Top Banner
1. Introduction 2. Constitutive STAT3 activation is associated with both cancer initiation and progression 3. Regulation of STAT3 signalling and its relevance to gastric neoplasia 4. STAT3 in murine models of gastric cancer 5. Upstream regulators of STAT3 and their association with H. pylori inflammation 6. H. pylori, STAT3 and IL-17 7. STAT3 and CagA 8. STAT3 and VacA 9. STAT3-activated genes that promote the gastric cancer phenotype 10. Development of IL-11Ra/ gp130 antagonists and STAT3 inhibitors as therapeutic targets 11. Expert opinion Review Targeting STAT3 in gastric cancer Andrew S Giraud , Trevelyan R Menheniott & Louise M Judd Murdoch Childrens Research Institute, Royal Childrens Hospital, Parkville, Australia Introduction: STAT3 is a key transcription factor for many regulatory factors that modulate gene transcription. Particularly important are cytokines and growth factors that maintain homeostasis by regulating immunocytes, stromal and epithelial cells. Dysregulation of STAT3 by constitutive activation plays an important role in the initiation of inflammation and cellular trans- formation in numerous cancers, especially of epithelial origin. This review focuses on STAT3 drive in gastric cancer initiation and progression, with emphasis on its activation by cytokines, and how targeting the primary drivers or gastric STAT3 therapeutically may prevent or slow stomach cancer development. Areas covered: This review will discuss the mechanics of STAT3 signalling, how constitutive STAT3 activation promotes gastric tumourigenesis in both human adenocarcinomas and mouse models, the nature of the upstream regulators of STAT3, and their association with chronic Helicobacter pylori infection, STAT3-activated genes that promote transformation and progression, and finally the development and use of STAT3 and upstream cytokine inhibitors as therapeutics. Expert opinion: Chronic STAT3 activation is a key event in gastric cancer induction and progression. Specific targeting of stomach epithelial STAT3 or blocking IL-11Ra/gp130 and/or EGFR signal transduction in chronic gastric inflammation and metaplasia may be therapeutically effective in preventing gastric carcinogenesis. Keywords: gastric cancer, IL-11, inhibitors, STAT3 Expert Opin. Ther. Targets [Early Online] 1. Introduction Gastric cancer is a significant cause of morbidity and mortality worldwide, and has the second highest death rate of all cancers. It was recently estimated that nearly 1,000,000 new cases of gastric cancer are diagnosed worldwide each year [1] (website: http://globocan.iarc.fr). Human gastric cancer is initiated after chronic infection with the bacterium Helicobacter pylori. Induction of the most common form, intestinal-type adenocarcinoma, is preceded by chronic gastritis (inflamma- tion), metaplasia and dysplasia in susceptible individuals. This stepwise progression is known as the Correa paradigm after its originator [2]. The prognosis for treatment is poor, largely because H. pylori infection is widespread and often asymptomatic from childhood, and so patients typically present after metastasis has occurred. In addition, antibiotic-based eradication is not globally practiced or available, and the prospect of a vaccine in the near future is unlikely. Thus, chronic infection with H. pylori and persistent inflammation predisposes susceptible individuals to disease, so alternative therapeutic options based on blocking chronic inflammatory and oncogenic signalling pathways, as well as new approaches to early cancer detection, need to be addressed. 10.1517/14728222.2012.709238 © 2012 Informa UK, Ltd. ISSN 1472-8222 889 All rights reserved: reproduction in whole or in part not permitted Expert Opin. Ther. Targets Downloaded from informahealthcare.com by Royal Childrens Hospital on 07/29/13 For personal use only.
13

Targeting STAT3 in gastric cancer

Jan 31, 2023

Download

Documents

Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Targeting STAT3 in gastric cancer

1. Introduction

2. Constitutive STAT3 activation

is associated with both cancer

initiation and progression

3. Regulation of STAT3

signalling and its relevance to

gastric neoplasia

4. STAT3 in murine models of

gastric cancer

5. Upstream regulators of

STAT3 and their association

with H. pylori inflammation

6. H. pylori, STAT3 and IL-17

7. STAT3 and CagA

8. STAT3 and VacA

9. STAT3-activated genes that

promote the gastric cancer

phenotype

10. Development of IL-11Ra/gp130 antagonists and

STAT3 inhibitors as

therapeutic targets

11. Expert opinion

Review

Targeting STAT3 in gastric cancerAndrew S Giraud†, Trevelyan R Menheniott & Louise M Judd†Murdoch Childrens Research Institute, Royal Childrens Hospital, Parkville, Australia

Introduction: STAT3 is a key transcription factor for many regulatory factors

that modulate gene transcription. Particularly important are cytokines and

growth factors that maintain homeostasis by regulating immunocytes,

stromal and epithelial cells. Dysregulation of STAT3 by constitutive activation

plays an important role in the initiation of inflammation and cellular trans-

formation in numerous cancers, especially of epithelial origin. This review

focuses on STAT3 drive in gastric cancer initiation and progression, with

emphasis on its activation by cytokines, and how targeting the primary drivers

or gastric STAT3 therapeutically may prevent or slow stomach cancer

development.

Areas covered: This review will discuss the mechanics of STAT3 signalling, how

constitutive STAT3 activation promotes gastric tumourigenesis in both human

adenocarcinomas and mouse models, the nature of the upstream regulators

of STAT3, and their association with chronic Helicobacter pylori infection,

STAT3-activated genes that promote transformation and progression, and

finally the development and use of STAT3 and upstream cytokine inhibitors

as therapeutics.

Expert opinion: Chronic STAT3 activation is a key event in gastric cancer

induction and progression. Specific targeting of stomach epithelial STAT3 or

blocking IL-11Ra/gp130 and/or EGFR signal transduction in chronic gastric

inflammation and metaplasia may be therapeutically effective in preventing

gastric carcinogenesis.

Keywords: gastric cancer, IL-11, inhibitors, STAT3

Expert Opin. Ther. Targets [Early Online]

1. Introduction

Gastric cancer is a significant cause of morbidity and mortality worldwide, andhas the second highest death rate of all cancers. It was recently estimated thatnearly 1,000,000 new cases of gastric cancer are diagnosed worldwide each year [1]

(website: http://globocan.iarc.fr). Human gastric cancer is initiated after chronicinfection with the bacterium Helicobacter pylori. Induction of the most commonform, intestinal-type adenocarcinoma, is preceded by chronic gastritis (inflamma-tion), metaplasia and dysplasia in susceptible individuals. This stepwise progressionis known as the Correa paradigm after its originator [2]. The prognosis for treatmentis poor, largely because H. pylori infection is widespread and often asymptomaticfrom childhood, and so patients typically present after metastasis has occurred. Inaddition, antibiotic-based eradication is not globally practiced or available, andthe prospect of a vaccine in the near future is unlikely. Thus, chronic infectionwith H. pylori and persistent inflammation predisposes susceptible individuals todisease, so alternative therapeutic options based on blocking chronic inflammatoryand oncogenic signalling pathways, as well as new approaches to early cancerdetection, need to be addressed.

10.1517/14728222.2012.709238 © 2012 Informa UK, Ltd. ISSN 1472-8222 889All rights reserved: reproduction in whole or in part not permitted

Exp

ert O

pin.

The

r. T

arge

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

oyal

Chi

ldre

ns H

ospi

tal o

n 07

/29/

13Fo

r pe

rson

al u

se o

nly.

Page 2: Targeting STAT3 in gastric cancer

2. Constitutive STAT3 activation is associatedwith both cancer initiation and progression

Constitutively activated STAT3 is associated with numerouscancers including prostate [3], head and neck [4], breast [5],and colon [6]. When constitutively activated by phosphoryla-tion, STAT3 can transform terminally differentiated andmitotically quiescent cells into proliferating cancer cells [7].Few functional mutations in STAT3 have been reported [8],rather persistent activation is due to overexpression of upstreamligands resulting in chronic JAK2 activity. STAT3 may initiatetumourigenesis by activating transcription of genes involved inproliferation/transformation, cell cycle regulation, inhibition ofprogrammed cell death, and angiogenesis. Once a tumour isinitiated, persistent STAT3 activation can promote invasivegrowth and metastasis.

3. Regulation of STAT3 signalling and itsrelevance to gastric neoplasia

STAT3 is a key transcription factor for many cytokines andgrowth factors that promote gene transcription. Thus EGFreceptor ligands such as HB-EGF, TGFa and amphiregulin,as well as HGF and FGF, all of which are all expressed inthe stomach, can activate STAT3 by phosphorylation afterbinding their respective receptors [9]. Of particular importancein gastric neoplasia, and the focus of this review, are theinflammation-associated cytokines, including members ofthe IL-6 family which signal by binding their cognatea-receptor, which in turn binds to and initiates dimerisationof the signal transducing unit (b receptor or gp130) at theplasma membrane (Figures 1A, 2). The predominantIL-6 family ligands expressed in the stomach are IL-6 andIL-11. Increased expression of IL-11 is associated with chronicH. pylori-associated inflammation [10-12] and gastric cancer [13]

and this cytokine, unlike IL-6, is absolutely required for neo-plastic initiation in animal models of gastric tumourigene-sis [14,15]. Ligand-induced formation of the gp130 receptorcomplex results in phosphorylation of critical intracellular

tyrosine residues by JAK kinases, and triggers two majorsignalling cascades: the SHP2/ras/erk/AP-1 and STAT3 path-ways, which independently activate transcription of targetgenes. Under normal circumstances, these signalling pathwaysare in homeostatic balance, however when imbalanced andbiased towards the STAT3 cascade, the outcome is inflamma-tion, hyperplasia and eventually neoplasia. This has also beenshown to occur in human gastric adenocarcinoma [16,17] wherehigh levels of activated STAT3 are already present in pre-neoplastic atrophic gastritis, particularly in epithelial cellsassociated with CagA+ H. pylori infection [17]. The early acti-vation of STAT3 suggests that it is associated with initiationof infectious pathology, particularly chronic inflammationwhich ultimately contributes to transformation. Togetherthese data indicate that selective inhibition of STAT3 activa-tion may be therapeutically useful in blocking gastric andpotentially other gut tumour growth.

