The emerging role of CXC chemokines in epithelial ovarian cancer

15
REPRODUCTION REVIEW The emerging role of CXC chemokines in epithelial ovarian cancer Adam Rainczuk, Jyothsna Rao, Jessica Gathercole and Andrew N Stephens Prince Henry’s Institute, Monash Medical Centre, Clayton, Victoria 3168, Australia Correspondence should be addressed to A N Stephens; Email: [email protected] A Rainczuk and J Rao contributed equally to this work Abstract In recent years, chemokines have generated intense investigations due to their involvement in both physiological and pathological processes of inflammation, particularly in ovarian biology. The physiological process of ovulation in the normal ovary involves various chemokines that mediate the healing of the ruptured endometrium. It is now being reported that many of these chemokines are also associated with the cancer of the ovary. Chronic inflammation underlies the progression of ovarian cancer; therefore, it raises the possibility that chemokines are involved in the inflammatory process and mediate immune responses that may favour or inhibit tumour progression. Ovarian cancer is a gynaecological cancer responsible for highest rate of mortality in women. Although there have been several investigations and advances in surgery and chemotherapy, the survival rate for this disease remains low. This is mainly because of a lack of specific symptoms and biomarkers for detection. In this review, we have discussed the emerging role of the CXC chemokines in epithelial ovarian cancer (EOC). The CXC group of chemokines is gaining importance in the field of ovarian cancer for being angiostatic and angiogenic in function. While there have been several studies on the angiogenesis function, emerging research shows that ELR K CXC chemokines, CXCL9 and CXCL10, are angiostatic. Importantly, the angiostatic chemokines can inhibit the progression of EOC. Given that there are currently no biomarkers or specific therapeutic targets for the disease, these chemokines are emerging as promising targets for therapy. Reproduction (2012) 144 303–317 Introduction As key mediators of the inflammatory response, chemokines represent an area of intense interest and study. Their influence on the chemotactic recruitment of leukocytes towards sites of inflammation is well established and involves multiple cell and tissue types (Charo & Ransohoff 2006). Apart from their established roles in chemotactic cell recruitment, however, there is a growing body of evidence suggesting that interference with their established functions may promote tumour development and metastasis (Mukaida & Baba 2012). In particular, a complex chemokine-signalling network may influence the development and progression of epithelial ovarian cancers (EOCs). The study of chemo- kines is therefore becoming important in furthering our understanding of this disease. An overview of the CXC chemokines and their functions Chemokines, comprising pairs of ligands and their associated receptors, are a superfamily of heparin- binding cytokine molecules with molecular weights between 8 and 10 kDa (Mukaida & Baba 2012). They are important mediators of the formation of new blood vessels by neovascularisation, involved in the angio- genic sprouting of vessels in both normal (e.g. embry- ogenesis and menstruation) and pathological states (Keeley et al. 2008). This role is achieved not solely through the promotion of angiogenesis but via a complex interplay between angiogenic and angiostatic signalling to influence the behaviour of the vascular endothelium (Kiefer & Siekmann 2011). Chemokine ligands exert their effects through their cognate G-protein-coupled receptors, a group of seven transmembrane-spanning proteins expressed on the cell surface and belonging to the class A rhodopsin-like family (Kiefer & Siekmann 2011; Fig. 1). On the basis of their primary structure, chemokines are classified as either C, CC, CXC or CX3C where ‘X’ represents a non-conserved amino acid substitution (Fig. 1). With the exception of the ‘C’ subgroup, all chemokines contain a common four-cysteine residue motif linked by disulphide bonds in conserved positions, one between the first and the third and one between the second and the fourth cysteine, to form triple-stranded b-sheet structures (Fernandez & Lolis 2002). The q 2012 Society for Reproduction and Fertility DOI: 10.1530/REP-12-0153 ISSN 1470–1626 (paper) 1741–7899 (online) Online version via www.reproduction-online.org

Transcript of The emerging role of CXC chemokines in epithelial ovarian cancer

Page 1: The emerging role of CXC chemokines in epithelial ovarian cancer

R

EPRODUCTIONREVIEW

The emerging role of CXC chemokines in epithelialovarian cancer

Adam Rainczuk, Jyothsna Rao, Jessica Gathercole and Andrew N Stephens

Prince Henry’s Institute, Monash Medical Centre, Clayton, Victoria 3168, Australia

Correspondence should be addressed to A N Stephens; Email: [email protected]

A Rainczuk and J Rao contributed equally to this work

Abstract

In recent years, chemokines have generated intense investigations due to their involvement in both physiological and pathological

processes of inflammation, particularly in ovarian biology. The physiological process of ovulation in the normal ovary involves various

chemokines that mediate the healing of the ruptured endometrium. It is now being reported that many of these chemokines are also

associated with the cancer of the ovary. Chronic inflammation underlies the progression of ovarian cancer; therefore, it raises the

possibility that chemokines are involved in the inflammatory process and mediate immune responses that may favour or inhibit tumour

progression. Ovarian cancer is a gynaecological cancer responsible for highest rate of mortality in women. Although there have been

several investigations and advances in surgery and chemotherapy, the survival rate for this disease remains low. This is mainly because of a

lack of specific symptoms and biomarkers for detection. In this review, we have discussed the emerging role of the CXC chemokines in

epithelial ovarian cancer (EOC). The CXC group of chemokines is gaining importance in the field of ovarian cancer for being angiostatic

and angiogenic in function. While there have been several studies on the angiogenesis function, emerging research shows that ELRK CXC

chemokines, CXCL9 and CXCL10, are angiostatic. Importantly, the angiostatic chemokines can inhibit the progression of EOC. Given

that there are currently no biomarkers or specific therapeutic targets for the disease, these chemokines are emerging as promising targets

for therapy.

Reproduction (2012) 144 303–317

Introduction

As key mediators of the inflammatory response,chemokines represent an area of intense interest andstudy. Their influence on the chemotactic recruitment ofleukocytes towards sites of inflammation is wellestablished and involves multiple cell and tissue types(Charo & Ransohoff 2006). Apart from their establishedroles in chemotactic cell recruitment, however, there is agrowing body of evidence suggesting that interferencewith their established functions may promote tumourdevelopment and metastasis (Mukaida & Baba 2012). Inparticular, a complex chemokine-signalling networkmay influence the development and progression ofepithelial ovarian cancers (EOCs). The study of chemo-kines is therefore becoming important in furthering ourunderstanding of this disease.

