with AOM-DSS is significantly smaller than that of their mutant
a role in tumorigenesis, as altered interactions between the host and counterparts housed separately, a result that might be attributable to
commensal bacteria can also contribute to malignant transformation86. the transferable intestinal bacterial population38.
We are tempted to speculate that the much greater prevalence of colon Autophagy is an important intracellular process known to be essen-
tumors than of small intestinal tumors in humans is related to the tial for cell survival, especially under stress conditions. Its regulation
greater bacterial load in the former. is important in tumor development92–94. Inhibition of autophagy by
The development of CRC is associated with a broad change in the ablation of the autophagy-related protein ATG7 results in a signifi-
intestinal microbe population: dysbiosis87. In various CRC mouse cant decrease in intestinal tumor load in mice heterozygous for the
models involving genetic manipulations and exposure to carcinogens, gene encoding the Wnt negative regulator APC. The inhibition of
tumor development can be modulated by growth of the mice in a germ- autophagy causes barrier dysfunction, with bacterial invasion of the
free environment, provision of antibiotic treatment, or co-housing crypts and changes in microbiome composition90. Different from
with wild-type counterparts. This highlights the possibility of a other models in which dysbiosis is a pro-tumorigenic driver39,41,88,
role for the microbiome in intestinal tumorigenesis, either as a direct inhibition of autophagy results in increased infiltration of CD8+
carcinogen or as a mediator in the crosstalk between tumor cells and T cells, the TH1 subset of helper T cells, and regulatory T cells, which
their microenvironment, including the immune system. Indeed, facilitates an anti-tumorigenic immune response. This protective
tumor-infiltrating microbes induce IL-23 production by tumor- response is abolished by antibiotic treatment, a result possibly attrib-
associated myeloid cells, which in turn upregulates the expression utable to the overgrowth of specific antibiotic-resistant bacterial
of pro-tumorigenic cytokines, including IL-17 (ref. 88) (Fig. 3). strains. These contradicting effects of dysbiosis on tumor develop-
IL-17 signals via its receptor IL-17A in transformed IECs and pro- ment highlight the complexity of microbiome-tumor interactions.
motes their growth and survival89. IL-23-dependent tumor growth Unless a microbiotic effect is resolved at single-species level, the effect

234 VOLUME 17  NUMBER 3  MARCH 2016  nature immunology


review

Figure 4  Therapeutic modulation of a Cytokines b


the inflammatory environment in CRC. Bacteria
Activated
The inflammatory environment of CRC tumors Barrier
Tumor immune cells
can be modulated by several therapeutic cells
strategies. (a) Treatment with NSAIDs
can attenuate the production of prostaglandin
E2 (PGE2), an inflammatory mediator that PGE2
Antibiotics
enhances tumor growth. Inhibition of NSAIDs
COX-2 by NSAIDs can prevent evasion of Immune Parainflamed
the immune system. In addition, NSAIDs cells tumor cells
target parainflammatory cells and thus delay
tumor formation or induce tumor regression.
(b) CRC tumors are often invaded by specific
pathogenic bacteria, which can promote

Marina Corral Spence/Nature Publishing Group


Immunological
checkpoint blockers: Anti-IL-6, JAK-STAT
tumorigenesis by evading the immune system
(for example, Fusobacterium nucleatum), c M1
anti-PD-1,
anti-CTLA-4
inhibitors, anti-IL-1β

inducing DNA damage (Escherichia coli, macrophage


ETBF and Enterococcus fecalis) or enhancing d
pro-tumorigenic signaling pathways.
Antibiotics can target those bacteria,
whose elimination might result in tumor
regression. (c) The blockade of immunological
checkpoints such as PD-1 or CTLA-4 can
enhance the recruitment and activation
© 2016 Nature America, Inc. All rights reserved.

of cells of the immune system and


anti-tumorigenic inflammatory cells,
such as anti-tumorigenic M1 macrophages, to
the tumor microenvironment, where they help to eliminate tumor cells. (d) Treatment with cytokine antagonists, such as inhibitors of IL-6 or IL-1β, or
blockade of their downstream signaling pathways, such as with inhibitors of JAK and STAT, can result in the attenuation of inflammatory signaling
and delay of tumor formation. In addition, inhibition of IL-1β can enhance barrier formation.

of a particular mutation of a gene involved in innate immunity on chronic inflammation and basal homeostasis 100 characterized by
dysbiosis and tumorigenesis cannot be predicted with certainty. activation of various genes encoding products involved in innate
Nutrition is an important factor in determining the risk for CRC. immunity, with a remarkable paucity of secreted factors, particularly
Specifically, consumption of red meat has been linked to an increased chemokines, and thus little recruitment of cells of the immune
incidence of CRC. Heme, which is present in high levels in red meat, system to the inflamed tissue. Parainflammation was first described
induces epithelial hyperproliferation, which leads to hyperplasia. This for a mouse model of Wnt activation in the intestine, in studies of
protumorigenic effect is microbiota dependent and can be eliminated mice deficient in CKIα73, a negative regulator of Wnt signaling101.
by antibiotic treatment. Bacterial mucolysis by hydrogen sulfide dam- In these mice, CKIα is deleted from the intestinal epithelium,
ages the protective mucus layer and exposes the epithelium to the which results in massive activation of Wnt, along with a prominent
toxic luminal environment, induced by consumption of heme95. DNA-damage response and activation of p53, with the result of
Although dysbiosis constitutes a broad change in microbial popu- senescence102. Gut homeostasis is maintained, with no tumors, in
lation, studies from the past decade have identified certain bacterial CKIα-deficient mice despite the hyperactivation of Wnt, probably
npg