4. STAT3 in murine models of gastric cancer

Murine models of gastric cancer can be broadly divided into twocategories: inflammatory and non-inflammatory. The inflam-matory models typically require infection with H. pylori [18-20]

or H. felis [21], particularly on the background of other geneticalterations including gastrin overexpression [22-24] or loss ofheterozygosity of tumour suppressor genes. There are alsomodels that involve genetic alterations and chronic inflamma-tion in the absence of Helicobacter infection includinggp1307575FF mutant [14,25-28], MHC Class II mutant, IL-1boverexpression [29] and COX2-2/PGES1 overexpression [30-32].

Non-inflammatory models include those involving muta-tions in key genes required for gastric function: gastrin [33,34],H/K-ATPase subunits [35-37], ion transporters [38-42] and trefoilfactor genes [43]. Many of these models have hypergastrinemiaas a key component although both the gp130757FF [25] andthe gastrin null mouse [44] develop stomach tumour phenotypesin the absence of gastrin. Other non-inflammatory models arethose involving mutations in gastric regulatory genes including:mutations in TGFb and its signalling pathways, and mutationsin cell proliferation pathways, for example p53 and p27. Thefinal examples of non-inflammatory models are those inducedby chemical carcinogenesis including administration of MNUor MNNG, as well as mutations in AhR. It is pertinent tonote that for while the initiating mutation or insult would tra-ditionally be considered non-inflammatory, the ensuing cancerthat develops almost always involves an inflammatory response,and modulating this would be expected to reduce the tumourburden. STAT3 activation is a common feature of bothnon-inflammatory and inflammatory models, although itsexpression is not obligatory for their development.

Interest in the activation of STAT3 in murine models ofgastric cancer grew with the description of the gp1307575FF

mutant mouse. This mouse was generated by engineering aknock-in mutation of the critical tyrosine residue at position757 of gp130, the common co-receptor for the IL-6 family of

Article highlights.

. Chronic STAT3 activation is oncogenic in the stomach.

. IL-11/STAT3 drive is absolutely required for gastrictumourigenesis in the gp130757FF mouse, and isassociated with many mouse models of gastriccarcinogenesis.

. Persistent CagA-positive H. pylori infection activatesSTAT3 in mice and humans.

. STAT3 mediates Th-17 lineage specification andIL-17 effector function via H. pylori-dependentIL-23 signalling

. Targeting STAT3 activation therapeutically maycontribute to preventing gastric cancer development.

This box summarises key points contained in the article.

A. S. Giraud et al.

890 Expert Opin. Ther. Targets [Early Online]

Exp

ert O

pin.

The

r. T

arge

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

oyal

Chi

ldre

ns H

ospi

tal o

n 07

/29/

13Fo

r pe

rson

al u

se o

nly.

Page 3: Targeting STAT3 in gastric cancer

cytokines. This mutation prevents SHP-2 (and SOCS3)docking after ligand binding, thus blocking signal transductionvia ras/ERK/AP-1 signalling cascade [45]. SOCS3 normallyfunctions in feedback inhibition of the alternate STAT3 signal-ling cascade emanating from gp130 [25,45]. In the absence ofSOCS3 feedback inhibition in the gp1307575FF mutant,increased oncogenic signalling by STAT3 dimers is observed.As a result, mice develop rapid gastric tumourigenesis in thedistal stomach showing several histopathological features typi-cally observed in human gastric adenocarcinoma (Figure 2B).These features include gastritis, atrophy, mucous metaplasiaand SPEM, dysplasia and submucosal invasion, consistentwith the Correa paradigm [2], but without metastasis. In thismodel, increased activation of STAT3 homodimers is crucial

to tumour development [27,46]. Complete absence of STAT3in mice results in embryonic lethality, but gp130 757FF micewith genetic STAT3 haploinsufficiency develop significantlysmaller gastric tumours (Figure 2B-D) [27,46].

In gp130757FF mutant mice, STAT3 activation and tumourdevelopment does not occur without gp130 activation.IL-11 is a gastric gp130 ligand and is absolutely required forgp130757FF tumourigenesis, since gp130757FF mutant micethat also lack the IL-11 co-receptor, IL-11Ra, neither haveincreased STAT3 activation, nor do they develop gastrictumours [14,15]. gp1307575FF mice housed under specifiedpathogen-free (SPF) conditions and treated with antibiotics(metronidazole and ciprofloxacin) also exhibit restrictedSTAT3 activation and significantly reduced tumour

gp130757F/F STAT3WT/WT

gp130757F/F STAT3WT/WT

gp130757F/F STAT3MUT/WT

gp130757F/F STAT3MUT/WT

gp130757F/F STAT3WT/WT gp130757F/F STAT3MUT/WT

12 weeks 25 weeks

250

200

150

100

Are

a o

f ad

eno

ma

(mm

2 )

Are

a o

f ad

eno

ma/

len

gth

mu

cosa

(m

m2 /m

m)

50

0

0.8*

*

*

0.7

0.6

0.5

0.4

0.3

0.2

0.1

0

B.

C.

D.

A.

IL 11

SOCS3

JAK757F

YYYY

STAT3P

PJAK

SHP2

Ras

MAPK

ERK1/2

tff1

IL 1

1Ra

IL 1

1Ra

gp

130

gp

130

IL 11

Gastrictumor

Figure 1. A. IL-11 signalling in the gp130757FF mouse model of gastric tumourigenesis. A knockin mutation (F for Y) at position

757 of gp130 prevents SHP2 and SOCS3 docking, thereby preventing MAP kinase signalling and activation of the tumour

suppressor trefoil factor 1 gene (tff1), as well as chronic activation of STAT3 respectively. B -- D. Antral tumours in gp130757FF

mice are much larger than those with haploinsufficiency for STAT3, macroscopically [B] in terms of surface area [C] and

microscopically in terms of cross-sectional area [D]. Reproduced from [27,28] with permission from Elsevier.

Targeting STAT3 in gastric cancer

Expert Opin. Ther. Targets [Early Online] 891

Exp

ert O

pin.

The

r. T

arge

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

oyal

Chi

ldre

ns H

ospi

tal o

n 07

/29/

13Fo

r pe

rson

al u

se o

nly.

Page 4: Targeting STAT3 in gastric cancer

growth [27]. Therefore STAT3 activation is crucial for tumourdevelopment in gp1307575FF mice, requiring both binding ofthe gp130 receptor by IL-11 and a gastric microbial presenceto initiate tumourigenesis.Increased activation of STAT3 and expression of IL-11 is

also observed in other models of gastric cancer that are not reli-ant on a mutation in gp130. Gastrin-deficient mice developgastric tumours in association with atrophy, inflammationand metaplasia by 12 months of age. These tumours exhibitelevated STAT3 activation driven by interferon (IFN)-g-biasedinflammation [44]. On the other hand, in the absence of aninflammatory stimulus, both activated STAT3 and tumouri-genesis are significantly reduced in gastrin-deficient mice. Iron-ically, activation of gp130 and subsequently STAT3 also occursin mice with hypergastrinaemia. Genetic ablation of the gastricH+K+ATPase b-subunit in mice leads to achlorhydria, hyper-gastrinaemia and tumour formation with co-incident IL-11expression and STAT3 drive [14]. Additionally, micewith experimental autoimmune gastritis exhibit elevatedIL-11 expression and STAT3 activation [37,47]. Finally, trans-genic mice that overexpress COX2-2/PGES1, K19-C2mEand K19-Wnt1C2mE develop fundic tumours with high levelsof IL-11 expression and STAT3 [14]. So although the moleculartriggers for gastric tumourigenesis are numerous and varied,most mouse models assessed to date are unified by an intrinsicmechanism of STAT3-driven inflammation. In keeping withthis, we have now established that intraperitoneal adminis-tration of high doses of IL-11 for 7 days, without any othermitigating factors, is sufficient to cause hyperactivation ofSTAT3 and injury to the gastric mucosa including gastritisand profound fundic atrophy [48].