An overview of the CXC chemokines and their functions

Chemokines, comprising pairs of ligands and theirassociated receptors, are a superfamily of heparin-binding cytokine molecules with molecular weightsbetween 8 and 10 kDa (Mukaida & Baba 2012). They are

q 2012 Society for Reproduction and Fertility

ISSN 1470–1626 (paper) 1741–7899 (online)

important mediators of the formation of new bloodvessels by neovascularisation, involved in the angio-genic sprouting of vessels in both normal (e.g. embry-ogenesis and menstruation) and pathological states(Keeley et al. 2008). This role is achieved not solelythrough the promotion of angiogenesis but via a complexinterplay between angiogenic and angiostatic signallingto influence the behaviour of the vascular endothelium(Kiefer & Siekmann 2011). Chemokine ligands exert theireffects through their cognate G-protein-coupledreceptors, a group of seven transmembrane-spanningproteins expressed on the cell surface and belonging tothe class A rhodopsin-like family (Kiefer & Siekmann2011; Fig. 1).

On the basis of their primary structure, chemokinesare classified as either C, CC, CXC or CX3C where ‘X’represents a non-conserved amino acid substitution(Fig. 1). With the exception of the ‘C’ subgroup, allchemokines contain a common four-cysteine residuemotif linked by disulphide bonds in conserved positions,one between the first and the third and one between thesecond and the fourth cysteine, to form triple-strandedb-sheet structures (Fernandez & Lolis 2002). The

DOI: 10.1530/REP-12-0153

Online version via www.reproduction-online.org

Page 2: The emerging role of CXC chemokines in epithelial ovarian cancer

Figure 1 Chemokine ligands and receptor:intramolecular disulphide bonds form betweencysteine C1–C3 and C2–C4 pairs, leading to theformation of the characteristic triple-strandedb-sheet structures (with the exception of the ‘C’chemokine subgroup, which has only one C–Cpair). CXC chemokines are further groupedaccording to the presence of the ‘ELR’ Glu-Leu-Arg motif. Chemokines bind to their cognateG-protein-coupled receptors (GPCRs), comprisingseven transmembrane-spanning domains andbelonging to the class A rhodopsin-like family.

304 A Rainczuk, J Rao and others

nomenclature for chemokines is extended by theaddition of receptor ‘R’ (e.g. CXCR1) and respectiveligand ‘L’ (e.g. CXCL8). CXC chemokines are further sub-grouped according to the presence or absence of a threeamino acid ‘ELR’ Glu-Leu-Arg motif preceding the CXCsequence. Thus, the CXC chemokines are often referredto as ELRC or ELRK (Strieter et al. 2004).

Over the past two decades, many studies haveexamined the dual roles of CXC chemokines in boththe promotion and the inhibition of angiogenesis. Theangiogenic sprouting of new, thin-walled capillarynetworks from pre-existing ones is a critical factor intumour growth and spread (Keeley et al. 2008, Kiefer &Siekmann 2011). As a general rule, ELRC CXCchemokines mediate angiogenesis, typically viainteraction with the CXCR2 receptor (see Table 1 for alisting of CXC chemokines and their cognate receptors)and are also involved in other diverse functions such asstem cell migration, lymphocyte migration and lym-phoid tissue neogenesis (Romagnani et al. 2004, Kiefer &Siekmann 2011). CXCR2 is typically expressed onendothelial cells but can be constitutively expressed byspecific tissues and cells (Table 1; Mukaida & Baba2012). By contrast, the ELRK CXC chemokines typicallypromote angiostasis (rather than angiogenesis), throughtheir common receptor CXCR3 (Mukaida & Baba 2012).Chemokine ligands lacking the ELR motif can promote

Reproduction (2012) 144 303–317

anti-tumour effects by recruiting both innate andadaptive immune effector cells (in the case of CXCL9and CXCL10 that can be secreted from tumour cells forexample) and can also inhibit endothelial cell migrationand proliferation (Keeley et al. 2008, Kiefer & Siekmann2011, Whitworth & Alvarez 2011).

There are, however, two exceptions to the rule. CXCL4is an ELRC chemokine, which also binds to CXCR3.Unlike the other ELRC members, CXCR4 promotesangiostasis similar to the ELRK chemokines. Contrast-ingly, the ELRK chemokine CXCL12 binds to the CXCR4receptor to promote angiogenesis (Kiefer & Siekmann2011). Therefore, both the ELR status and the cognatereceptor for each chemokine must be considered whenevaluating the function (either angiogenic or angiostatic)of the CXC chemokine family. The functions of the CXCchemokines are also extensively regulated through post-translational modification. This is a well-establishedmechanism for regulating their bioactivity and functionand has been reviewed elsewhere (Mortier et al. 2011).

It is now well established that besides leukocytes,fibroblasts and endothelial cells, tumour cells alsoexpress chemokines and their receptors (Balkwill 2004,Romagnani et al. 2004, Kiefer & Siekmann 2011,Mukaida & Baba 2012). Importantly, chemokines aresecreted by both the stromal and the malignant part ofthe cancer tissue, and functionally, the chemokine

www.reproduction-online.org

Page 3: The emerging role of CXC chemokines in epithelial ovarian cancer

Table

1G

ener

alove

rvie

wof

CX

Cch

emoki

nes

invo

lved

inth

ere

gula

tion

of

angi

oge

nes

isan

dly

mphocy

tere

cruitm

ent.

CXCL

ELR

Previous

nomen

clature

CXCR

Rec

eptor

expression

Function

Gen

eral

origin

Commondisea

seReferen

ces

CX

CL1

CX

CL2

CX

CL3

CG

ROa

/MG

SA-a

,G

ROb/M

GSA

-b,

GR

Og

/MG

SA-g

CX

CR

2Ep

i/en

do

Chem

ota

xis

of

neu

trophil

s.En

doth

elia

lce

llm

igra

tion

and

pro

life

rati

on

Neu

trophils

Mel

anom

a,ga

stri

can

dco

l-ore

ctal

cance

rs,

ova

rian

Cuen

caet

al.

(1992),

Wuyt

set

al.