strains that have a primary role in tumor initiation and propagation due to the parallel activation of several tumor-suppressive pathways.
(Box 2 and Fig. 2). Streptococcus gallolyticus has long been associated Parainflammation might contribute to the maintenance of home­
with colonic pathologies. Although a causal relationship to CRC is ostasis by acting together with the activation of p53 in tumor suppres-
not clear, increased production of inflammatory factors, including the sion. After loss of p53, parainflammation loses its tumor-suppressive
cyclooxygenase COX-2, IL-1 and IL-8, in S. gallolyticus–bearing tumor nature to become tumor promoting (Fig. 1). Remarkably, parainflam-
tissue might indicate its possible contribution to tumor progression96. mation on a background of p53 deficiency can be attenuated by treat-
Enterotoxinogenic Bacteroides fragillis (ETBF) induces cleavage of ment with non-steroidal anti-inflammatory drugs (NSAIDs), which
E-cadherin97, increases Wnt signaling and triggers the activation results in tumor regression73 (Fig. 4).
of NF-κB98. Together these processes contribute to increased epi- A buildup of parainflammation in early tumors might be one of
thelial permeability and mucosal inflammation, which might sup- the mechanisms that account for the observation that adenomatous
port tumorigenesis. A protumorigenic role for ETBF has also been polyps only rarely progress to invasive carcinomas. Activation of
demonstrated in mice heterozygous for the ‘min’ mutation (‘multiple wild-type p53, which is preserved in half of the human polyps, might
intestinal neoplasia’) of the gene encoding APC (Apcmin/+), in which act in conjunction with parainflammation, similar to its role in the
colonization by ETBF is associated with increased formation of colon CKIα-deficient mouse model, and halt tumor progression; only pol-
adenomas mediated at least in part by induction of a mucosal response yps that subsequently undergo mutation of the gene encoding p53
by the TH17 subset of helper T cells99. Antibiotic treatment target- would progress to full-blown tumors (Fig. 1). This indicates that
ing specific bacterial strains might thus help to prevent or suppress NSAID treatment might be beneficial only after such mutation,
colorectal tumorigenesis (Fig. 4). whereas by reducing the tumor-suppressor effect of parainflamma-
tion, NSAID treatment in the absence of such mutation would prove
Parainflammation and CRC harmful. On the other hand, NSAID treatment might also suppress
A novel type of inflammation with a considerable effect on CRC pro- pro-tumorigenic pathways, such as Wnt signaling103; thus, it remains
gression is parainflammation. This is an intermediate state between to be determined if the status of p53 in a tumor influences the efficacy

nature immunology  VOLUME 17  NUMBER 3  MARCH 2016 235


review

Table 1  Therapeutic strategies for modulating the inflammatory environment in CRC


Drug Pathway targeted Target population CRC mouse model Status (as of December 2015)
Aspirin COX-2 and/or Patients at medium or Apcmin/+ mice, in which Sulindac Aspirin is used mainly for cardiovascular diseases
parainflammation high risk of both has been shown to delay treatment and prevention. Multiple large-scale
sporadic CRC and tumorigenesis105; mice deficient observational retrospective studies have shown
familial CRC in both CKIα and p53, a model that long-term aspirin use has a protective
of gut parainflammation in effect of against CRC in patients of average
which NSAIDs have been shown risk, with a modest effect on all-cause mortality.
to suppress parainflammation In prospective studies of high-risk patients,
and tumorigenesis73 such as those with FAP or Lynch syndrome or
with a history of colon adenoma, aspirin
significantly reduces tumor frequency142,143.
Ongoing clinical studies of aspirin in CRC include
adjuvant therapy for advanced-stage CRC144 and
prevention of recurrence after polyp resection145.
Sulindac, Patients with FAP, NSAIDs and selective COX-2 inhibitors (‘coxibs’) are
celecoxib Lynch syndrome or used for analgesia and for the treatment of some
mutations involving rheumatological diseases.
the DNA-repair pathway These agents result in a reduction in CRC in
high-risk populations146–148, but their long term
use is limited mainly by cardiovascular toxicity.
Ongoing studies include adjuvant therapy added
to standard chemotherapy in advanced CRC149,
© 2016 Nature America, Inc. All rights reserved.

a phase I/II study assessing the combination of


celecoxib and EPO 906 (a microtubule stabilizer)
in metastatic CRC150, and the combination of
celecoxib, sulindac and eflornithine (an inhibitor of
ornithine decarboxylase) in the prevention of primary
and secondary CRC151,152
PD-1 inhibitors PD-1 Patients with mismatch A phase II clinical trial of pembrolizumab has
(nivolumab; repair defects in whom reported increased response and survival in patients
pembrolizumab) a high rate of mutations with mutations in the mismatch-repair pathway141;
facilitates the generation clinical trials of nivolumab are now underway153.
of novel epitopes
Antibody to CTLA-4 CTLA-4 Clinical trials to evaluate the efficacy of treatment
(ipilimumab) with ipilimumab alone or in combination with
nivolumab are underway153.
Tocilizumab and IL-6 Patients with IBD or CAC, in Deletion of IL-6 or its receptor Antibodies to IL-6 or its receptor are in use in the
siltuximab which IL-6 is known to have in mouse models of CAC clinic for the treatment of inflammatory diseases
a major role in tumorigen- leads to a reduction in tumor such as rheumatoid arthritis or Castleman’s disease.
esis; combined therapy with burden66,67 A phase I/II clinical trial has failed to show
drugs that target TNF and a substantial clinical effect of siltuximab,
IL-6 might be beneficial for a monoclonal antibody to IL-6, on patients with
patients with CAC advanced solid tumors, including CRC154.
npg

Ruxolitinib and JAK-STAT STAT3 has been shown to have a JAK2 inhibitors are in use for myeloproliferative
tofacitinib major role in CRC downstream disorders. The JAK1/3 inhibitor tofacitinib is used
of IL-6 signaling66,67; inhibition for rheumatoid arthritis. Clinical trials for IBD are
of the JAK-STAT pathway rather underway155,156.
than the IL-6 pathway can prevent
the activation of alternative
signaling pathways such as IL-11
Infliximab, etaner- TNF Patients with IBD or CAC, in Antibody to TNF is in use in the clinic for the
cept and adali- which IL-6 is known to have treatment of patients with various rheumatological
mumab a major role in tumorigenesis; diseases and IBD.
combined therapy with drugs
that target TNF and IL-6
might be beneficial for
patients with CAC
IL-1β inhibitor IL-1β Patients with CRC and In a mouse model of CAC, IL-1β These agents are in use for the treatment of
anakinra CAC, including metastatic released from tumor-infiltrating autoimmune and autoinflammatory disease.
disease; the precursor neutrophils has a critical role in An ongoing phase II clinical trial is assessing
of IL-1β is activated tumorigenesis158; mice with 5-fluorouracil and bevacizumab (monoclonal
by caspase-1 upon single deficiency in the immu­ antibody to VEGF) in combination with anakinra
inflammasome activation noglobulin IL-1 receptor–related in the treatment of metastatic CRC160.
in tumor cells, and IL-1β molecule SIGIRR show enhanced
induces various growth Toll–IL-1 receptor signaling and
factors and angiogenic increased tumorigenesis induced
factors (such as VEGF) by DSS-AOM159
and promotes tumor
growth and metastasis157