5. Upstream regulators of STAT3 and theirassociation with H. pylori inflammation

H. pylori is a stomach-restricted bacterium that exerts a pro-found influence on the development of intestinal-type gastricadenocarcinoma. Significant advances have been made inunderstanding the molecular mechanisms by which H. pyloritriggers, but largely evades, host mucosal immunity. Thehost response to H. pylori is mediated by a highly evolvedmolecular dialogue between the bacterium, gastric mucosalcells and immune cells, involving an array of cytokines, theirreceptors and intracellular transduction cascades. Currentliterature indicates that pro-inflammatory cytokines producedby both infiltrating immune, as well as epithelial cells directlyinitiate and maintain a chronic inflammatory state, ulti-mately leading to gastric cancer in susceptible individuals.The widely described oncogenic role of STAT3 in severalorgan systems naturally poses the question of whetherSTAT3 activity is pivotal in the immune response to H. pyloriinfection and indeed whether it may mediate some of theoncogenic effects of the bacterium.STAT3 exerts a strong functional influence in both the

gastric mucosal and immune compartments [14,25-27,45] and,

by controlling specific intracellular signalling events, is well-positioned to dictate inflammatory and oncogenic outcomesin the context of chronic mucosal colonisation by H. pylori.With the potential of STAT3 as a target for pharmacotherapynow recognised, research efforts should prioritise STAT3inhibition at the formative stages of gastric cancer (H. pylorigastritis) as a strategy to prevent the emergence of increasinglysevere (and mostly irreversible) pathologies of atrophicgastritis, metaplasia and adenocarcinoma respectively. Herewe consider the role of STAT3 in the host immune responseto H. pylori infection from both the mucosal and immuno-logical perspectives, as well as delineating STAT3-dependentprocesses amenable to drug targeting.

We previously reported STAT3 hyperactivation inH. pylori-dependent gastritis [17] and on that basis argued fora functional role for STAT3 in the earliest stages of gastriccancer development. We observed nuclear localisation ofphosphorylated STAT3 in mucosal surface and pit epithelialcells of infected, but not uninfected individuals, suggestingthe active engagement of target gene promoters. This suppo-sition was supported by molecular studies showing increasedmRNA for the STAT3 transcriptional target, SOCS3. Ourfindings are also supported by functional studies ingp130757FF mice, as elucidated above.

6. H. pylori, STAT3 and IL-17

Aside from well-described mucosal homeostatic roles, it isincreasingly apparent that STAT3 activity in immune cellsmay also influence the outcome of H. pylori-related disease.H. pylori infection in humans is associated with a dramaticaccumulation of gastric mucosal mononuclear cells includingCD4+ T cells of the T helper (Th)-1 and Th17-type line-ages [49,50]. Th-1 cell recruitment and associated cytokinerelease are well-described components of the host immuneresponse to H. pylori. On the other hand, increased mucosalprevalence of Th-17 cells, together with elevated expressionof their lineage-defining cytokine, interleukin (IL)-17, haveonly recently gained recognition as factors mediatingH. pylori-specific immunity [49,50]. IL-17 collectively refers toa family of six members (IL-17A-F; reviewed in [51]). None-theless, current evidence argues strongly that IL-17 familycytokines play a central role in H. pylori-related gastritis andmay influence subsequent oncogenic progression [52,53]

although which members participate in the host response toH. pylori is unclear.

STAT3 plays an essential role in Th-17 lineage specifica-tion by activating expression of the critical transcriptionfactor, RORgt which in turn promotes IL-6-dependent,Th-17 polarisation of naive CD4-positive cells. Disruptionof (IL-6-dependent) STAT3 signalling blocks Th-17 differen-tiation without altering the abundance of Th-17 inhibitoryregulatory T cells (Tregs) [54].

Besides playing a key role in the specification of Th-17 fatevia induction of RORgt, STAT3 may also be required for the

A. S. Giraud et al.

892 Expert Opin. Ther. Targets [Early Online]

Exp

ert O

pin.

The

r. T

arge

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

oyal

Chi

ldre

ns H

ospi

tal o

n 07

/29/

13Fo

r pe

rson

al u

se o

nly.

Page 5: Targeting STAT3 in gastric cancer

maintenance of Th-17 effector functions such as IL-17secretion. Caruso and colleagues, in efforts to elucidate themolecular basis of IL-17 production by Th-17 cells duringH. pylori infection, demonstrated a pivotal role for STAT3,acting in concert with the pro-inflammatory cytokineIL-23 [55]. They showed that exogenous IL-23 treatmentdirectly stimulated STAT3 phosphorylation and IL-17 secre-tion in acutely isolated gastric lamina propria mononuclearcells (LPMC), while pharmacological inhibition of STAT3blocked IL-23-dependent IL-17 release. Furthermore, IL-23immunoneutralisation in LPMC cultures derived fromH. pylori-infected patients suppressed both STAT3 activationand IL-17 expression [55]. Therefore, STAT3 positivelyregulates IL-17 transcription in infiltrating Th-17 cells bytransducing H. pylori-related IL-23 signals in infected gastricmucosal tissue.

Elevated IL-23 expression is well-documented in H. pyloriinfection [49] although the cellular basis of this response hasnot yet been formally substantiated in vivo. Nevertheless, thefindings of two recent in vitro studies implicate dendritic cells(DCs) as a likely mucosal source of H. pylori-relatedIL-23 [56,57]. Furthermore, H. pylori-primed DCs are able totrigger IL-17 production in autologous CD4+ T cells [57].Together these studies suggest an attractive model in whichH. pylori-dependent IL-23 release from DCs, acting viaSTAT3, leads to increased mucosal Th-17 cell prevalenceand pro-inflammatory IL-17 production (Figure 2).

IL-17 family cytokines do not exert direct chemotacticactivity but indirectly direct specific cell-mediated immuneresponses by stimulating the release of unrelated pro-inflammatory molecules such as IL-6, a canonical STAT3activator, and a range of chemokines. The process is evidentduring H. pylori infection, where IL-17 has been shown topromote secretion of the chemokine IL-8, by signalling viathe ERK 1/2 MAP kinase pathway [50]. Therefore,IL-17 may indirectly drive cell-mediated immunity (in partvia IL-8) which is a potent chemoattractant for neutrophils.Speculatively, future therapeutic targeting of STAT3 may,by uncoupling the IL-23/STAT3/IL-17 regulatory cascade,provide an effective strategy to subdue IL-17 activity (anddownstream pro-inflammatory actions) and potentiallyimprove the prognosis after chronic H. pylori infection.

The immune responses of immunised and challenged miceinclude T-lymphocyte recruitment and the production ofIL-17 [52]. As a consequence, neutrophil-attracting chemo-kines are released, and the bacterial load is considerablyreduced. Besides regulating gastric inflammation, IL-17 mayalso participate in immune remodelling after immunisation.During infection, Tregs restrain IL-17-specific inflammatoryresponses thus acting in favour of bacterial persistence [52].However a recently published study shows that IL-17 parti-cipates in reducing H. pylori colonisation levels in micefollowing immunisation and challenge [58]. Because immuni-sation is generally thought to produce Helicobacter-specificmemory T-helper cells that potentially alter the ratio between

Th-17 and Treg responses, these findings [58] suggest thatIL-17-mediated inflammatory reactions overwhelm the Tregresponse leading to bacterial clearance. Thus augmentationof STAT3 signalling, rather than inhibition of it, might bepredicted to improve the outcomes of immunisation againstH. pylori by strengthening IL-17-mediated bacterial clearance.

7. STAT3 and CagA

In the past decade, no other aspect of H. pylori biology hasreceived more attention than the cytotoxin-associated antigenA (CagA) [59,60]. CagA is a high molecular weight(120 -- 145 kDa) protein and principal cytotoxin of H. pyloriencoded by the cagA virulence gene. The cagA gene is locatedwithin the cag pathogenicity island (cagPAI), a 40 kb genomicDNA region containing 27 -- 31 virulence genes, some ofwhich encode components of a bacterial type IV secretionsystem (T4SS) that mediate the delivery of CagA into gastricepithelial cells [59,61]. A substantial body of evidence showsthat CagA-positive H. pylori strains exhibit greater virulencethan their CagA-negative counterparts, and carry an increasedrisk of gastric adenocarcinoma [62]. Functional evidence tosupport these studies comes from mouse transgenic experi-ments in which CagA overexpression led to uniformovergrowth of the gastric epithelium and low frequency,late-onset focal tumourigenesis, without significant inductionof gastritis or atrophy [63]. CagA therefore deregulates gastricepithelial cell homeostasis autonomously, but additionalfactors such as secondary somatic mutations, or inflammatorystimuli are evidently required for fully penetrant oncogenesis.The concept is supported by in vitro experiments showingthat CagA mediates oncogenic transformation of primarygastric epithelial cells but only in combination with otherimmortalising agents [64]. The functions of CagA are bestsummarised as follows: it has a demonstrated capacity to altercell polarity and growth kinetics, but its intrinsic transformingeffects are limited (when uncomplemented) and may contri-bute to oncogenic progression only in the subset ofH. pylori-infected individuals with pre-existing genetic orinflammatory susceptibility.

CagA is translocated from the bacterium to gastric epithelialcells via a bacterial type-IV secretion system [65]. Once interna-lised, CagA localises to the inner surface of the plasma mem-brane and is tyrosine phosphorylated at specific C-terminalGlu-Pro-Ile-Tyr-Ala (EPIYA) repeat motifs by src and c-Ablkinases [66,67]. Translocated CagA has been shown to interactwith several intracellular signal transduction cascades associatedwith cell growth and motility [66,68-73]. Current literature iden-tifies Src-homology protein tyrosine phosphatase (SHP)2 as themost widely described intracellular target of CagA [59]. Phos-phorylated CagA specifically binds and activates SHP2, leadingto inappropriate signalling through the SHP2-(Ras)-ERK(MAP-kinase) cascade, deregulated epithelial cell polarityand increased motility; the ‘hummingbird phenotype’ [68,74].Additionally, RNA interference (RNAi)-mediated knockdown

Targeting STAT3 in gastric cancer

Expert Opin. Ther. Targets [Early Online] 893

Exp

ert O

pin.