(1999)

and

Eck

etal

.(2

003)

CX

CL4

CPF4

CX

CR

3B

Endo

Anti

-angi

oge

nic

,in

hib

its

tum

our

vasc

ula

risa

tion

Thym

us,

nat

ura

lki

ller

cell

,m

onocy

tes

Mel

anom

a,ova

rian

cance

rG

ott

lieb

etal

.(1

988),

Lasa

gni

etal

.(2

003),

Stru

yfet

al.

(2007),

Cal

lese

net

al.

(2010)

and

Ha

etal

.(2

011)

CX

CL5

CEN

A-7

8C

XC

R2

Epi/

endo

Angi

oge

nes

is,

pro

mote

stu

mour

grow

than

dm

etas

tasi

s

Neu

trophils

Lung,

pan

crea

tic,

ova

rian

cance

rsW

isle

zet

al.

(2004),

Furu

yaet

al.(

2007,2

011),

Fric

ket

al.

(2008)

and

Liet

al.

(2011)

CX

CL6

CG

ranulo

cyte

chem

o-

tact

icpro

tein

2C

XC

R1,

CX

CR

2Ep

i/en

do

Angi

oge

nes

isN

eutr

ophils

Gas

troin

test

inal

tum

our

Gij

sber

set

al.

(2005)

CX

CL7

CN

AP-2

,PB

PC

XC

R2

Epi/

endo

Med

iate

san

gioge

nes

isvi

aM

APK

and

mTO

Rpat

hw

ay

Neu

trophils,

pla

tele

ts,

monocy

tes

Pancr

eati

cca

nce

rsM

atsu

bar

aet

al.

(2011)

CX

CL8

CIL

8C

XC

R1,

CX

CR

2Ep

i/en

do

Neu

trophil

chem

oat

trac

-ta

nta

nd

activa

ting

fact

or.

Angi

oge

nic

agen

t

Neu

trophils

Gas

tric

carc

inom

a,ova

rian

cance

rEc

ket

al.

(2003)

and

Yig

itet

al.

(2011)

CX

CL9

–M

onoki

ne

induce

dby

IFN

-g(M

IG)

CX

CR

3En

do

Att

ract

tum

our-

infilt

rating

lym

phocy

tes,

inhib

iten

doth

elia

lce

llm

igra

tion

and

pro

life

r-at

ion

Thym

us,

nat

ura

lki

ller

cell

,w

ides

pre

adG

astr

ic,

colo

rect

alca

nce

rsM

ush

aet

al.

(2005)

and

Ohta

ni

etal

.(2

009)

CX

CL1

0–

IFN

-g-i

nduci

ble

pro

tein

10

(IP10)

CX

CR

3En

do

Pro

mote

anti

-tum

our

effe

cts

by

attr

acting

T-ly

mphocy

tes

Thym

us,

nat

ura

lki

ller

cell

,w

ides

pre

adG

astr

ic,

colo

rect

al,

endo-

met

rial

,ova

rian

cance

rsM

ush

aet

al.

(2005),

Son

etal

.(2

007),

Ohta

ni

etal

.(2

009)

and

Smir

nova

etal

.(2

012)

CX

CL1

1–

IFN

-induci

ble

T-ce

lla

chem

oat

trac

tant

(I-T

AC

)

CX

CR

3,

CX

CR

7En

do/e

pi

Inhib

iten

doth

elia

lce

llm

igra

tion

and

pro

life

r-at

ion

Thym

us,

nat

ura

lki

ller

cell

Cle

arce

llova

rian

cance

rsFu

ruya

etal

.(2

011)

CX

CL1

2–

SDF-

1a

/bC

XC

R4,

CX

CR

7En

do/e

pi

Neo

vasc

ula

risa

tion

duri

ng

foet

alorg

anoge

nes

is.

Tum

our

cell

mig

rati

on

and

pro

life

rati

on

Wid

espre

adB

reas

t,co

lore

ctal

carc

inom

a,en

dom

etri

al,

ova

rian

can-

cers

Jian

get

al.

(2006),

Aki

shim

a-Fu

kasa

wa

etal

.(2

009),

Gel

min

iet

al.

(2009)

and

Has

san

etal

.(2

009)

CX

CL1

3C

BLC

/BC

A-1

CX

CR

5–

Mig

rati

on

of

T-ce

lls

Bce

lls

Hum

anlu

ng

cance

rde

Chai

sem

arti

net

al.

(2011)

CX

CL1

4C

BR

AK

/bole

kine

Unkn

ow

nU

nkn

ow

nM

igra

tion

of

T-ce

lls,

che-

moat

trac

tion

of

NK

cell

sin

epit

hel

ium

of

the

endom

etri

um

Monocy

tes

Endom

etri

alca

nce

rva

nder

Hors

tet

al.

(2012)

CX

CL1

6C

SR-P

SOX

OX

;C

XC

LG16;

SR-P

SOX

CX

CR

6Ep

iSo

luble

CX

CL1

6en

han

ces

pro

life

rati

on

and

mig

ration.

Tran

s-m

embra

ne

CX

CL1

6in

hib

its

pro

life

rati

on

Act

ivat

edT-

cells

Gli

om

a,co

lore

ctal

carc

i-nom

a,re

nal

cance

r,pro

s-ta

teca

nce

r,bre

ast

cance

r

Ludw

iget

al.(2

005),

Hojo

etal

.(2

007),

Dar

ash-Y

ahan

aet

al.

(2009),

Gutw

ein

etal

.(2

009)

and

Den

get

al.

(2010)

Epi,

epit

hel

ial

cell

expre

ssio

n;

Endo,

endoth

elia

lce

llex

pre

ssio

n.

CXC chemokines in epithelial ovarian cancer 305

www.reproduction-online.org Reproduction (2012) 144 303–317

Page 4: The emerging role of CXC chemokines in epithelial ovarian cancer

306 A Rainczuk, J Rao and others

ligand and receptor pairs can exert a direct effect ontumour proliferation and survival (Erreni et al. 2009,Ha et al. 2011, Boimel et al. 2012). The chemokinesfrom stromal cells may influence the survival ofmalignant cells by binding to the functional receptorsacquired on the cancerous cells, enhancing metastasis ina chemokine-rich environment (Mantovani et al. 2010,Balkwill 2012).

This review will discuss the emerging roles of theCXC chemokines in the pathogenesis of EOC, in thecontext of their inflammatory, angiogenic and metastaticproperties.