236 VOLUME 17  NUMBER 3  MARCH 2016  nature immunology


review

of NSAID treatment. Given the role of mutation of the gene encoding available for the treatment of rheumatoid arthritis or Castelman’s
p53 in enhancing NF-κB activation in CAC76, such mutations might disease127,128, and it is possible that these inhibitors, when adminis-
also possibly enhance parainflammation, which again might explain tered alone or together with conventional chemotherapy, might also
the switch in its nature from tumor suppressor to tumor promoter. have a beneficial effect in the treatment of CAC. Furthermore, inhibi-
tion of IL-6 in patients with IBD might prevent or delay the onset of
Anti-inflammatory therapeutic strategies for CRC CAC. Another possibility is that inhibition of the downstream effec-
Due to its deep inflammatory roots, CRC might be a candidate for tors of IL-6 signaling, JAK-STAT129, would prevent the activation of
treatment or prevention by anti-inflammatory agents (Fig. 4 and alternative pathways following the inhibition of IL-6, such as that of
Table 1). Most striking is the role of NSAIDs in CRC prevention, first the IL-6 family member IL-11, also shown to have a major role in
noticed when preliminary studies of small groups of patients with FAP tumors of the gastrointestinal tract67,130. Patients with IBD are often
demonstrated that after 1 year of treatment with the NSAID sulin- treated with inhibitors of the cytokine TNF131, which is also a major
dac, patients tend to develop less polyps104. Those studies have also participant in the development of CAC. It is possible that the combina-
been recapitulated in a mouse model of FAP (Apcmin/+) with similar tion of treatment with IL-6 inhibitors plus TNF inhibitors might have
results105. In 1991, a large-population observational study reported enhanced or synergistic effects in the prevention of CAC (Table 1).
that NSAID use reduces the risk for fatal CRC106. Subsequently, ret- However, the benefit of such preventive strategies must be weighed
rospective studies demonstrated that NSAID treatment is associated against the treatment hazards. NSAID treatment is associated with
with a reduced risk for colorectal polyps and tumor recurrence107, an increased risk of bleeding, particularly in the gastrointestinal
with an overall reduction of 40–50% in the risk of CRC for patients tract, even at low doses132. Certain patients with fragile blood vessels
who reported long-term use (more than 5 years) of low-dose aspirin. (for example, those with amyloid angiopathy) might be at particular
NSAID treatment also has a positive effect in patients with advanced risk of bleeding after treatment with aspirin 133, a condition that is
CRC, in whom long-term use reduces tumor recurrence108. difficult to diagnose for the exclusion of patients from such treatment.
© 2016 Nature America, Inc. All rights reserved.

Following studies demonstrating a protective effect of NSAIDs in Treatment with antibody to TNF is associated with an increased risk
CRC, additional studies have demonstrated a similar effect in several of certain infectious diseases, such as tuberculosis134 or infection with
other cancer types109–111. However, there is controversy about the Clostridium difficile135, as well as increased risk of fungal infections136
exact mechanism for the function of NSAIDs in tumor prevention. and skin cancer137, and blockade with antibody to IL-6 might lead to
It is conceivable that the anti-carcinogenic effect of NSAIDs stems upper respiratory tract infections138.
at least in part from their anti-inflammatory properties. NSAIDs are Inhibitors of immunological checkpoints, which serve to reactivate
non-selective inhibitors of COX-2 (ref. 112), and indeed, expression of the adaptive immune system in tumors, are an emerging therapeutic
COX-2 is elevated in many tumors, including colorectal113, breast114, strategy in many cancer types. The adaptive immune system has a
pancreas115, stomach116, bladder117 and lung118. COX-2 catalyzes the major role in battling CRC, and the tumor ‘immunoscore’, which quan-
production of prostaglandins involved in myriad tumor-promoting tifies the presence of infiltrating cells of the immune system in tumors,
pathways119–121. COX-2 also has a documented role in modulating the is a better predictor of survival than is tumor stage 139. In particular,
immunological environment and can mediate evasion of the immune in CRC, polarization to the TH1 subset of helper T cells and the pres-
system in a mouse model of melanoma122. Thus, it is not surprising ence of T cells in tumors are beneficial and are positively correlated
that inhibition of COX-2 by NSAIDs has an anti-tumorigenic effect with patient survival140. In this context, using inhibitors of immuno-
in cancers that arise following prolonged chronic inflammation, in logical checkpoints might be a particularly effective strategy for the
which COX-2 is indeed active. However, NSAIDs are also effective treatment of patients with CRC who have a low ‘immunoscore’ and
in the prevention of tumors not preceded by inflammation, such as might possibly alter it to an efficacious high ‘immunoscore’ (Fig. 4).
in patients with FAP or hereditary Lynch syndrome 7,8. Through the Indeed, treatment of patients with CRC with an inhibitor of the cos-
npg

years, many conflicting mechanisms have been proposed to explain timulatory molecule PD-1 has a beneficial effect in patients with
the anti-tumorigenic effects of NSAIDs. One such mechanism sug- advanced CRC and mismatch-repair deficiency141, in whom the high
gests that cytokines released during inflammation have a role in rate of mutation might generate new epitopes that are recognized upon
reprogramming adult stem cells to become malignant cells123. NSAID activation of the immune system. Likewise, COX-2 inhibitors and PD-
use can block this reprogramming and thus prevent tumor develop- 1 inhibitors act synergistically in a mouse model of melanoma122, and
ment. Another proposed mechanism is related to activation of the given the major role of COX-2 in CRC, it is possible that a similar
Wnt pathway by prostaglandins124. This pathway is activated in many synergistic effect might be apparent also in CRC.
different tumors, most notably in CRC, in which NSAID efficacy was
originally detected. NSAIDs have been reported to have a significant Conclusion
advantage in patients with CRC who have a mutation in the gene Whereas researchers estimate that approximately 15% of CRC occurs
encoding the lipid kinase PI(3)K125, yet a subsequent study reported in patients with IBD, epidemiological studies have demonstrated a
no beneficial effects of NSAID use in patients with this mutation126. much greater incidence of beneficial effects of anti-inflammatory
We note that most studies of NSAIDs in cancer therapy are obser- treatment in patients with CRC, particularly prolonged treatment
vational and do not point out a clear mechanism of action. It is thus with NSAIDs, mostly more than 5 years. Although there is only cur-
possible that NSAIDs act by both alleviating overt inflammation, as sory understanding of the mechanisms underpinning the beneficial
seen in patients with CAC, and suppressing covert inflammation, such effects of NSAIDs, the reported studies suggest that the inflamma-
as parainflammation, which might account for the tumor-suppressive tory basis of common sporadic CRC is much wider than previously
effects in patients with hereditary CRC and in sporadic CRC. estimated and perhaps underlies most CRC cases. Confirming that
Given the major role of IL-6 in CAC, inhibition of the IL-6 pathway conjecture has important clinical value, as it might tilt the balance of
might constitute another therapeutic strategy for CRC (Fig. 4). In benefit versus hazard in the preventive use of NSAID or new means
mouse models of CAC, inhibition or ablation of IL-6 or its receptor of anti-inflammatory therapy, depending on CRC risk factors, or
reduces tumor burden66,67. Inhibitors of IL-6 or its receptor are clinically even universally, only after a patient has aged. Moreover, aspirin, the