The

r. T

arge

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

oyal

Chi

ldre

ns H

ospi

tal o

n 07

/29/

13Fo

r pe

rson

al u

se o

nly.

Page 6: Targeting STAT3 in gastric cancer

of SHP2 prevents CagA-dependent ERK activation and deve-lopment of the hummingbird phenotype [74]. Therefore,SHP2 is a pivotal mediator of CagA function in gastricepithelial cells.The association of SHP2 with gastric gp130 signalling [45],

together with the necessity of correctly regulated gp130/JAK/STAT3 pathway activation for the maintenance of gastricmucosal homeostasis [25,45] prompted us to investigate the func-tional impact of CagA on STAT3. We observed significantlyhigher STAT3 activation levels in CagA-positive comparedto CagA-negative, H. pylori-infected human gastric mucosalbiopsies [17], providing empirical support for the perceivedlink between STAT3 and CagA, and have substantiatedthis by demonstrating that transfected CagA triggers STAT3phosphorylation in gastric epithelial cells [73].EPIYA-motif phosphorylation is a major determinant of

CagA-mediated signalling and target gene transcription withgeneral consensus that CagA functions predominantly, thoughnot exclusively, in the phosphorylated state [66,68-73,75,76]. Ourrecently published work shows that phosphorylation of tyrosineresidues within the CagA EPIYA repeat motifs is necessary foractivation of STAT3 since mutation of these tyrosines to serines(EPISA) abolished CagA-dependent STAT3 activation [73].

Two other groups have also demonstrated a link betweenCagA and STAT3 activation [76,77], however, all three studiesdisagree on the mechanism of STAT3 activation. As discussedabove, we have shown a requirement for CagA tyrosine phos-phorylation [73] whereas the other two studies report thatSTAT3 activation occurs either independently of CagA tyro-sine phosphorylation [76] or exclusively by unphosphorylatedCagA [77].

These discrepancies are not easily reconciled but may beexplained by the contrasting cell types and different experi-mental approaches used. One of the groups performed adetailed investigation of CagA-dependent STAT3 activationin non-gastric, laryngeal carcinoma-derived HEp-2 cells [76],whereas the second group assessed STAT3 responses in gastricAGS cells following infection with different H. pylori strainsincluding an internally deleted phosphorylation defectiveCagA mutant lacking an EPIYA motif. [77]. Our studies basedin gastric cell lines used inducible transgenes carrying eitherwild-type CagA or an amino acid substitution mutant inwhich the (EPIYA) tyrosine residues had been replaced withserines (EPISA) [73]. This approach allowed specific abolitionof EPIYA tyrosine phosphorylation without compromisingthe plasma membrane tethering of CagA [78].

IL-8CCL6CCL20

IL-17

Th-17differentiation

IL-6 (STAT3)TGFb

Naive CD4+ T-cell

Sustained SHP2/ERKactivation by CagA

leading to deregulatedepithelial cell polarity,

proliferation andincreased motility

RORgt REG3g

ERK1/2P

PMAPK

Raf

SOS Grb2

Ras

T4SS SRCCagA

CagA

IL-11

IL-11

CagA promotes IL-11release from gastricepithelial cells

H. pylori (CagA)dependent IL-23 releasefrom mucosal DCs

Dendritic cell

IL-11

SHP2Y757

YYYY

JAK2 P

STAT3

gp13

0

gp13

0IL

11R

α

IL 1

1Rα

P P

P

Gastric epithelial cell

STAT3 activationdirectly by CagA, or

indirectly via increasedIL-11 release, leads toincreased bactericidal

REG3γ synthesis

Th-17 cell

Neutrophilrecruitment leadingto reduced H. pyloricolonisation

Neutrophil

IL-23

IL-23

STAT3

JAK2

IL23

RIL

12R

β1

H. pylori

PP

P

Figure 2. The H. pylori cytotoxin CagA activates signalling pathways downstream of gp130 to increase proliferation, motility

and cell polarity as well as synthesis of bactericidal Reg3g in an IL-11/STAT3-dependent fashion. In addition, CagA causes

IL-23 release from mucosal dendritic cells which activates STAT3 in CD4+ T cells and targets such as RORgt which can alter

Th17 differentiation and neutrophil recruitment. Reproduced from [27] with permission from Elsevier.

A. S. Giraud et al.

894 Expert Opin. Ther. Targets [Early Online]

Exp

ert O

pin.

The

r. T

arge

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

oyal

Chi

ldre

ns H

ospi

tal o

n 07

/29/

13Fo

r pe

rson

al u

se o

nly.

Page 7: Targeting STAT3 in gastric cancer

8. STAT3 and VacA

The H. pylori vacuolating toxin VacA induces apoptosisof gastric epithelial cells after activation of Bcl-2 familyproteins. Since STAT3 activity is anti-apoptotic, it is notsurprising that VacA mediates STAT3 inhibition as well asreducing expression of Bcl-2 and Bcl-XL in gastric cancercell lines [79]. Whether functional parallels betweenSTAT3 and VacA exist in vivo is unknown, however thestrong and sustained activation of STAT3 in chronicH. pylori infection in both humans and mouse models (seeprevious sections) suggests that the STAT3-inhibitoryactions of VacA are overwhelmed by distinct mechanismsof H. pylori pathogenicity.

9. STAT3-activated genes that promote thegastric cancer phenotype

STAT3 is known to transcriptionally activate numerous targetgenes. We and others have investigated the spectrum of genesregulated by gp130/STAT3 drive that may have particularrelevance to gastric cancer. It has been established that the dis-tal stomach, where most gastric cancers initiate is particularlyresponsive to IL-11 (but is refractory to IL-6), and thatexogenous IL-11 preferentially activates only STAT and notMAPK/Erk signalling pathways [14]. Therefore the simplestway to ascertain which genes are directly activated bySTAT3 in the stomach in the absence of other confoundingpathologies is to administer IL-11 in vivo, and then quantifyeffects on the gastric transcriptome prior to significantmorphological changes to the gastric mucosa. In this contextit is important to understand that the stomach is composedof a number of functionally distinct compartments: theacid/enzyme secreting fundus (body) and the distal antrum.The spectrum of genes induced by IL-11/STAT3 is differentin these two compartments. The early gene changes in theantral stomach driven directly by activation of the IL-11/gp130/STAT3 pathway include: Regenerating-islet derived(Reg) family members, Clusterin, Gremlin-1, gp130,SOCS3, Jak1, TIMP1 and Gas1 [14]. Significantly, the expres-sion pattern for this IL-11-induced gene signature is alsoobserved in mouse genetic models of gastric disease indepen-dently of exogenous IL-11 administration. These modelsinclude gp130757FF mice, K19-C2mE transgenic mice andH+K+ATPase b-subunit-/- knockout mice. Incidentally, agene expression profile bearing remarkable similarity to themurine IL-11-induced gene signature is observed in humangastric biopsies collected from patients with H. pyloriinfection, pre-neoplastic and neoplastic gastric disease [14].

In contrast, the IL-11/STAT3 gene signature in the fundicmucosa is much more diverse and includes: i) proliferative genessuch as the Reg family members, Igfbp4, Gsdmc1, Grem1 andBlm1, ii) immune-related genes, e.g., Serpin a family members,Dmbt1, IL-33, Spp1, MyD88, and iii) signal transduction genes,e.g., Socs3, Jak3 and Jun b [14,28]. Many of these genes are

reported to be associated with human cancers especially theReg family members [11,80], Grem1 [81,82] and Blm1 [83].

Other studies have indirectly investigated STAT3-regulatedgenes by examining the correlation between STAT3 activationand gene expression, particularly in the setting of gastriccancer. Han and colleagues demonstrated a high correlationbetween STAT3 activation and expression of VEGF, c-Myc,survivin, MMP-7, CD44v6 and cyclinD1 [84], many of whichhave been confirmed to be STAT3-responsive in mouse [46].Others have shown that the expression of survivin and alsoBcl-2 is dependent on the activation of STAT3 [16,46,85]. Interms of the inflammatory response to H. pylori, it has beendemonstrated that the levels of IL-6 and IL-8 expression cor-relate with the activation of STAT3 in both the human [17]

and mouse [27]. In summary, a significant number of genesare proposed to be STAT3-responsive in the stomach. Someof these genes have been identified in mouse genetic studiesin which gastric STAT3 is activated in the absence of othersignificant pathogenic changes. Others have been identifiedin comparative mouse models and human studies in whichthere is a correlation between STAT3 activation and geneexpression, but on the background of complex gastric patho-logies. In spite of this complication all of these studies suggestthat, under permissive conditions, chronic activation ofSTAT3 is oncogenic in the stomach.

10. Development of IL-11Ra/gp130antagonists and STAT3 inhibitors astherapeutic targets

There are numerous points in the IL-11/STAT3 signallingpathway that are amenable to inhibitory blockade. Theseinclude antagonism of IL-11 binding to the IL-11Ra, blockadeof JAK kinase activation by de-phosphorylation, inhibition ofSTAT3 phosphorylation, preventing STAT3 dimerisation, pro-moting STAT3 de-phosphorylation, and inhibiting STAT3-mediated gene transcription. As well as endogenous STAT3inhibitors which include suppressor of cytokine signalling3 (SOCS3), protein inhibitor of activated STAT3 (PIAS3),the SHP2 tyrosine phosphatase, GRIM19 and Grb2 [28] thereare now many small molecule inhibitors of STAT3 that specifi-cally target each part of the IL-6/11 signalling cascade describedpreviously. These have been detailed recently in several compre-hensive reviews [86-89] and the most salient in relation to gastriccancer development are summarised below. To date, only a fewSTAT3 inhibitors have entered clinical trials despite the utilityof blocking constitutive STAT3 activation pathways inepithelial cancers.