The role of CXC chemokines in normal ovarianphysiology

The surface epithelium of the ovary is a natural extensionof the peritoneal lining and is directly exposed to anymetabolic and environmental stress present in theperitoneal cavity (Maccio & Madeddu 2012). Regularcycling and release of ova from the ovary causes theovarian epithelium to undergo regular cycles of wound-ing and repair throughout a female’s reproductive life.This process is generally considered to induce aninflammatory response and triggers (Murdoch et al.2010) the secretion of a large number of chemokines,cytokines, enzymes and growth factors (Murdoch &McDonnel 2002).

A model of CXC chemokine-coordinated regulation ofangiogenesis and inflammation after wounding of anormal epithelial cell layer is summarised in Fig. 2(Griffioen & Molema 2000, Spinetti et al. 2001,Romagnani et al. 2004), and a more detailed review ofwound repair involving chemokines can be found inRomagnani et al. (2004). At the wound site, plateletsrelease growth factors including vascular endothelialgrowth factor (VEGF), platelet-derived growth factor andvarious cytokines including CXCL7, CXCL1, CXCL5(which initiate neutrophil recruitment) and CXCL4(to assist in blood-clot formation) (Griffioen & Molema2000, Spinetti et al. 2001, Romagnani et al. 2004).Wounded epithelial cells then express CXCL8, inducingan inflammatory response and resulting in angiogenesisand the sprouting of new blood vessels with high CXCR2expression (Romagnani et al. 2004). The epithelial layeralso produces angiostatic CXCL9, -10 and -11, arrestingthe migration of proliferating CXCR3C endothelial cellsand leading to lymphocyte recruitment during the periodof healing (Romagnani et al. 2004). CXCL8 is alsoinvolved in re-epithelialisation of the wound site (notshown in Fig. 2; Romagnani et al. 2004). Other non-CXCchemokines are also involved in wound healing; forexample, CCL2 expression during wound healing alsorecruits CCR2C macrophages and monocytes to theinflamed ovarian epithelium (Fader et al. 2010). The roleof non-CXC chemokines in this process is reviewedelsewhere (Charo & Ransohoff 2006).

Reproduction (2012) 144 303–317

Chemokines and cytokines also modulate ovarianfunction via their involvement in follicular developmentand ovulation and the formation, function and death ofthe corpus luteum (Auersperg et al. 1998). CXCL8, forexample, is significantly elevated in normal folliculo-genesis, suggesting a role in mediating neutrophilattraction (Ness & Modugno 2006). Moreover, anti-CXCL8 antiserum can suppress hCG-induced ovulationand neutrophil activity in women undergoing IVF (Ness& Modugno 2006). CXCL8 levels also correlate withcollagenases, which are crucial to the remodelling ofthe cervical matrix (Auersperg et al. 1998). CXCL1,another neutrophil attractant, is present in periovulatoryfollicular fluid and is expressed by ovarian stromalcells. On completion of ovulation, the rupturedepithelium undergoes repair to form the corpus luteum,characterised by significantly increased infiltration ofleukocytes and modulated by pro-angiogenic factors inthe follicular fluid such as CXCL8 and CCL2 (Merritt &Cramer 2011).

Recent studies have shown that CXCL12 is expressedin the cells of the normal ovarian surface epithelium andthe epithelium of the fallopian tube (Machelon et al.2011). Interestingly, CXCL12 is absent in the follicles andthe oocytes (Machelon et al. 2011, Merritt & Cramer2011). Constitutive expression of CXCL10 has also beenreported in normal ovary, with cyclic increases observedduring ovulation (Wong et al. 2002, Zhou et al. 2004).While the precise roles played by these chemokines inthe physiology of the ovary are still unclear, theirpresence firmly establishes their importance in normalovarian function.

Epithelial ovarian cancer

EOCs account for between 80 and 95% of all ovariantumours diagnosed and exhibit the highest mortality rateof any gynaecological tumour type (Roett & Evans 2009).EOCs are commonly classified according to histopatho-logical appearance as clear cell, endometrioid, mucinousor serous; amongst these, serous epithelial tumoursare the most commonly diagnosed tumour type (Kobelet al. 2008). However, wide varieties of less commonovarian neoplasms exist and can include mixed, undiffer-entiated or Brenner-type tumours (Kaku et al. 2003,Arnogiannaki et al. 2011). While traditionally sub-grouped according to FIGO stage and grade, a growingbody of evidence now suggests that low- and high-gradetumours have different aetiologies arising from unrelated,underlying molecular pathologies (Gilks et al. 2008,Kobel et al. 2008). Low-grade tumours typically exhibitmicropapillary structures and originate from borderlinetumours. These tumours are believed to progress throughbenign and borderline stages to malignancy and aregenerally associated with good prognosis. They also havewell-defined mutational pathways and often expressmutated KRAS or BRAF (Cho 2009). By contrast, a

www.reproduction-online.org

Page 5: The emerging role of CXC chemokines in epithelial ovarian cancer

Figure 2 An overview of CXC chemokine involvement in normal wound repair (expanded details found in Romagnani et al. (2004)). (A) Generationof blood clot and neutrophil recruitment: (A1) growth factors, cytokines and chemokines are released by clotted platelets, while CXCL4, -5 and -7initiate neutrophil recruitment to the wound site; (A2) epithelial cells at the wound site express CXCL8, while endothelial cells express CXCL1; (A3)neutrophils expressing CXCR2 migrate into the wound site along the CXCL1/CXCL8 chemotactic gradient. (B) Recruitment of lymphocytes andmonocytes: (B1) epithelial cells continue to express CXCL8, promoting the growth of new blood cells. Epithelial cells also express CXCL9, -10 and-11 to limit and regulate endothelial cell proliferation and new blood vessel growth; (B2) CXCL9 and -10 recruit lymphocytes during wound healing;(B3) ongoing CXCL1 expression by endothelial cells promotes continued neutrophil recruitment, while CXCL9, -10 and -11 regulate blood vesselgrowth and the migration of proliferating endothelial cells; (B4) CXCL4 helps promote and maintain blood clotting.