nature immunology  VOLUME 17  NUMBER 3  MARCH 2016 237


review

most commonly used NSAID, provides cancer-protective benefits 23. Sharma, S., Fitzgerald, K.A., Cancro, M.P. & Marshak-Rothstein, A. Nucleic acid-
sensing receptors: rheostats of autoimmunity and autoinflammation. J. Immunol.
after years of treatment. The mechanism of action of NSAIDs must
195, 3507–3512 (2015).
be understood for the improvement of preventive therapy and 24. Härtlova, A. et al. DNA damage primes the type I interferon system via the
possibly for the combination of anti-inflammatory therapy with cytosolic DNA sensor STING to promote anti-microbial innate immunity. Immunity
42, 332–343 (2015).
immunotherapy, chemotherapy and targeted cancer therapies in 25. Zhu, Q. et al. Cutting edge: STING mediates protection against colorectal
progressive cancer (Fig. 4). Part of this might be achieved by eluci- tumorigenesis by governing the magnitude of intestinal inflammation. J. Immunol.
dation of the diversity of inflammatory responses and their possible 193, 4779–4782 (2014).
26. Dihlmann, S. et al. Lack of Absent in Melanoma 2 (AIM2) expression in tumor
documentation in cancer-susceptible people, especially by non- cells is closely associated with poor survival in colorectal cancer patients.
invasive procedures. Mechanistic studies that explain the origin of Int. J. Cancer 135, 2387–2396 (2014).
27. Hou, J. et al. Hepatic RIG-I predicts survival and interferon-α therapeutic response
various inflammatory responses and their progression might also
in hepatocellular carcinoma. Cancer Cell 25, 49–63 (2014).
result in more-effective, possibly less-hazardous means of inter­ 28. Strowig, T., Henao-Mejia, J., Elinav, E. & Flavell, R. Inflammasomes in health
vention than those currently available. and disease. Nature 481, 278–286 (2012).
29. Hoque, R. et al. TLR9 and the NLRP3 inflammasome link acinar cell death with
inflammation in acute pancreatitis. Gastroenterology 141, 358–369 (2011).
Acknowledgments 30. Pétrilli, V., Dostert, C., Muruve, D.A. & Tschopp, J. The inflammasome: a danger
Supported by Israeli Science Foundation Centers of Excellence, the European sensing complex triggering innate immunity. Curr. Opin. Immunol. 19, 615–622
Research Council Framework Programme 7 (294390 PICHO), The Dr. Miriam and (2007).
31. Vanaja, S.K., Rathinam, V.A. & Fitzgerald, K.A. Mechanisms of inflammasome
Sheldon G. Adelson Medical Research Foundation, the I-CORE program of the
activation: recent advances and novel insights. Trends Cell Biol. 25, 308–315
Israel Science Foundation (41/11) and the Israel Cancer Research Fund (Y.B.-N.). (2015).
32. Kanarek, N. et al. Critical role for IL-1β in DNA damage-induced mucositis.
COMPETING FINANCIAL INTERESTS Proc. Natl. Acad. Sci. USA 111, E702–E711 (2014).
The authors declare no competing financial interests. 33. Nowarski, R. et al. Epithelial IL-18 equilibrium controls barrier function in colitis.
Cell 163, 1444–1456 (2015).
© 2016 Nature America, Inc. All rights reserved.

Reprints and permissions information is available online at http://www.nature.com/ 34. Levy, M. et al. Microbiota-modulated metabolites shape the intestinal
reprints/index.html. microenvironment by regulating NLRP6 inflammasome signaling. Cell 163,
1428–1443 (2015).
1. Hanahan, D. & Weinberg, R.A. Hallmarks of cancer: the next generation. 35. Harrison, O.J. et al. Epithelial-derived IL-18 regulates Th17 cell differentiation
Cell 144, 646–674 (2011). and Foxp3+ Treg cell function in the intestine. Mucosal Immunol. 8, 1226–1236
2. Aggarwal, B.B., Vijayalekshmi, R.V. & Sung, B. Targeting inflammatory pathways (2015).
for prevention and therapy of cancer: short-term friend, long-term foe. Clin. Cancer 36. Sellin, M.E., Maslowski, K.M., Maloy, K.J. & Hardt, W.D. Inflammasomes of the
Res. 15, 425–430 (2009). intestinal epithelium. Trends Immunol. 36, 442–450 (2015).
3. Balkwill, F.R. & Mantovani, A. Cancer-related inflammation: common themes and 37. Hu, B. et al. Inflammation-induced tumorigenesis in the colon is regulated by
therapeutic opportunities. Semin. Cancer Biol. 22, 33–40 (2012). caspase-1 and NLRC4. Proc. Natl. Acad. Sci. USA 107, 21635–21640
4. Rowan, A.J. et al. APC mutations in sporadic colorectal tumors: A mutational (2010).
“hotspot” and interdependence of the “two hits”. Proc. Natl. Acad. Sci. USA 97, 38. Man, S.M. et al. Critical role for the DNA sensor AIM2 in stem cell proliferation
3352–3357 (2000). and cancer. Cell 162, 45–58 (2015).
5. Groden, J. et al. Identification and characterization of the familial adenomatous 39. Rommereim, L.M. & Subramanian, N. AIMing 2 Curtail Cancer. Cell 162, 18–20
polyposis coli gene. Cell 66, 589–600 (1991). (2015).
6. Jess, T., Frisch, M. & Simonsen, J. Trends in overall and cause-specific mortality 40. Wlodarska, M. et al. NLRP6 inflammasome orchestrates the colonic host-microbial
among patients with inflammatory bowel disease from 1982 to 2010. interface by regulating goblet cell mucus secretion. Cell 156, 1045–1059
Clin. Gastroenterol. Hepatol. 11, 43–48 (2013). (2014).
7. Labayle, D. et al. Sulindac causes regression of rectal polyps in familial 41. Elinav, E. et al. NLRP6 inflammasome regulates colonic microbial ecology and
adenomatous polyposis. Gastroenterology 101, 635–639 (1991). risk for colitis. Cell 145, 745–757 (2011).
8. Burn, J. et al. Long-term effect of aspirin on cancer risk in carriers of hereditary 42. Normand, S. et al. Nod-like receptor pyrin domain-containing protein 6 (NLRP6)
colorectal cancer: an analysis from the CAPP2 randomised controlled trial. Lancet controls epithelial self-renewal and colorectal carcinogenesis upon injury.
378, 2081–2087 (2011). Proc. Natl. Acad. Sci. USA 108, 9601–9606 (2011).
9. Turner, J.R. Intestinal mucosal barrier function in health and disease. Nat. Rev. 43. Khor, B., Gardet, A. & Xavier, R.J. Genetics and pathogenesis of inflammatory
Immunol. 9, 799–809 (2009). bowel disease. Nature 474, 307–317 (2011).
npg