10.1 IL-11Ra/gp130 antagonistsRelatively few IL-11 receptor antagonists have been described,likely because of limited information about the potentialpathogenic actions of this cytokine in the stomach, and itsclinical use (oprelvekin) in promoting megakaryocytopoiesisafter chemotherapy. In addition IL-11 has been proposed as

Targeting STAT3 in gastric cancer

Expert Opin. Ther. Targets [Early Online] 895

Exp

ert O

pin.

The

r. T

arge

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

oyal

Chi

ldre

ns H

ospi

tal o

n 07

/29/

13Fo

r pe

rson

al u

se o

nly.

Page 8: Targeting STAT3 in gastric cancer

a therapeutic agent in chronic inflammatory diseases such asCrohn’s disease [90], lending support to the erroneous ideathat it may have generalised anti-inflammatory actions inthe gut.One of the early antagonists to be evaluated for its therapeu-

tic potential was Madindoline A, a low molecular weight, non-peptide inhibitor of IL-6 and IL-11 based on a fulindolinestructure. Madindoline A was active in the 10 -- 100 µM rangefor osteoclastogenesis and proposed to act by suppression ofgp130 and respective ligand a-receptor dimerisation [91].More recently, a high affinity IL-11-specific antagonistW147A has been described [92]. This tryptophan-modifiedIL-11 analogue competitively disrupts gp130/IL-11Ra multi-mer formation and resultant signalling, with biological activityon human endometrial cells [92]. Since IL-11 and LIF areabsolutely required for blastocyst implantation [93], antago-nists of IL-11 action might also have application as contracep-tives. In response to this, polyethylene glycol (PEG)-ylatedIL-11 antagonist has been synthesised [93] and shown to bindthe IL-11Ra, thereby preventing proper interaction ofIL-11Ra and gp130 [93] and blocking STAT3 activation.Although effective after injection, PEG-ylated IL-11 is toolarge to be commercially synthesised as an IL-11 antagonistand is also unlikely to be sufficiently bioavailable if givenorally. To date, the successful application of PEG-ylatedIL-11 as an anti-cancer agent in the stomach has notbeen reported.

10.2 STAT3 inhibitorsInhibition of STAT3 in cancer progression is desirablebecause STAT3 plays a major role in a broad spectrum oftumours, promoting cell growth, survival, angiogenesis andthe local inflammatory response [89]. Since STAT3 is a tran-scriptional regulatory end-point for numerous and diverseregulatory pathways, it has been traditionally viewed as“undruggable” with respect to inhibition in cancer. However,more recently this view has changed [86-88], in part because ofthe realisation that STAT3 also acts as a signalling mediatorand its activation can be specifically blocked at numerouspoints in the signal transduction pathway. Additionally,because STAT3 activity in many normal tissues is low or tran-sient, but constitutively activated to high levels in cancer, ithas a high therapeutic index as a target for inhibition [94]

(but see section 10.3 below). Recent advances in STAT3inhibitor development have demonstrated that new genera-tion inhibitors not only have direct anti-cancer effects, butcan also synergise with other therapies [94,95] and reducemetastatic success. The main types of inhibitors are smallpeptides or peptidomimetics, small molecules derived fromlibrary or natural compound screens, oligonucleotides, andplatinum-based inhibitors [89]. Although medicinal chemistshave generally concentrated on inhibitors which targetSTAT dimerisation and DNA binding, there are many recentreports in the literature of STAT3 inhibitors which act bytyrosine kinase (especially JAK kinase) inhibition. Although

regarded as sub-optimal [86] likely due to concerns aboutspecificity, JAK kinase inhibitors have been described whichare of low molecular weight, orally bioavailable, stable, effica-cious in reducing tumour volume in vivo, and which haveIC50 values in the same range as STAT3 dimer inhibitors.An example is the caffeic acid derivative, WP1066, which pro-motes glioma cell apoptosis in vivo via STAT3 inhibition [96].Moreover, in unpublished work we have shown WP1066 toeffectively block gastric tumour progression in thegp130757FF mouse when given orally (Louise M. Judd,unpublished), and at similar doses to antisense oligonucleoti-des given intraperitoneally in the same model as reportedpreviously [15].

There are relatively few reports in the literature on the useand outcome of using STAT3 inhibitors for blocking humangastric cancer progression, despite the fact that increasedSTAT3 expression and activation has been shown to be associ-ated with gastric cancer development [17,84,97] and with poorsurvival [98-100]. Exceptions to this are three recent studies, test-ing novel phytochemicals against STAT3 activation in vitro andin vivo in the context of VEGF stimulation [101,102]. Thus, eupa-tilin, a flavone from Artemisia asiatica dose-dependentlyreduced STAT3 phosphorylation and VEGF promoter bindingunder hypoxic conditions in MKN45 gastric cancer cells, andalso inhibited VEGF neovascularisation of xenotransplantedcancer cells and tumour growth [101]. Likewise nitidine chloride,derived from Zanthoxylum nitidum had similar effects onSGC-7901 gastric cancer cells and tumours, against the sametargets and at a comparable dose range (7-10 mg/kg/day) [102].This latter cell line and its cisplatin-resistant derivative havealso been used to demonstrate that the porphyrin compoundDPP which prevents STAT3 dimerisation [103] is effective inreducing cisplatin resistance and inducing apoptosis in aSTAT3-dependent fashion [104]. Finally, the green tea deriva-tive (-)-epigallocatechin-3-gallate inhibited STAT3-mediatedVEGF expression in the gastric cancer cell line AGS [105].

Although gastric cancer is a multifaceted disease that hasproved resistant to standard chemotherapeutic treatment [106]

unless detected early, recent advances have begun to define themain molecular targets, for which focussed therapies might bedeveloped. For instance, a recent comprehensive genomic anal-ysis using SNP arrays to interrogate a panel of more than200 gastric cancers concluded that nearly 40% of those investi-gated could be independently classified according to discretegenomic alterations in one of EGFR (ERBB1), ERBB2,KRAS, FGFR2 or MET genes [107]. Since these receptor orreceptor-associated genes either have intrinsic tyrosine kinaseactivity, or are associated with particular tyrosine kinases whichcontribute to downstream signalling, it was concluded thatthese patients would be likely to respond to tyrosine kinase/RAS directed therapies. Changes in STAT3 copy numberwere not reported in this study, however all identified genegroups utilise STAT3-dependent signalling pathways, sugges-ting that inhibition of activated STAT3 might also havetherapeutic value in these patients along with targeted receptor

A. S. Giraud et al.

896 Expert Opin. Ther. Targets [Early Online]

Exp

ert O

pin.

The

r. T

arge

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

oyal

Chi

ldre

ns H

ospi

tal o

n 07

/29/

13Fo

r pe

rson

al u

se o

nly.

Page 9: Targeting STAT3 in gastric cancer

approaches. In support of this, dual use of PI3-kinase andSTAT3 inhibitors have shown utility in inhibiting proliferationin gastric cancer cell lines with KRAS mutations [95], andFGFR2-amplified gastric cancers were sensitive to dovitinib [107]

a multi-kinase inhibitor that also targets STAT3 [108].

10.3 Off-target effects of STAT3 inhibitorsBecause STAT3 is a highly utilised transcription factor inmany tissues, global inhibition of its activity is likely toimpact normal cellular function in both health and disease.Therefore, off-target effects of STAT3 inhibition need to beconsidered when designing inhibitors.

Two potential undesirable actions of global STAT3 inhibi-tion are briefly considered here. The first relates to the obser-vation that STAT3 plays an important role in cardiacprotection and regulation of cardiac inflammation [109]. Theformer is thought to occur by induction of anti-oxidantenzymes as well as by promoting cell survival after inhibitionof key anti-apoptotic mediators [110]. STAT3 appears to bepleiotrophic with respect to regulation of inflammation; itcan mediate pro-inflammatory outcomes but can also inhibitinflammation when activated downstream of cardiacIL-11 [111] and IL-10 [112] in models of myocardial ischaemia.In addition STAT3 is intimately involved in mitochondrialrespiration [113], required for maintenance of energy balancein normal myocardial function [110].

A second area of concern for STAT3 inhibition is inIL-10 signalling. IL-10 is an important anti-inflammatorycytokine, particularly in the gut, where it is expressed by cellsof both the innate and adaptive immune systems, signallingvia the JAK/STAT3 pathway [114]. It is well established thatblockade of IL-10 signalling leads to the development ofinflammatory bowel disease (IBD) in mouse models [115]

and loss of function polymorphisms in IL-10R1 andIL-10R2 increase the chance of IBD development inhumans [116]. These examples highlight the contrasting rolesthat STAT3 plays in normal physiology and tumourigenesis,as well as emphasising the importance of selectively targeting

acute and chronic STAT3 activation in different tissuecompartments by therapeutic STAT3 inhibitors.