CXC chemokines in epithelial ovarian cancer 307

significantly large number of TP53 gene mutations havebeen found in high-grade serous tumours (Milner et al.1993), and they also have a poorly defined mutationalpathway with mutated TP53 expression found in up to80% of cases (Vang et al. 2009). They appear to arisespontaneously, metastasise early in their progression andare associated with poor prognosis (Cho 2009, Cho &Shih Ie 2009, Roett & Evans 2009).

www.reproduction-online.org

Balancing inflammation, T-cell recruitment,angiogenesis and angiostasis: the role of chemokinesin ovarian cancer development and progression

Inflammation and the ovarian surface epithelium

Similar to other tumour types, chronic inflammation isan important condition underlying the growth andprogression of ovarian tumours (Ness & Cottreau 1999,

Reproduction (2012) 144 303–317

Page 6: The emerging role of CXC chemokines in epithelial ovarian cancer

308 A Rainczuk, J Rao and others

Ness et al. 2000, Fleming et al. 2006, Son et al. 2007,Coffelt & Scandurro 2008, Maccio & Madeddu 2012). Askey mediators of inflammation, chemokines play twomajor roles: i) they are responsible for the recruitmentand activation of immune cells into sites of malignancy(and possibly areas of pre-neoplastic growth) and ii) theymediate pro- and anti-angiogenic effects, processesrequired for the vascularisation and progression oftumours. In addition, the multiple biological effectsexerted by CXC chemokines mean that each of theseprocesses is inextricably linked. Therefore, it is ofparamount importance to identify molecules that areinvolved and their contribution towards anti-tumouractivity and in preventing recurrence – or, conversely,how their activity may promote tumour growth andmetastasis.

Although an accepted view is that a majority ofovarian cancer cases originate from the ovarian surfaceepithelium, emerging evidence suggests the involvementof the fimbriae of fallopian tubes. Crum et al. showedthat fallopian tubes and ovaries from women with BRCAmutations often contained precursor lesions in thefimbriae of the tube (Medeiros et al. 2006, Crum et al.2007). By contrast, Clarke et al. have demonstrated anassociation between BRCA and inflammation in serousovarian carcinomas. BRCA mutations (or epigenetic loss)were associated with favourable prognosis via recruit-ment of intraepithelial CD8C (and to a lesser extentCD3C) tumour-infiltrating lymphocytes (TILs). This novelassociation between intraepithelial T-cell infiltration andBRCA mutation in serous EOC has also been observedwith certain breast cancer types and can lead to afavourable patient prognosis (Clarke et al. 2009).Chronic inflammation from diseases such as hepatitis,human papilloma virus in the cervix and ulcerativecolitis are well known to cause cancer of the tube(Demopoulos et al. 2001). Similarly, it has beensuggested that proliferation and hyperplasia in thefallopian tube are associated with lesions that may becarcinogenic in nature (Moore & Enterline 1975).Compelling evidence for this hypothesis was recentlypresented by Kim et al. (2012) who demonstrated, usinga Dicer–Pten knockout mouse, that serous epithelialtumours can arise in the fallopian tube and rapidlymetastasise to the ovary. Mice in this study exhibited a100% mortality rate from tumours that bore a strikingresemblance to clinical disease (Kim et al. 2012).

Retrograde menstruation via the fallopian tube is acommon condition, where the flow of endometrial fluidbathes the tubes with inflammatory molecules such asCXCL8 (interleukin-8 (IL8)), tumour necrosis factor alpha(TNF-a) and granulocyte–macrophage colony-stimulat-ing factor (GM–CSF). Each of these are elevated in ovariancarcinomas (Strandell et al. 2004). It is well establishedthat a combination of tubal ligation and hysterectomy isprotective against ovarian cancer (Green et al. 1997,Seidman et al. 2002). Together, these findings strongly

Reproduction (2012) 144 303–317

support the notion that inflammation in the fallopiantube, the ovarian epithelium or both are contributingfactors in the development of ovarian cancer.

The inflammation-mediated release of chemokines,cytokines and growth factors can also promote migrationand proliferation of leukocytes, epithelial and endo-thelial cells (Fig. 2; Mukaida & Baba 2012). At amolecular level, there is evidence that pro-inflammatorycytokines such as IL1, IL6 and TNF-a are linked to a poorprognosis in EOC patients (Clendenen et al. 2011,Maccio & Madeddu 2012). Oxidative stress, cell necrosisand rapid cell division are hallmarks of chronicinflammation (Ness & Modugno 2006). Rapid celldivision gives rise to a possibility of replication errorand the need for DNA repair, such as at the key regulatorysite TP53, increasing the risk of mutagenesis. This isfurther supported by the high rate of TP53 mutationcommonly observed in high-grade serous EOC (Merritt &Cramer 2011).

Recently, inflammatory cytokine production in thehuman ovarian cancer microenvironment has beendescribed in terms of an autocrine cytokine network.The TNF network includes TNF-a, IL6 and CXCL12(Kulbe et al. 2012); in particular, the CXCL12/CXCR4axis plays a major role in the proliferation and metastasisof ovarian tumour cells (Barbieri et al. 2010). EnhancedTNF network activity has been associated with genesinvolving angiogenesis, inflammation and leukocyteinfiltration. Targeting members of the TNF network viasiRNA or neutralising antibodies reduced experimentalperitoneal ovarian tumour growth, as well as angiogen-esis in human ovarian tumour biopsies and mousexenograft models (Kulbe et al. 2012).

The nuclear factor-kB (NF-kB) pathway is also import-ant in modulating autocrine and paracrine signallingbetween inflammatory markers in the cancer microenvir-onment (Son et al. 2007). Notably, NF-kB regulates theCXCL12/CXCR4 autocrine signalling in ovarian cancercell lines (Miyanishi et al. 2010). Over-expression ofprostaglandins, leading to altered expression of cytokinesand chemokines, increases the invasiveness of ovariantumour cells (and other tumour types) (Aitokallio-Tallberget al. 1988, Subbaramaiah et al. 1997, Ness & Modugno2006). The expression of cytokines and chemokines ininflamed malignancies of the female genital tract may,therefore, lead to dysfunctional autocrine loops, resultingin increased tumour progression and invasion.