10. Zhang, K., Hornef, M.W. & Dupont, A. The intestinal epithelium as guardian of 44. Muise, A.M. et al. Polymorphisms in E-cadherin (CDH1) result in a mis-localised
gut barrier integrity. Cell. Microbiol. 17, 1561–1569 (2015). cytoplasmic protein that is associated with Crohn’s disease. Gut 58, 1121–1127
11. Tlaskalová-Hogenová, H. et al. The role of gut microbiota (commensal bacteria) (2009).
and the mucosal barrier in the pathogenesis of inflammatory and autoimmune 45. Sabath, E. et al. Galpha12 regulates protein interactions within the MDCK cell
diseases and cancer: contribution of germ-free and gnotobiotic animal models of tight junction and inhibits tight-junction assembly. J. Cell Sci. 121, 814–824
human diseases. Cell. Mol. Immunol. 8, 110–120 (2011). (2008).
12. Tsiaoussis, G.I., Assimakopoulos, S.F., Tsamandas, A.C., Triantos, C.K. & 46. Jostins, L. et al. Host-microbe interactions have shaped the genetic architecture
Thomopoulos, K.C. Intestinal barrier dysfunction in cirrhosis: Current concepts in of inflammatory bowel disease. Nature 491, 119–124 (2012).
pathophysiology and clinical implications. World J Hepatol 7, 2058–2068 (2015). 47. Keshavarzian, A. et al. Excessive production of reactive oxygen metabolites by
13. Kiesslich, R. et al. Local barrier dysfunction identified by confocal laser inflamed colon: analysis by chemiluminescence probe. Gastroenterology 103,
endomicroscopy predicts relapse in inflammatory bowel disease. Gut 61, 177–185 (1992).
1146–1153 (2012). 48. Rioux, J.D. et al. Genome-wide association study identifies new susceptibility loci
14. Salim, S.Y. & Soderholm, J.D. Importance of disrupted intestinal barrier in for Crohn disease and implicates autophagy in disease pathogenesis. Nat. Genet.
inflammatory bowel diseases. Inflamm. Bowel Dis. 17, 362–381 (2011). 39, 596–604 (2007).
15. Velcich, A. et al. Colorectal cancer in mice genetically deficient in the mucin 49. Hampe, J. et al. A genome-wide association scan of nonsynonymous SNPs
Muc2. Science 295, 1726–1729 (2002). identifies a susceptibility variant for Crohn disease in ATG16L1. Nat. Genet. 39,
16. Dunn, G.P., Koebel, C.M. & Schreiber, R.D. Interferons, immunity and cancer 207–211 (2007).
immunoediting. Nat. Rev. Immunol. 6, 836–848 (2006). 50. McCarroll, S.A. et al. Deletion polymorphism upstream of IRGM associated with
17. Saleh, M. & Trinchieri, G. Innate immune mechanisms of colitis and colitis- altered IRGM expression and Crohn’s disease. Nat. Genet. 40, 1107–1112
associated colorectal cancer. Nat. Rev. Immunol. 11, 9–20 (2011). (2008).
18. Lin, W.W. & Karin, M. A cytokine-mediated link between innate immunity, 51. Kaser, A. et al. XBP1 links ER stress to intestinal inflammation and confers genetic
inflammation, and cancer. J. Clin. Invest. 117, 1175–1183 (2007). risk for human inflammatory bowel disease. Cell 134, 743–756 (2008).
19. Siegal, F.P. et al. The nature of the principal type 1 interferon-producing cells in 52. Cattin, A.L. et al. Hepatocyte nuclear factor 4alpha, a key factor for homeostasis,
human blood. Science 284, 1835–1837 (1999). cell architecture, and barrier function of the adult intestinal epithelium.
20. Schoggins, J.W. et al. A diverse range of gene products are effectors of the type Mol. Cell. Biol. 29, 6294–6308 (2009).
I interferon antiviral response. Nature 472, 481–485 (2011). 53. Villani, A.C. et al. Common variants in the NLRP3 region contribute to Crohn’s
21. Takeuchi, O. & Akira, S. Pattern recognition receptors and inflammation. disease susceptibility. Nat. Genet. 41, 71–76 (2009).
Cell 140, 805–820 (2010). 54. Allen, I.C. et al. The NLRP3 inflammasome functions as a negative regulator of
22. Kemp, M.G. & Sancar, A. DNA excision repair: where do all the dimers go? tumorigenesis during colitis-associated cancer. J. Exp. Med. 207, 1045–1056
Cell Cycle 11, 2997–3002 (2012). (2010).