11. Expert opinion

STAT3 is a pivotal signalling molecule and transcriptionfactor in gastric mucosal function. Constitutive activation ofSTAT3 by hyperphosphorylation (often accompanied byonly a modest increase in gene expression as quantified bysteady-state mRNA analysis) contributes to pre-neoplasticprogression in the stomach, where it promotes inflammation,cell proliferation, angiogenesis, and inhibits programmed celldeath. Recent data from both animal models and humangastric cancers suggest that a major endogenous driver ofoncogenic STAT3 is the cytokine IL-11. IL-11 has pleiotro-phic actions with respect to inflammation and tumourigene-sis, which are directed in a tissue-specific fashion. Furtherresearch is required to understand the relative roles at eachstage of gastric cancer development of IL-11 and IL-6, bothof which can signal through gp130 along several differentsignal transduction pathways. Selective inhibition of STAT3appears to be a potentially useful therapeutic option inblocking gastric neoplastic progression. Caveats include off-target effects especially in the cardiovascular system, theability to develop selective, orally available antagonists, aswell as establishing whether STAT3 blockade will be effectivein ameliorating metastasis to secondary sites; a commonsituation at first presentation for therapy.

Acknowledgements

Supported by NHMRC (Australia) and the VictorianGovernment’s Operational Infrastructure Support Program.

Declaration of interest

The authors state no conflict of interest and have received nopayment in preparation of this manuscript.

Targeting STAT3 in gastric cancer

Expert Opin. Ther. Targets [Early Online] 897

Exp

ert O

pin.

The

r. T

arge

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

oyal

Chi

ldre

ns H

ospi

tal o

n 07

/29/

13Fo

r pe

rson

al u

se o

nly.

Page 10: Targeting STAT3 in gastric cancer

BibliographyPapers of special note have been highlighted as

either of interest (�) or of considerable interest(��) to readers.

1. Ferlay JSH, Bray F, Forman D, et al.

(2008) GLOBOCAN 2008 v1.2, Cancer

Incidence and Mortality Worldwide:

IARC CancerBase No. 10 [Internet].

International Agency for Research on

Cancer; 2010. 2008 ed

2. Correa P. A human model of gastric

carcinogenesis. Cancer Res

1988;48:3554-60.. Initial presentation of a model for

progression of intestinal-type

gastric cancer.

3. Mora LB, Buettner R, Seigne J, et al.

Constitutive activation of Stat3 in human

prostate tumors and cell lines: direct

inhibition of Stat3 signaling induces

apoptosis of prostate cancer cells.

Cancer Res 2002;62:6659-66

4. Song JI, Grandis JR. STAT signaling in

head and neck cancer. Oncogene

2000;19:2489-95

5. Burke WM, Jin X, Lin HJ, et al.

Inhibition of constitutively active

Stat3 suppresses growth of human

ovarian and breast cancer cells. Oncogene

2001;20:7925-34

6. Corvinus FM, Orth C, Moriggl R, et al.

Persistent STAT3 activation in colon

cancer is associated with enhanced cell

proliferation and tumor growth.

Neoplasia 2005;7:545-55

7. Bromberg JF, Wrzeszczynska MH,

Devgan G, et al. Stat3 as an oncogene.

Cell 1999;98:295-303.. Demonstration that STAT3 is

oncogenic when

constitutively expressed.

8. Willson TA, Kuhn BR, Jurickova I, et al.

STAT3 Genotypic variation and cellular

STAT3 activation and colon leukocyte

recruitment in pediatric crohn disease.

J Pediatr Gastroenterol Nutr

2011;55:32-43

9. Rho O, Kim DJ, Kiguchi K, et al.

Growth factor signaling pathways as

targets for prevention of epithelial

carcinogenesis. Mol Carcinog

2011;50:264-79

10. Economescu MC, Necula LG, Dragu D,

et al. Identification of potential

biomarkers for early and advanced gastric

adenocarcinoma detection.

Hepatogastroenterology 2010;57:1453-64

11. Ellmark P, Ingvarsson J, Carlsson A,

et al. Identification of protein expression

signatures associated with helicobacter

pylori infection and gastric

adenocarcinoma using recombinant

antibody microarrays.

Mol Cell Proteomics 2006;5:1638-46

12. Sugimoto M, Ohno T, Graham DY,

et al. Helicobacter pylori outer

membrane proteins on gastric mucosal

interleukin 6 and 11 expression in

mongolian gerbils. J Gastroenterol

Hepatol 2011;26:77-84

13. Nakayama T, Yoshizaki A, Izumida S,

et al. Expression of interleukin-11

(IL-11) and IL-11 receptor alpha in

human gastric carcinoma and IL-11

upregulates the invasive activity of

human gastric carcinoma cells.

Int J Oncol 2007;30:825-33

14. Howlett M, Giraud AS, Lescesen H,

et al. The interleukin-6 family cytokine

interleukin-11 regulates homeostatic

epithelial cell turnover and promotes

gastric tumor development.

Gastroenterology 2009;136:967-77. Direct demonstration that IL-11 drives

gastric oncogenesis in a mouse model.

15. Ernst M, Najdovska M, Grail D, et al.

STAT3 and STAT1 mediate

IL-11-dependent and

inflammation-associated gastric

tumorigenesis in gp130 receptor mutant

mice. J Clin Invest 2008;118:1727-38. Direct demonstration that IL-11 drives

gastric oncogenesis in a mouse model.

16. Kanda N, Seno H, Konda Y, et al.

STAT3 is constitutively activated and

supports cell survival in association with

survivin expression in gastric cancer cells.

Oncogene 2004;23:4921-9. One of the first associations of

activated STAT3 with human

gastric cancer.

17. Jackson CB, Judd LM, Menheniott TR,

et al. Augmented gp130-mediated

cytokine signalling accompanies human

gastric cancer progression. J Pathol

2007;213:140-51. STAT3 accompanies pre-neoplastic

progression in human gastric cancer.

18. Testino G. Helicobacter pylori, cell

proliferation, and carcinogenesis.

Am J Gastroenterol 2001;96:2514

19. Mandell L, Moran AP, Cocchiarella A,

et al. Intact gram-negative helicobacter

pylori, helicobacter felis, and helicobacter

hepaticus bacteria activate innate

immunity via toll-like receptor 2 but not

toll-like receptor 4. Infect Immun

2004;72:6446-54

20. Correa P, Houghton J. Carcinogenesis of

helicobacter pylori. Gastroenterology

2007;133:659-72

21. Court M, Robinson PA, Dixon MF,

et al. The effect of gender on

helicobacter felis-mediated gastritis,

epithelial cell proliferation, and apoptosis

in the mouse model. J Pathol

2003;201:303-11

22. Takaishi S, Cui G, Frederick DM, et al.

Synergistic inhibitory effects of gastrin

and histamine receptor antagonists on

helicobacter-induced gastric cancer.

Gastroenterology 2005;128:1965-83

23. Lee CG, Hartl D, Matsuura H, et al.

Endogenous IL-11 signaling is essential

in Th2- and IL-13-induced inflammation

and mucus production. Am J Respir Cell

Mol Biol 2008;39:739-46

24. Takaishi S, Tu S, Dubeykovskaya ZA,

et al. Gastrin is an essential cofactor for

helicobacter-associated gastric corpus

carcinogenesis in C57BL/6 mice.

Am J Pathol 2009;175:365-75

25. Judd LM, Alderman BM, Howlett M,

et al. Gastric cancer development in mice

lacking the SHP2 binding site on the

IL-6 family co-receptor gp130.

Gastroenterology 2004;126:196-207

26. Howlett M, Judd LM, Jenkins B, et al.

Differential regulation of gastric tumor

growth by cytokines that signal

exclusively through the coreceptor gp130.

Gastroenterology 2005;129:1005-18

27. Judd LM, Bredin K, Kalantzis A, et al.

STAT3 activation regulates growth,

inflammation, and vascularization in a

mouse model of gastric tumorigenesis.

Gastroenterology 2006;131:1073-85. Demonstrates the broad effects of

chronic STAT3 activation in a gastric

cancer model.

28. Howlett M, Menheniott TR, Judd LM,

et al. Cytokine signalling via gp130 in

gastric cancer. Biochim Biophys Acta

2009;1793:1623-33

29. Tu S, Bhagat G, Cui G, et al.

Overexpression of interleukin-1beta

induces gastric inflammation and cancer

and mobilizes myeloid-derived suppressor

A. S. Giraud et al.

898 Expert Opin. Ther. Targets [Early Online]

Exp

ert O

pin.

The

r. T

arge

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

oyal

Chi

ldre

ns H

ospi

tal o

n 07

/29/

13Fo

r pe

rson

al u

se o

nly.

Page 11: Targeting STAT3 in gastric cancer

cells in mice. Cancer Cell

2008;14:408-19

30. Oshima H, Oshima M, Inaba K, et al.

Hyperplastic gastric tumors induced by

activated macrophages in COX-2/

mPGES-1 transgenic mice. EMBO J

2004;23:1669-78

31. Oshima H, Matsunaga A, Fujimura T,

et al. Carcinogenesis in mouse stomach

by simultaneous activation of the Wnt

signaling and prostaglandin E2 pathway.

Gastroenterology 2006;131:1086-95

32. Itadani H, Oshima H, Oshima M, et al.

Mouse gastric tumor models with

prostaglandin E2 pathway activation

show similar gene expression profiles to

intestinal-type human gastric cancer.

BMC Genomics 2009;10:615

33. Tomita H, Takaishi S, Menheniott TR,

et al. Inhibition of gastric carcinogenesis

by the hormone gastrin is mediated by

suppression of TFF1 epigenetic silencing.