Recruitment of T-cells to epithelial ovarian tumours

In the normal anti-tumour immune response,interactions between CXCL9 (MiG), CXCL10 (IP10) andtheir receptor CXCR3 influences the chemoattraction ofnatural killer (NK) cells, activated Th1 cells, gamma-delta T-cells, macrophages and dendritic cells towardstumours (Strieter et al. 2006, Whitworth & Alvarez2011). The importance of CXCL9 and CXCL10 to

www.reproduction-online.org

Page 7: The emerging role of CXC chemokines in epithelial ovarian cancer

CXC chemokines in epithelial ovarian cancer 309

generate an anti-tumour response is supported by theirroles as mediators of IL12, a potent immunoregulatorycytokine (also being trialled as a therapeutic agent inovarian cancer patients, although with modest results)(Whitworth & Alvarez 2011). Additionally, in aninvestigation into links between coagulation and inflam-mation, it was shown that culture of cells derived fromovarian cancer ascites and peripheral blood mono-nuclear cells (PBMCs) from healthy donors also inducedsignificant up-regulation of inflammatory cytokines(IL1b and IL6), along with CCL2 and CXCL8 (Naldiniet al. 2011). This suggests that the ovarian cancermicroenvironment is inflammatory in nature and candirect PBMCs to increase the release of inflammatorychemokines and cytokines.

Recent gene expression studies identified CXCL10,CXCL11 and their receptor CXCR3 as amplified in asubset of ovarian cancer patients from a sample of 489high-grade, serous epithelial tumours. These studiesdefined this group of tumours as ‘immunoreactive’(CGARN 2011), suggesting an important role for thesechemokines in the tumour microenvironment. A primaryfunction for these chemokine ligands is the trafficking ofleukocytes and the recruitment of activated CD4C Th1cells, CD8CT-cells and NK cells towards sites ofinflammation (Clark-Lewis et al. 2003). High concen-trations of CXCL9 and CXCL10 are also associated withthe presence of TILs, and this is a positive predictor ofpatient outcome with patients exhibiting lower recur-rence, less dissemination and longer survival (Zhanget al. 2003, Sato et al. 2005, Tomsova et al. 2008,Kryczek et al. 2009). By contrast, negative patientoutcomes are associated with tumour infiltration byregulatory T-cells (Treg) that mediate immune tolerance(Curiel et al. 2004, Sato et al. 2005, Kryczek et al. 2009).Sato et al. (2005) demonstrated that high intraepithelial

www.reproduction-online.org

CD8C/CD4CT-cell and CD8CT-cell/Treg ratios resultedin improved survival in EOC. Moreover, neitherintraepithelial CD4C or other CD3C TILs alone wereassociated with survival in this study (Sato et al. 2005).Others have more recently shown that longer survival ispositively correlated with both CD8C and CD3C TILs(Hwang et al. 2012). However, CD8C positive TILsshowed a more consistent association with patientsurvival in this meta-analysis (Hwang et al. 2012).

EOC tissue and ascites fluid contain high levels ofCD4C CD25C FOXP3C Treg cells, which are not presentin normal ovarian tissues (Curiel et al. 2004). FOXP3 is amarker of Treg cells, crucial for maintaining immunologichomoeostasis by preventing autoimmunity in a normalimmune environment (Kelley & Parker 2010). In a studyof immune cell infiltration into tumours, a high level ofthe chemokine CCL22 (secreted by tumour-associatedmacrophages) was detected in tumour ascites, possiblyinducing Treg trafficking into tumours and suppressingthe response of TILs (Curiel et al. 2004). In an apparentcontradiction, however, under certain conditions in thetumour microenvironment, CXCL9, CXCL10 and thecytokine IL17 can also be produced by tumour-associated macrophages and contribute to positiveoutcomes in EOC (Kryczek et al. 2009).

An explanation as to why TILs are effective atimproving EOC patient outcomes can be partiallyattributed to the recent identification of T helper17 (Th17) cells. These cells can contribute to pathogenesisof autoimmune disease and initiate tumour growth incertain models but drive anti-tumour immunity in someepithelial malignancies (Zou & Restifo 2010, Wilke et al.2011). Th17 cells are postulated not to be immunosup-pressive, as they do not express FOX3P (Wilke et al.2011). An important study by Kryczek et al. (2009)showed that in addition to NK cells and Th1 effector

Figure 3 Promotion of a favourable outcome inEOC involving CXC chemokines. (1) Th17 cells(without Treg suppression in this example) secreteIL17 and anti-tumour promoting IFN-g, afterinduction by tumour-associated macrophagessecreting IL1b (Kryczek et al. 2009); (2) IL17secretion and IFN-g induce tumour cells tosecrete CXCL9 and CXCL10; (3) angiostaticchemokines establish a gradient whereby adap-tive and innate immune cells migrate to thetumour site. A high CD8CT-cell to low CD4CT-cell/Treg ratio can lead to positive outcomes inEOC (Sato et al. 2005).

Reproduction (2012) 144 303–317

Page 8: The emerging role of CXC chemokines in epithelial ovarian cancer

310 A Rainczuk, J Rao and others

T-cells, Th17 cells secreting IFN-g and IL17 werecorrelated with the elevation of Th1-type chemokinesCXCL9 and CXCL10 in the ovarian cancer microenvir-onment. CXCL9 and CXCL10 were produced by primaryovarian cancer cells and macrophages, leading to therecruitment of CXCR3-expressing effector CD8CT-cellsand NK cells to the tumour. A significant association wasfound with the presence of IL17 in patient ascites, withhigh IL17 levels (median patient survival 78 months)significantly improving survival time compared with lowIL17 levels (median patient survival 27 months) (Kryczeket al. 2009). The levels of CXCL9 and CXCL10 weredirectly correlated with tumour and tumour-infiltratingNK cells and CD8CT-cells, highlighting the importanceof these ELR-chemokines in limiting the spread of EOC.Figure 3 attempts to summarise the effect of increasedCXCL9 and CXCL10 concentrations in the EOC micro-environment, which are reported to lead to a positivesurvival outcome.

Angiostatic chemokine ligands: CXCL9 and CXCL10expression can inhibit EOC progression

The ELRK chemokines CXCL9, CXCL10 and CXCL11can influence tumour progression through their angio-static effects (Strieter et al. 2006). Primary ovarian cancercells stimulated with IL17 and IFN-g can be induced tosecrete CXCL9 and CXCL10 (Kryczek et al. 2009), whileCXCL11 (and CXCL9) expression has been detected inserous ovarian carcinoma cell lines and tissue byimmunohistochemistry and reverse transcriptase PCR(Furuya et al. 2007). These CXCR3-binding chemokinescan inhibit endothelial cell migration and proliferation,attract cells involved in innate and adaptive immunityand increase MHC class I processing by tumour cells,

Reproduction (2012) 144 303–317

thereby enhancing recognition by the immune system(Groom & Luster 2011, Wilke et al. 2011; Fig. 3).