238 VOLUME 17  NUMBER 3  MARCH 2016  nature immunology


review

55. Zaki, M.H. et al. The NLRP3 inflammasome protects against loss of epithelial integrity 91. Kirchberger, S. et al. Innate lymphoid cells sustain colon cancer through production
and mortality during experimental colitis. Immunity 32, 379–391 (2010). of interleukin-22 in a mouse model. J. Exp. Med. 210, 917–931 (2013).
56. Adolph, T.E. et al. Paneth cells as a site of origin for intestinal inflammation. 92. Mathew, R., Karantza-Wadsworth, V. & White, E. Role of autophagy in cancer.
Nature 503, 272–276 (2013). Nat. Rev. Cancer 7, 961–967 (2007).
57. Cadwell, K. et al. A key role for autophagy and the autophagy gene Atg16l1 in 93. Vitale, I., Manic, G., Dandrea, V. & De Maria, R. Role of autophagy in the
mouse and human intestinal Paneth cells. Nature 456, 259–263 (2008). maintenance and function of cancer stem cells. Int. J. Dev. Biol. 59, 95–108
58. Biswas, A. et al. Induction and rescue of Nod2-dependent Th1-driven (2015).
granulomatous inflammation of the ileum. Proc. Natl. Acad. Sci. USA 107, 94. Jiang, X., Overholtzer, M. & Thompson, C.B. Autophagy in cellular metabolism
14739–14744 (2010). and cancer. J. Clin. Invest. 125, 47–54 (2015).
59. Porter, E.M., Bevins, C.L., Ghosh, D. & Ganz, T. The multifaceted Paneth cell. 95. Ijssennagger, N. et al. Gut microbiota facilitates dietary heme-induced epithelial
Cellular and molecular life sciences. Cell. Mol. Life Sci. 59, 156–170 (2002). hyperproliferation by opening the mucus barrier in colon. Proc. Natl. Acad. Sci.
60. Sato, T. et al. Paneth cells constitute the niche for Lgr5 stem cells in intestinal USA 112, 10038–10043 (2015).
crypts. Nature 469, 415–418 (2011). 96. Abdulamir, A.S., Hafidh, R.R. & Bakar, F.A. Molecular detection, quantification,
61. Ben-Neriah, Y. & Karin, M. Inflammation meets cancer, with NF-κB as the and isolation of Streptococcus gallolyticus bacteria colonizing colorectal tumors:
matchmaker. Nat. Immunol. 12, 715–723 (2011). inflammation-driven potential of carcinogenesis via IL-1, COX-2, and IL-8. Mol.
62. Karin, M. & Ben-Neriah, Y. Phosphorylation meets ubiquitination: the control of Cancer 9, 249 (2010).
NF-κB activity. Annu. Rev. Immunol. 18, 621–663 (2000). 97. Wu, S., Lim, K.C., Huang, J., Saidi, R.F. & Sears, C.L. Bacteroides fragilis
63. Shaked, H. et al. Chronic epithelial NF-κB activation accelerates APC loss and enterotoxin cleaves the zonula adherens protein, E-cadherin. Proc. Natl. Acad.
intestinal tumor initiation through iNOS up-regulation. Proc. Natl. Acad. Sci. USA Sci. USA 95, 14979–14984 (1998).
109, 14007–14012 (2012). 98. Housseau, F. & Sears, C.L. Enterotoxigenic Bacteroides fragilis (ETBF)-mediated
64. Greten, F.R. et al. IKKβ links inflammation and tumorigenesis in a mouse model colitis in Min (Apc+/−) mice: a human commensal-based murine model of colon
of colitis-associated cancer. Cell 118, 285–296 (2004). carcinogenesis. Cell Cycle 9, 3–5 (2010).
65. West, N.R., McCuaig, S., Franchini, F. & Powrie, F. Emerging cytokine networks 99. Wu, S. et al. A human colonic commensal promotes colon tumorigenesis via activation
in colorectal cancer. Nat. Rev. Immunol. 15, 615–629 (2015). of T helper type 17 T cell responses. Nat. Med. 15, 1016–1022 (2009).
66. Grivennikov, S. et al. IL-6 and Stat3 are required for survival of intestinal 100. Bondar, T. & Medzhitov, R. The origins of tumor-promoting inflammation. Cancer
epithelial cells and development of colitis-associated cancer. Cancer Cell 15, Cell 24, 143–144 (2013).
103–113 (2009). 101. Amit, S. et al. Axin-mediated CKI phosphorylation of β-catenin at Ser 45: a
67. Bollrath, J. et al. gp130-mediated Stat3 activation in enterocytes regulates molecular switch for the Wnt pathway. Genes Dev. 16, 1066–1076 (2002).
© 2016 Nature America, Inc. All rights reserved.

cell survival and cell-cycle progression during colitis-associated tumorigenesis. 102. Elyada, E. et al. CKIα ablation highlights a critical role for p53 in invasiveness
Cancer Cell 15, 91–102 (2009). control. Nature 470, 409–413 (2011).
68. Rokavec, M. et al. IL-6R/STAT3/miR-34a feedback loop promotes EMT-mediated 103. Bos, C.L. et al. Effect of aspirin on the Wnt/β-catenin pathway is mediated via
colorectal cancer invasion and metastasis. J. Clin. Invest. 124, 1853–1867 (2014). protein phosphatase 2A. Oncogene 25, 6447–6456 (2006).
69. Feldmann, M. Development of anti-TNF therapy for rheumatoid arthritis. 104. Waddell, W.R., Ganser, G.F., Cerise, E.J. & Loughry, R.W. Sulindac for polyposis
Nat. Rev. Immunol. 2, 364–371 (2002). of the colon. Am. J. Surg. 157, 175–179 (1989).
70. Reich, K. et al. Infliximab induction and maintenance therapy for moderate- 105. Beazer-Barclay, Y. et al. Sulindac suppresses tumorigenesis in the Min mouse.
to-severe psoriasis: a phase III, multicentre, double-blind trial. Lancet 366, Carcinogenesis 17, 1757–1760 (1996).
1367–1374 (2005). 106. Thun, M.J., Namboodiri, M.M. & Heath, C.W. Jr. Aspirin use and reduced risk of
71. Moller, D.E. Potential role of TNF-α in the pathogenesis of insulin resistance and fatal colon cancer. N. Engl. J. Med. 325, 1593–1596 (1991).
type 2 diabetes. Trends Endocrinol. Metab. 11, 212–217 (2000). 107. Sandler, R.S. et al. A randomized trial of aspirin to prevent colorectal adenomas in
72. Schwitalla, S. et al. Intestinal tumorigenesis initiated by dedifferentiation and patients with previous colorectal cancer. N. Engl. J. Med. 348, 883–890 (2003).
acquisition of stem-cell-like properties. Cell 152, 25–38 (2013). 108. Ng, K. et al. Aspirin and COX-2 inhibitor use in patients with stage III colon
73. Pribluda, A. et al. A senescence-inflammatory switch from cancer-inhibitory to cancer. J. Natl. Cancer Inst. 107, 345 (2015).
cancer-promoting mechanism. Cancer Cell 24, 242–256 (2013). 109. Rothwell, P.M. et al. Effect of daily aspirin on long-term risk of death due to
74. Tilstra, J.S. et al. NF-kappaB inhibition delays DNA damage-induced senescence cancer: analysis of individual patient data from randomised trials. Lancet 377,
and aging in mice. J. Clin. Invest. 122, 2601–2612 (2012). 31–41 (2011).
75. Schwitalla, S. et al. Loss of p53 in enterocytes generates an inflammatory 110. Fraser, D.M., Sullivan, F.M., Thompson, A.M. & McCowan, C. Aspirin use and
microenvironment enabling invasion and lymph node metastasis of carcinogen- survival after the diagnosis of breast cancer: a population-based cohort study.
induced colorectal tumors. Cancer Cell 23, 93–106 (2013). Br. J. Cancer 111, 623–627 (2014).
76. Cooks, T. et al. Mutant p53 prolongs NF-κB activation and promotes chronic 111. Streicher, S.A., Yu, H., Lu, L., Kidd, M.S. & Risch, H.A. Case-control study of
inflammation and inflammation-associated colorectal cancer. Cancer Cell 23, aspirin use and risk of pancreatic cancer. Cancer Epidemiol. Biomarkers Prev.
634–646 (2013). 23, 1254–1263 (2014).
77. Rodrigues, N.R. et al. p53 mutations in colorectal cancer. Proc. Natl. Acad. Sci. 112. Hawkey, C.J. COX-2 inhibitors. Lancet 353, 307–314 (1999).
USA 87, 7555–7559 (1990). 113. Williams, C.S., Shattuck-Brandt, R.L. & DuBois, R.N. The role of COX-2 in
npg