Gastroenterology 2011;140:879-91

34. Fox JG, Wang TC, Rogers AB, et al.

Host and microbial constituents

influence Helicobacter pylori-induced

cancer in a murine model of

hypergastrinemia. Gastroenterology

2003;124:1879-90

35. Spicer Z, Miller ML, Andringa A, et al.

Stomachs of mice lacking the gastric H,

K-ATPase alpha -subunit have

achlorhydria, abnormal parietal cells, and

ciliated metaplasia. J Biol Chem

2000;275:21555-65

36. Judd LM, Andringa A, Rubio CA, et al.

Gastric achlorhydria in H/K-ATPase-

deficient (Atp4a(-/-)) mice causes severe

hyperplasia, mucocystic metaplasia and

upregulation of growth factors.

J Gastroenterol Hepatol

2005;20:1266-78

37. Franic TV, van Driel IR, Gleeson PA,

et al. Reciprocal changes in trefoil 1 and

2 expression in stomachs of mice with

gastric unit hypertrophy and

inflammation. J Pathol 2005;207:43-52

38. Xu J, Song P, Miller ML, et al. Deletion

of the chloride transporter Slc26a9 causes

loss of tubulovesicles in parietal cells and

impairs acid secretion in the stomach.

Proc Natl Acad Sci USA

2008;105:17955-60

39. Song P, Groos S, Riederer B, et al.

KCNQ1 is the luminal K+ recycling

channel during stimulation of gastric acid

secretion. J Physiol 2009;587:3955-65

40. Sandhiya S, Dkhar SA. Potassium

channels in health, disease &

development of channel modulators.

Indian J Med Res 2009;129:223-32

41. Recalde S, Muruzabal F, Looije N, et al.

Inefficient chronic activation of parietal

cells in Ae2a,b(-/-) mice. Am J Pathol

2006;169:165-76

42. Heitzmann D, Warth R. No potassium,

no acid: K+ channels and gastric acid

secretion. Physiology (Bethesda)

2007;22:335-41

43. Lefebvre O, Chenard MP, Masson R,

et al. Gastric mucosa abnormalities and

tumorigenesis in mice lacking the

pS2 trefoil protein. Science

1996;274:259-62

44. Zavros Y, Eaton KA, Kang W, et al.

Chronic gastritis in the hypochlorhydric

gastrin-deficient mouse progresses to

adenocarcinoma. Oncogene

2005;24:2354-66

45. Tebbutt NC, Giraud AS, Inglese M,

et al. Reciprocal regulation of

gastrointestinal homeostasis by SHP2 and

STAT-mediated trefoil gene activation in

gp130 mutant mice. Nat Med

2002;8:1089-97.. First description of the

gp130757FF mouse.

46. Jenkins BJ, Grail D, Nheu T, et al.

Hyperactivation of Stat3 in

gp130 mutant mice promotes gastric

hyperproliferation and desensitizes

TGF-beta signaling. Nat Med

2005;11:845-52

47. Franic TV, Judd LM, Robinson D, et al.

Regulation of gastric epithelial cell

development revealed in H(+)/K(+)-

ATPase beta-subunit- and

gastrin-deficient mice. Am J Physiol

Gastrointest Liver Physiol

2001;281:G1502-11

48. Howlett M, Chalinor HV, Buzzelli JN,

et al. IL-11 is a parietal cell cytokine that

induces atrophic gastritis. Gut

2012; In press

49. Caruso R, Pallone F, Monteleone G.

Emerging role of IL-23/IL-17 axis in H

pylori-associated pathology.

World J Gastroenterol 2007;13:5547-51

50. Kabir S. The role of interleukin-17 in

the helicobacter pylori induced infection

and immunity. Helicobacter 2011;16:1-8

51. Gaffen SL. Biology of recently discovered

cytokines: interleukin-17--a unique

inflammatory cytokine with roles in bone

biology and arthritis. Arthritis Res Ther

2004;6:240-7

52. Zou W, Restifo NP. T(H)17 cells in

tumour immunity and immunotherapy.

Nat Rev Immunol 2010;10:248-56

53. Jarnicki A, Putoczki T, Ernst M. Stat3:

linking inflammation to epithelial cancer

- more than a "gut" feeling? Cell Div

2010;5:14

54. Nishihara M, Ogura H, Ueda N, et al.

IL-6-gp130-STAT3 in T cells directs the

development of IL-17+ Th with a

minimum effect on that of Treg in the

steady state. Int Immunol

2007;19:695-702

55. Caruso R, Fina D, Paoluzi OA, et al.

IL-23-mediated regulation of

IL-17 production in helicobacter

pylori-infected gastric mucosa.

Eur J Immunol 2008;38:470-8

56. Mitchell P, Germain C, Fiori PL, et al.

Chronic exposure to helicobacter pylori

impairs dendritic cell function and

inhibits Th1 development. Infect Immun

2007;75:810-19

57. Khamri W, Walker MM, Clark P, et al.

Helicobacter pylori stimulates dendritic

cells to induce interleukin-17 expression

from CD4+ T lymphocytes.

Infect Immun 2010;78:845-53

58. Flach CF, Ostberg AK, Nilsson AT,

et al. Proinflammatory cytokine gene

expression in the stomach correlates with

vaccine-induced protection against

Helicobacter pylori infection in mice:

an important role for

interleukin-17 during the effector phase.

Infect Immun 2011;79:879-86

59. Hatakeyama M. Oncogenic mechanisms

of the helicobacter pylori CagA protein.

Nat Rev Cancer 2004;4:688-94

60. Hatakeyama M, Higashi H. Helicobacter

pylori CagA: a new paradigm for

bacterial carcinogenesis. Cancer Sci

2005;96:835-43

61. Hatakeyama M. SagA of CagA in

helicobacter pylori pathogenesis.

Curr Opin Microbiol 2008;11:30-7

62. Kuipers EJ, Perez-Perez GI,

Meuwissen SG, et al. Helicobacter pylori

and atrophic gastritis: importance of the

cagA status. J Natl Cancer Inst

1995;87:1777-80

63. Ohnishi N, Yuasa H, Tanaka S, et al.

Transgenic expression of helicobacter

pylori CagA induces gastrointestinal and

hematopoietic neoplasms in mouse.

Targeting STAT3 in gastric cancer

Expert Opin. Ther. Targets [Early Online] 899

Exp

ert O

pin.

The

r. T

arge

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

oyal

Chi

ldre

ns H

ospi

tal o

n 07

/29/

13Fo

r pe

rson

al u

se o

nly.

Page 12: Targeting STAT3 in gastric cancer

Proc Natl Acad Sci USA

2008;105:1003-8

64. Zhu Y, Zhong X, Zheng S, et al.

Transformed immortalized gastric

epithelial cells by virulence factor

CagA of helicobacter pylori through Erk

mitogen-activated protein kinase

pathway. Oncogene 2005;24:3886-95

65. Stein M, Rappuoli R, Covacci A.

Tyrosine phosphorylation of the

helicobacter pylori CagA antigen after

cag-driven host cell translocation.

Proc Natl Acad Sci USA 2000;97:1263-8

66. Tsutsumi R, Higashi H, Higuchi M,

et al. Attenuation of helicobacter pylori

CagA x SHP-2 signaling by interaction

between CagA and C-terminal Src

kinase. J Biol Chem 2003;278:3664-70

67. Poppe M, Feller SM, Romer G, et al.

Phosphorylation of helicobacter pylori

CagA by c-Abl leads to cell motility.

Oncogene 2007;26:3462-72

68. Higashi H, Tsutsumi R, Muto S, et al.

SHP-2 tyrosine phosphatase as an

intracellular target of helicobacter pylori

CagA protein. Science 2002;295:683-6

69. Mimuro H, Suzuki T, Tanaka J, et al.

Grb2 is a key mediator of helicobacter

pylori CagA protein activities. Mol Cell

2002;10:745-55

70. Churin Y, Al-Ghoul L, Kepp O, et al.

Helicobacter pylori CagA protein targets

the c-Met receptor and enhances the

motogenic response. J Cell Biol

2003;161:249-55

71. Saadat I, Higashi H, Obuse C, et al.

Helicobacter pylori CagA targets PAR1/

MARK kinase to disrupt epithelial cell

polarity. Nature 2007;447:330-3

72. Murata-Kamiya N, Kurashima Y,

Teishikata Y, et al. Helicobacter pylori

CagA interacts with E-cadherin and

deregulates the beta-catenin signal that

promotes intestinal transdifferentiation in

gastric epithelial cells. Oncogene

2007;26:4617-26

73. Lee KS, Kalantzis A, Jackson CB, et al.

Helicobacter pylori CagA triggers

expression of the bactericidal lectin

REG3gamma via gastric

STAT3 activation. PLoS One

2012;7:e30786

74. Higashi H, Nakaya A, Tsutsumi R, et al.

Helicobacter pylori CagA induces

Ras-independent morphogenetic response

through SHP-2 recruitment and

activation. J Biol Chem

2004;279:17205-16

75. Suzuki M, Mimuro H, Suzuki T, et al.

Interaction of CagA with Crk plays an

important role in Helicobacter

pylori-induced loss of gastric epithelial

cell adhesion. J Exp Med

2005;202:1235-47

76. Bronte-Tinkew DM, Terebiznik M,

Franco A, et al. Helicobacter pylori

cytotoxin-associated gene A activates the

signal transducer and activator of

transcription 3 pathway in vitro and in

vivo. Cancer Res 2009;69:632-9

77. Lee IO, Kim JH, Choi YJ, et al.

Helicobacter pylori

CagA phosphorylation status determines

the gp130-activated SHP2/ERK and

JAK/STAT signal transduction pathways

in gastric epithelial cells. J Biol Chem

2010;285:16042-50

78. Higashi H, Yokoyama K, Fujii Y, et al.

EPIYA motif is a membrane-targeting

signal of Helicobacter pylori virulence

factor CagA in mammalian cells.