CXC-mediated regulation of the EOC microenviron-ment appears to be primarily due to CXCL9 and CXCL10;the anti-tumour effect of CXCL11 in EOC is reportedlyless efficient in mounting an anti-tumour response(Lasagni et al. 2003, Furuya et al. 2007). Similar to theexpression of CXCL9 and CXCL10 ligands, increasedexpression of CXCR3 has been reported on the surface ofEOC cells (Furuya et al. 2007) and clear cell ovariancancer cells (Furuya et al. 2011). CXCR3 has been foundto be expressed by endothelial cells while undergoingG2M and late S phases of the cell cycle (Romagnani et al.2001). Although the expression of CXCR3 in multiplecell types and specific cell cycle phases have beenidentified (Romagnani et al. 2001), their biologicalrelevance has yet to be conclusively demonstrated(Lasagni et al. 2003, Giuliani et al. 2006, Gacci et al.2009, Campanella et al. 2010).

Angiogenic chemokine ligands can promote tumourdevelopment

Pathological angiogenesis involves multiple cell typesand growth factors and is observed in wound healing,arthritis and diabetic retinopathy. It is closely related tochronic inflammation and is an important event in theprogression and spread of cancer (Keeley et al. 2008).ELRC chemokines including CXCL8 (IL8), CXCL5(ENA-78) and CXCL1 (GRO-a) are known to induceendothelial migration and proliferation through bindingto CXCR2, leading to metastatic spread (Mukaida &Baba 2012). They can also act indirectly via theexpression of CXCR2 on leukocytes, leading to thesecretion of pro-angiogenic factors such as VEGF(Kiefer & Siekmann 2011). All three CXC chemokines

Figure 4 Modification of CXCL5 increaseschemotactic potency and neutrophil recruit-ment. (1) As in normal wound healing, neutro-phils containing CXCR2 continue to migrate tothe wound site towards CXCL1 and -8;(2) neutrophils secrete cathepsin G and cleaveCXCL5, increasing chemotactic potency.(3) Enhanced CD11b/CD18 expression on theneutrophil surfaces increases adhesion toendothelial tumour cells and initiatesremodelling of the endothelium, making it easierfor tumour cells to metastasise.

www.reproduction-online.org

Page 9: The emerging role of CXC chemokines in epithelial ovarian cancer

CXC chemokines in epithelial ovarian cancer 311

have the ability to attract polymorphonuclear leukocytesincluding neutrophils (Mukaida & Baba 2012).

The significance of inflammation in the potentialinitiation and progression of EOC makes CXCL5 apotentially important (and largely unreported) factor inthis disease. CXCL5 is expressed in both lung andpancreatic cancer and is up-regulated in endometriosisand ovarian carcinoma where it can attract neutrophils(Wislez et al. 2004, Furuya et al. 2007, Frick et al. 2008,Li et al. 2011). Once present, neutrophils secrete a broadspectrum of proteases including cathepsin G that cancleave CXCL5; N-terminal truncation of CXCL5 signi-ficantly enhances the chemotactic potency of CXCL5both in vitro and in vivo (Wuyts et al. 1999, Mortier et al.2010). Neutrophils isolated from the peripheral blood ofovarian cancer patients also produce increased reactiveoxygen species and display increased adhesion toovarian cancer cells through enhanced CD11b/CD18 expression on their cell surfaces (Klink et al.2008). The adhesion of neutrophils to endothelial cellsinitiates remodelling of the endothelium, making iteasier for tumour cells to metastasise (Wu et al. 2001,De Larco et al. 2004, Klink et al. 2008). Figure 4 attemptsto summarise the influence of neutrophils and modifiedCXCL5 in the metastasis of ovarian tumour cells.

Both CXCL8 (Schutyser et al. 2002) and CXCL1(Bolitho et al. 2010) are up-regulated in ascites fluid,while CXCL8 protein and anti-CXCL8 antibodies are alsopresent in serum from advanced EOC patients (Lokshinet al. 2006). Stimulation by CXCL8 of various ovariantumour cell lines leads to their proliferation in vitro(Wang et al. 2005) and is correlated with increasedmetastatic potential in SKOV3 cells (Yin et al. 2011).CXCL8 also has a direct role in angiogenesis, stimulatingcapillary tube formation via CXCR1 and CXCR2 (Li et al.2003). In the context of advanced EOC, development

www.reproduction-online.org

of ascites and possible metastasis is promoted byangiogenesis through the key regulator VEGF (MasoumiMoghaddam et al. 2011). In an early study to determinethe genes expressed in angiogenesis, five differenthuman ovarian carcinomas were implanted into theperitoneal cavities of nude mice (Yoneda et al. 1998).The formation of ascites was associated with theexpression of VEGF and CXCL8, and this was inverselyassociated with survival.

Similar to CXCL5, CXCL1 has chemotactic activity forneutrophils and its N-terminal truncation leads toenhanced chemotaxis in vitro (Wuyts et al. 1999).Over-expression of CXCL1 in vitro also results inincreased cell proliferation (Bolitho et al. 2010).CXCL1 has been shown to promote malignant transfor-mation of epithelial cells through a complex RASpathway-mediated transformation, which inducesnormal stroma to ‘re-program’ and become tumorigenic(Yang et al. 2006). Silencing of CXCL1 using siRNAeliminated RAS-induced transformation (Yang et al.2006). In the same study, CXCL1 expression waselevated in both serum and tumour tissue from ovariancancer patients (Yang et al. 2006). CXCL1 (in com-bination with CCL18) has also been suggested as a serumbiomarker of ovarian cancer (Wang et al. 2011).Multivariate ELISA gave a sensitivity of 92% andspecificity of 97% for detecting ovarian cancers, whichwas significantly higher than values obtained using CA125 alone (Wang et al. 2011).