78. Brentnall, T.A. et al. Mutations in the p53 gene: an early marker of neoplastic intestinal cancer. Expert Opin. Investig. Drugs 8, 1–12 (1999).
progression in ulcerative colitis. Gastroenterology 107, 369–378 (1994). 114. Holmes, M.D. et al. COX-2 expression predicts worse breast cancer prognosis and
79. Tjalsma, H., Boleij, A., Marchesi, J.R. & Dutilh, B.E. A bacterial driver-passenger does not modify the association with aspirin. Breast Cancer Res. Treat. 130,
model for colorectal cancer: beyond the usual suspects. Nat. Rev. Microbiol. 10, 657–662 (2011).
575–582 (2012). 115. Cascinu, S. et al. COX-2 and NF-κB overexpression is common in pancreatic
80. Parsonnet, J. et al. Helicobacter pylori infection and the risk of gastric carcinoma. cancer but does not predict for COX-2 inhibitors activity in combination with
N. Engl. J. Med. 325, 1127–1131 (1991). gemcitabine and oxaliplatin. Am. J. Clin. Oncol. 30, 526–530 (2007).
81. The EUROGAST Study Group. An international association between Helicobacter 116. Han, S.L., Tang, H.J., Hua, Y.W., Ji, S.Q. & Lin, D.X. Expression of COX-2 in
pylori infection and gastric cancer. Lancet 341, 1359–1362 (1993). stomach cancers and its relation to their biological features. Dig. Surg. 20,
82. Wotherspoon, A.C., Ortiz-Hidalgo, C., Falzon, M.R. & Isaacson, P.G. Helicobacter 107–114 (2003).
pylori-associated gastritis and primary B-cell gastric lymphoma. Lancet 338, 117. Kömhoff, M. et al. Enhanced expression of cyclooxygenase-2 in high grade human
1175–1176 (1991). transitional cell bladder carcinomas. Am. J. Pathol. 157, 29–35 (2000).
83. Wotherspoon, A.C. et al. Regression of primary low-grade B-cell gastric lymphoma 118. Achiwa, H. et al. Prognostic significance of elevated cyclooxygenase 2 expression
of mucosa-associated lymphoid tissue type after eradication of Helicobacter pylori. in primary, resected lung adenocarcinomas. Clin. Cancer Res. 5, 1001–1005
Lancet 342, 575–577 (1993). (1999).
84. Zhang, Y.J. et al. Impacts of gut bacteria on human health and diseases. 119. Vane, J.R. Inhibition of prostaglandin synthesis as a mechanism of action for
Int. J. Mol. Sci. 16, 7493–7519 (2015). aspirin-like drugs. Nat. New Biol. 231, 232–235 (1971).
85. O’Hara, A.M. & Shanahan, F. The gut flora as a forgotten organ. EMBO Rep. 7, 120. Sheng, H., Shao, J., Washington, M.K. & DuBois, R.N. Prostaglandin E2 increases
688–693 (2006). growth and motility of colorectal carcinoma cells. J. Biol. Chem. 276,
86. Elinav, E. et al. Inflammation-induced cancer: crosstalk between tumours, immune 18075–18081 (2001).
cells and microorganisms. Nat. Rev. Cancer 13, 759–771 (2013). 121. Williams, C.S., Mann, M. & DuBois, R.N. The role of cyclooxygenases in
87. Sears, C.L. & Garrett, W.S. Microbes, microbiota, and colon cancer. Cell Host inflammation, cancer, and development. Oncogene 18, 7908–7916 (1999).
Microbe 15, 317–328 (2014). 122. Zelenay, S. et al. Cyclooxygenase-dependent tumor growth through evasion of
88. Grivennikov, S.I. et al. Adenoma-linked barrier defects and microbial products immunity. Cell 162, 1257–1270 (2015).
drive IL-23/IL-17-mediated tumour growth. Nature 491, 254–258 (2012). 123. Herfs, M., Hubert, P. & Delvenne, P. Epithelial metaplasia: adult stem cell
89. Wang, K. et al. Interleukin-17 receptor a signaling in transformed enterocytes reprogramming and (pre)neoplastic transformation mediated by inflammation?
promotes early colorectal tumorigenesis. Immunity 41, 1052–1063 (2014). Trends Mol. Med. 15, 245–253 (2009).
90. Lévy, J. et al. Intestinal inhibition of Atg7 prevents tumour initiation through a 124. Liu, X.H. et al. Prostaglandin E2 modulates components of the Wnt signaling
microbiome-influenced immune response and suppresses tumour growth. Nat. system in bone and prostate cancer cells. Biochem. Biophys. Res. Commun. 394,
Cell Biol 17, 1062–1073 (2015). 715–720 (2010).