J Biol Chem 2005;280:23130-7

79. Matsumoto A, Isomoto H,

Nakayama M, et al. Helicobacter pylori

VacA reduces the cellular expression of

STAT3 and pro-survival Bcl-2 family

proteins, Bcl-2 and Bcl-XL, leading to

apoptosis in gastric epithelial cells.

Dig Dis Sci 2011;56:999-1006

80. Kim B, Bang S, Lee S, et al. Expression

profiling and subtype-specific expression

of stomach cancer. Cancer Res

2003;63:8248-55

81. Sneddon JB, Zhen HH, Montgomery K,

et al. Bone morphogenetic protein

antagonist gremlin 1 is widely expressed

by cancer-associated stromal cells and can

promote tumor cell proliferation.

Proc Natl Acad Sci USA

2006;103:14842-7

82. Namkoong H, Shin SM, Kim HK, et al.

The bone morphogenetic protein

antagonist gremlin 1 is overexpressed in

human cancers and interacts with

YWHAH protein. BMC Cancer

2006;6:74

83. Gallagher PG, Bao Y, Prorock A, et al.

Gene expression profiling reveals

cross-talk between melanoma and

fibroblasts: implications for host-tumor

interactions in metastasis. Cancer Res

2005;65:4134-46

84. Han JC, Zhang KL, Chen XY, et al.

Expression of seven gastric

cancer-associated genes and its relevance

for Wnt, NF-kappaB and Stat3 signaling.

APMIS 2007;115:1331-43

85. Deng JY, Sun D, Liu XY, et al.

STAT-3 correlates with lymph node

metastasis and cell survival in gastric

cancer. World J Gastroenterol

2010;16:5380-7

86. Deng J, Grande F, Neamati N. Small

molecule inhibitors of Stat3 signaling

pathway. Curr Cancer Drug Targets

2007;7:91-107

87. Yue P, Turkson J. Targeting STAT3 in

cancer: how successful are we?

Expert Opin Investig Drugs

2009;18:45-56

88. Mankan AK, Greten FR. Inhibiting

signal transducer and activator of

transcription 3: rationality and rationale

design of inhibitors. Expert Opin

Investig Drugs 2011;20:1263-75

89. Page BD, Fletcher S, Yue P, et al.

Identification of a non-phosphorylated,

cell permeable, small molecule ligand for

the Stat3 SH2 domain. Bioorg Med

Chem Lett 2011;21:5605-9

90. Trepicchio WL, Dorner AJ. The

therapeutic utility of Interleukin-11 in

the treatment of inflammatory disease.

Expert Opin Investig Drugs

1998;7:1501-4

91. Hayashi M, Rho MC, Enomoto A, et al.

Suppression of bone resorption by

madindoline A, a novel nonpeptide

antagonist to gp130. Proc Natl Acad

Sci USA 2002;99:14728-33

92. Underhill-Day N, McGovern LA,

Karpovich N, et al. Functional

characterization of W147A:

a high-affinity interleukin-11 antagonist.

Endocrinology 2003;144:3406-14

93. Dimitriadis E, Menkhorst E. New

generation contraceptives: interleukin

11 family cytokines as non-steroidal

contraceptive targets. J Reprod Immunol

2011;88:233-9

94. Nelson EA, Sharma SV, Settleman J,

et al. A chemical biology approach to

developing STAT inhibitors: molecular

strategies for accelerating clinical

translation. Oncotarget 2011;2:518-24

95. Park E, Park J, Han SW, et al. NVP-

BKM120, a novel PI3K inhibitor, shows

synergism with a STAT3 inhibitor in

human gastric cancer cells harboring

A. S. Giraud et al.

900 Expert Opin. Ther. Targets [Early Online]

Exp

ert O

pin.

The

r. T

arge

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

oyal

Chi

ldre

ns H

ospi

tal o

n 07

/29/

13Fo

r pe

rson

al u

se o

nly.

Page 13: Targeting STAT3 in gastric cancer

KRAS mutations. Int J Oncol

2012;40:1259-66

96. Iwamaru A, Szymanski S, Iwado E, et al.

A novel inhibitor of the STAT3 pathway

induces apoptosis in malignant glioma

cells both in vitro and in vivo.

Oncogene 2006;26:2435-2444

97. Gong W, Wang L, Yao JC, et al.

Expression of activated signal transducer

and activator of transcription 3 predicts

expression of vascular endothelial growth

factor in and angiogenic phenotype of

human gastric cancer. Clin Cancer Res

2005;11:1386-93

98. Kim DY, Cha ST, Ahn DH, et al.

STAT3 expression in gastric cancer

indicates a poor prognosis.

J Gastroenterol Hepatol 2009;24:646-51

99. Yakata Y, Nakayama T, Yoshizaki A,

et al. Expression of p-STAT3 in human

gastric carcinoma: significant correlation

in tumour invasion and prognosis.

Int J Oncol 2007;30:437-42

100. Lee J, Kang WK, Park JO, et al.

Expression of activated signal transducer

and activator of transcription 3 predicts

poor clinical outcome in gastric

adenocarcinoma. APMIS

2009;117:598-606

101. Cheong JH, Hong SY, Zheng Y, et al.

Eupatilin inhibits gastric cancer cell

growth by blocking STAT3-mediated

VEGF expression. J Gastric Cancer

2011;11:16-22

102. Chen J, Wang J, Lin L, et al. Inhibition

of STAT3 signaling pathway by nitidine

chloride suppressed the angiogenesis and

growth of human gastric cancer.

Mol Cancer Ther 2012;11:277-87

103. Uehara Y, Mochizuki M, Matsuno K,

et al. Novel high-throughput screening

system for identifying

STAT3-SH2 antagonists.

Biochem Biophys Res Commun

2009;380:627-31

104. Huang S, Chen M, Shen Y, et al.

Inhibition of activated Stat3 reverses

drug resistance to chemotherapeutic

agents in gastric cancer cells. Cancer Lett

2012;315:198-205

105. Zhu BH, Chen HY, Zhan WH, et al.

(-)-Epigallocatechin-3-gallate inhibits

VEGF expression induced by IL-6 via

Stat3 in gastric cancer. World J

Gastroenterol 2011;17:2315-25

106. Bang YJ, Van Cutsem E, Feyereislova A,

et al. Trastuzumab in combination with

chemotherapy versus chemotherapy alone

for treatment of HER2-positive advanced

gastric or gastro-oesophageal junction

cancer (ToGA): a phase 3, open-label,

randomised controlled trial. Lancet

2010;376:687-97

107. Deng N, Goh LK, Wang H, et al.

A comprehensive survey of genomic

alterations in gastric cancer reveals

systematic patterns of molecular

exclusivity and co-occurrence among

distinct therapeutic targets. Gut

2012;61:673-84

108. Tai WT, Cheng AL, Shiau CW, et al.

Dovitinib induces apoptosis and

overcomes sorafenib resistance in

hepatocellular carcinoma through

SHP-1-mediated inhibition of STAT3.

Mol Cancer Ther 2012;11:452-63

109. Lim CP, Fu XY. Multiple roles of

STAT3 in cardiovascular inflammatory

responses. Prog Mol Biol Transl Sci

2012;106:63-73

110. Haghikia A, Stapel B, Hoch M, et al.

STAT3 and cardiac remodeling.

Heart Fail Rev 2011;16:35-47

111. Obana M, Maeda M, Takeda K, et al.

Therapeutic activation of signal

transducer and activator of transcription

3 by interleukin-11 ameliorates cardiac

fibrosis after myocardial infarction.

Circulation 2010;121:684-91

112. Manukyan MC, Alvernaz CH,

Poynter JA, et al. Interleukin-10 protects

the ischemic heart from reperfusion

injury via the STAT3 pathway. Surgery

2011;150:231-9

113. Wegrzyn J, Potla R, Chwae YJ, et al.

Function of mitochondrial Stat3 in

cellular respiration. Science

2009;323:793-7

114. Murray PJ. Understanding and exploiting

the endogenous interleukin-10/STAT3-

mediated anti-inflammatory response.

Curr Opin Pharmacol 2006;6:379-86

115. Kuhn R, Lohler J, Rennick D, et al.

Interleukin-10-deficient mice develop

chronic enterocolitis. Cell

1993;75:263-74

116. Glocker EO, Kotlarz D, Boztug K, et al.

Inflammatory bowel disease and

mutations affecting the

interleukin-10 receptor. N Engl J Med

2009;361:2033-45

AffiliationAndrew S Giraud†1,2 PhD,

Trevelyan R Menheniott1 PhD &

Louise M Judd1 PhD†Author for correspondence1Murdoch Childrens Research Institute,

Royal Childrens Hospital,

Parkville, Australia2Research Director (Infection & Immunity),

Murdoch Childrens Research Institute,

Royal Childrens Hospital,

Flemington Rd,

Parkville, 3052, Australia

Tel: +61+3 8341 6446;

E-mail: [email protected]

Targeting STAT3 in gastric cancer

Expert Opin. Ther. Targets [Early Online] 901

Exp

ert O

pin.

The

r. T

arge

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y R

oyal

Chi

ldre

ns H

ospi

tal o

n 07

/29/

13Fo

r pe

rson

al u

se o

nly.