nature immunology  VOLUME 17  NUMBER 3  MARCH 2016 239


review

125. Liao, X. et al. Aspirin use, tumor PIK3CA mutation, and colorectal-cancer survival. 147. Steinbach, G. et al. The effect of celecoxib, a cyclooxygenase-2 inhibitor, in
N. Engl. J. Med. 367, 1596–1606 (2012). familial adenomatous polyposis. N. Engl. J. Med. 342, 1946–1952 (2000).
126. Domingo, E. et al. Evaluation of PIK3CA mutation as a predictor of benefit from 148. Bertagnolli, M.M. et al. Five-year efficacy and safety analysis of the Adenoma
nonsteroidal anti-inflammatory drug therapy in colorectal cancer. J. Clin. Oncol. Prevention with Celecoxib Trial. Cancer Prev. Res. (Phila.) 2, 310–321 (2009).
31, 4297–4305 (2013). 149. https://clinicaltrials.gov/ct2/show/NCT01150045?term=NCT01150045&rank=1.
127. Deisseroth, A. et al. FDA approval: siltuximab for the treatment of patients with 150. https://clinicaltrials.gov/ct2/show/NCT00159484?term=NCT00159484&rank=1.
multicentric Castleman disease. Clin. Cancer Res. 21, 950–954 (2015). 151. https://clinicaltrials.gov/ct2/show/NCT00033371?term=NCT00033371&rank=1.
128. Karsdal, M.A. et al. IL-6 receptor inhibition positively modulates bone balance in 152. https://clinicaltrials.gov/ct2/show/NCT01483144?term=NCT01483144&rank=1.
rheumatoid arthritis patients with an inadequate response to anti-tumor necrosis 153. https://clinicaltrials.gov/ct2/show/NCT02060188?term=NCT02060188&rank=1.
factor therapy: biochemical marker analysis of bone metabolism in the tocilizumab 154. Angevin, E. et al. A phase I/II, multiple-dose, dose-escalation study of siltuximab,
RADIATE study (NCT00106522). Semin. Arthritis Rheum. 42, 131–139 (2012). an anti-interleukin-6 monoclonal antibody, in patients with advanced solid tumors.
129. Heinrich, P.C. et al. Principles of interleukin (IL)-6-type cytokine signalling and Clin. Cancer Res. 20, 2192–2204 (2014).
its regulation. Biochem. J. 374, 1–20 (2003). 155. https://clinicaltrials.gov/ct2/show/NCT01458574?term=NCT01458574&rank=1.
130. Putoczki, T.L. et al. Interleukin-11 is the dominant IL-6 family cytokine during 156. https://clinicaltrials.gov/ct2/show/NCT01470599?term=NCT01470599&rank=1.
gastrointestinal tumorigenesis and can be targeted therapeutically. Cancer Cell 157. Dinarello, C.A. Why not treat human cancer with interleukin-1 blockade? Cancer
24, 257–271 (2013). Metastasis Rev. 29, 317–329 (2010).
131. Peyrin-Biroulet, L. Anti-TNF therapy in inflammatory bowel diseases: a huge 158. Wang, Y. et al. Neutrophil infiltration favors colitis-associated tumorigenesis by
review. Minerva Gastroenterol. Dietol. 56, 233–243 (2010). activating the interleukin-1 (IL-1)/IL-6 axis. Mucosal Immunol. 7, 1106–1115
132. Huang, E.S., Strate, L.L., Ho, W.W., Lee, S.S. & Chan, A.T. Long-term use of (2014).
aspirin and the risk of gastrointestinal bleeding. Am. J. Med. 124, 426–433 159. Xiao, H. et al. The Toll-interleukin-1 receptor member SIGIRR regulates colonic
(2011). epithelial homeostasis, inflammation, and tumorigenesis. Immunity 26, 461–475
133. Biffi, A. et al. Aspirin and recurrent intracerebral hemorrhage in cerebral amyloid (2007).
angiopathy. Neurology 75, 693–698 (2010). 160. https://clinicaltrials.gov/ct2/show/NCT02090101?term=NCT02090101&rank=1.
134. Bruns, H. et al. Anti-TNF immunotherapy reduces CD8+ T cell-mediated 161. Pitzalis, C., Jones, G.W., Bombardieri, M. & Jones, S.A. Ectopic lymphoid-like
antimicrobial activity against Mycobacterium tuberculosis in humans. J. Clin. structures in infection, cancer and autoimmunity. Nat. Rev. Immunol. 14,
Invest. 119, 1167–1177 (2009). 447–462 (2014).
135. Ali, T. et al. Clinical use of anti-TNF therapy and increased risk of infections. 162. Dieu-Nosjean, M.C., Goc, J., Giraldo, N.A., Sautes-Fridman, C. & Fridman, W.H. Tertiary
Drug Healthc. Patient Saf. 5, 79–99 (2013). lymphoid structures in cancer and beyond. Trends Immunol. 35, 571–580 (2014).
© 2016 Nature America, Inc. All rights reserved.

136. Ordonez, M.E., Farraye, F.A. & Di Palma, J.A. Endemic fungal infections in 163. Coppola, D. et al. Unique ectopic lymph node-like structures present in human
inflammatory bowel disease associated with anti-TNF antibody therapy. Inflamm. primary colorectal carcinoma are identified by immune gene array profiling.
Bowel Dis. 19, 2490–2500 (2013). Am. J. Pathol. 179, 37–45 (2011).
137. Mercer, L.K. et al. The influence of anti-TNF therapy upon incidence of keratinocyte 164. Finkin, S. et al. Ectopic lymphoid structures function as microniches for tumor
skin cancer in patients with rheumatoid arthritis: longitudinal results from progenitor cells in hepatocellular carcinoma. Nat. Immunol. 16, 1235–1244
the British Society for Rheumatology Biologics Register. Ann. Rheum. Dis. 71, (2015).
869–874 (2012). 165. Kaplan, K.B. et al. A role for the Adenomatous Polyposis Coli protein in
138. Hoshi, D. et al. Incidence of serious respiratory infections in patients with chromosome segregation. Nat. Cell Biol. 3, 429–432 (2001).
rheumatoid arthritis treated with tocilizumab. Mod. Rheum. 22, 122–127 166. Peltomäki, P. Deficient DNA mismatch repair: a common etiologic factor for colon
(2012). cancer. Hum. Mol. Genet. 10, 735–740 (2001).
139. Galon, J. et al. Towards the introduction of the ‘Immunoscore’ in the classification 167. Ferguson, B.J., Mansur, D.S., Peters, N.E., Ren, H. & Smith, G.L. DNA-PK is a
of malignant tumours. J. Pathol. 232, 199–209 (2014). DNA sensor for IRF-3-dependent innate immunity. eLife 1, e00047 (2012).
140. Tosolini, M. et al. Clinical impact of different classes of infiltrating T cytotoxic 168. Kondo, T. et al. DNA damage sensor MRE11 recognizes cytosolic double-stranded
and helper cells (Th1, th2, treg, th17) in patients with colorectal cancer. Cancer DNA and induces type I interferon by regulating STING trafficking. Proc. Natl.
Res. 71, 1263–1271 (2011). Acad. Sci. USA 110, 2969–2974 (2013).
141. Le, D.T. et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. 169. Goodwin, A.C. et al. Polyamine catabolism contributes to enterotoxigenic
J. Med. 372, 2509–2520 (2015). Bacteroides fragilis-induced colon tumorigenesis. Proc. Natl. Acad. Sci. USA 108,
142. Cole, B.F. et al. Aspirin for the chemoprevention of colorectal adenomas: 15354–15359 (2011).
meta-analysis of the randomized trials. J. Natl. Cancer Inst. 101, 256–266 170. Bergounioux, J. et al. Calpain activation by the Shigella flexneri effector VirA
(2009). regulates key steps in the formation and life of the bacterium’s epithelial niche.
143. Burn, J. et al. A randomized placebo-controlled prevention trial of aspirin and/or Cell Host Microbe 11, 240–252 (2012).
resistant starch in young people with familial adenomatous polyposis. Cancer 171. Cuevas-Ramos, G. et al. Escherichia coli induces DNA damage in vivo and
Prev. Res. (Phila.) 4, 655–665 (2011). triggers genomic instability in mammalian cells. Proc. Natl. Acad. Sci. USA 107,
144. https://clinicaltrials.gov/ct2/show/NCT00565708?term=NCT00565708&rank=1. 11537–11542 (2010).
145. https://clinicaltrials.gov/ct2/show/NCT02301286?term=NCT02301286&rank=1. 172. Gur, C. et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human
npg

146. Ait Ouakrim, D. et al. Aspirin, ibuprofen, and the risk of colorectal cancer in inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity 42,
Lynch syndrome. J. Natl. Cancer Inst. 107, djv170 (2015). 344–355 (2015).

240 VOLUME 17  NUMBER 3  MARCH 2016  nature immunology

Das könnte Ihnen auch gefallen