In normal ovarian function, both CXCL8 and CXCL1are produced by the ovulating follicle to attract CXCR1C

and CXCR2C leukocytes to assist with ovulation(Karstrom-Encrantz et al. 1998). Using mouse modelsof peritoneal ovarian cancer, it has been shown thatmatrix metalloprotease-1 (MMP1) activation of theG protein-coupled receptor, protease-activated

Figure 5 Promotion of an unfavourable outcome inEOC involving CXC chemokines. (1) Tumour-associated macrophages secrete CCL22 to attractTreg cells. This results in suppression of Th17 cells,thereby reducing IL17 and IFN-g and impairingproduction of CXCL9 and -10 (Kryczek et al.2009). CXCL1 and -8 continue to be produced,enhancing angiogenesis (Romagnani et al. 2004);(2) CXCL12 production by tumour cells andenhanced CXCR4 expression by epithelial cancercells facilitate metastasis; (3) the lack of angio-static chemokines and presence of Treg cellsprevent immune cell migration to the immuno-suppressive tumour site (Sato et al. 2005).

Reproduction (2012) 144 303–317

Page 10: The emerging role of CXC chemokines in epithelial ovarian cancer

312 A Rainczuk, J Rao and others

receptor-1 (PAR1), is also an important stimulator ofangiogenesis and metastasis in EOC (Agarwal et al.2008). The paracrine control of chemokine productionby CXCR1 and CXCR2 receptors on ovarian carcinomaendothelial cells is stimulated by the MMP1-PAR1signalling system (Agarwal et al. 2010). After MMP1-PAR1 stimulation, it was shown that ovarian carcinomacells both in vitro and in mice secreted CXCL8 andCXCL1, with the introduction of X1/2pal-i3 pepducincompletely inhibiting angiogenesis (Agarwal et al.2010). Inhibition of this pathway blocks tumour growthand metastasis in breast cancer models (Yang et al.2009); the MMP1-PAR1-CXCR1/2 pathway may there-fore be a new target for preventing EOC-relatedangiogenesis.

Direct involvement of the ELRK chemokine CXCL12in metastatic spread

In early studies to determine the influence of chemokinegradients on the metastatic spread of ovarian tumours, itwas shown that the receptor CXCR4 induced calciumflux and could direct migration of tumour cells (Scottonet al. 2001, 2002). The expression of CXCR4 mRNAwas detected in six ovarian cancer cell lines, ascites (19 of20 samples), and primary ovarian tumours (eight of tensamples). The study showed that the cell lines displayedchemotaxis towards CXCL12, which was also present athigh levels in ovarian cancer ascites fluid from 63 patients(Scotton et al. 2001). However, not all tumour cells in theprimary ovarian tumours expressed CXCR4 mRNA,suggesting that other factors present in the tumourmicroenvironment contributed to receptor expression.After in vitro stimulation of the ovarian cancer cell lineIGROV with CXCL12, cells could invade through Matrigeltowards a CXCL12 gradient (Scotton et al. 2002). This alsopromoted the activation of Akt/protein kinase B, biphasicphosphorylation of p44/42 MAP kinase and induction ofthe cytokine TNF-a (Scotton et al. 2002).

The metastatic spread of EOC tumour cells to theperitoneal mesothelium also involves the CXCL12/CXCR4axis (Kajiyama et al. 2008, Miyanishi et al. 2010).Furthermore, a significant correlation has been shownbetween CXCR4 expression in both primary and meta-static ovarian tumours, as well as lymph node metastases(Jiang et al. 2006). In a mouse model of ovarian cancer,siRNA-mediated silencing of CXCL12 reduced tumourgrowth in vivo, and a CXCR4 antagonist (AMD3100)could increase T-cell-mediated anti-tumour responses(Righi et al. 2011). Human primary ovarian tumour cellsalso express CXCL12 and VEGF following exposure to thehypoxic tumour microenvironment (Kryczek et al. 2005).In this in vivo model, CXCL12 and VEGF promotedangiogenesis and the migration of human vascularendothelial cell expressing CXCR4 (Kryczek et al. 2005).Importantly, CXCL12 and CXCR4 are not detected in

Reproduction (2012) 144 303–317

normal ovarian tissues (Jiang et al. 2006, Kajiyama et al.2008), even in women with a family history of ovariancancer (Scotton et al. 2002).

Recently, sequential genetic changes were found in theTP53 allele as well as the CXCR4 gene locus (chromo-some 2q21.3) of human transformed ovarian surfaceepithelial cell lines (Archibald et al. 2012). These wereinvestigated to determine early events in the develop-ment of serous EOC. Following the screening of 50 serousEOC patient biopsies, the mutated TP53 expression genesignature (with decreased TP53 target gene expression)was associated with activation of epidermal growthfactor receptor pathways, as well as high expression ofCXCR4 genes (with corresponding mRNA and protein).This suggests that these events may occur early in thedevelopment of EOC (Archibald et al. 2012). Amongstother recently discovered genetic changes to potentiallyup-regulate CXCR4 expression in EOC is the abrogationof breast cancer metastasis suppressor 1 (BRMS1; Shenget al. 2012). Suppression of BRMS1 by short-hairpin RNAtransfection of OVCAR3 cell lines significantly enhancedCXCR4 expression at the mRNA and protein levels, withenhanced expression mediated by the NF-kB signallingpathway (Sheng et al. 2012).

Figure 5 offers a simplistic summary of the immuno-suppressive tumour microenvironment in EOC. The lackof angiogenic ELR-chemokines results in uncheckedangiogenesis due to the presence of Treg cells, while CXCchemokines and their receptors, in particular theCXCL12/CXCR4 axis, can promote tumour metastasis.

CXC chemokines as future therapeutic targetsfor EOC

Due to the importance of CXC chemokine signalling,there are active development strategies to exploit thesepathways to reduce the morbidity and mortalityassociated with EOC. One strategy involves the stimu-lation of Th17 cells through the use of vaccines orimmunotherapy to stimulate IFN-g and IL17, resulting inelevated CXCL9 and CXCL10 levels (Munn 2009,Cannon et al. 2011, Goyne et al. 2011). However,animal or clinical trials are yet to be reported. The use ofCXC chemokine inhibitors and antagonists for a varietyof different cancers, including EOC (Zlotnik et al. 2011,Balkwill 2012), is emerging as a promising strategy fornew treatments.

Of the CXC chemokine ligands and receptors involvedin EOC, inhibition of CXCR4 has received a great deal ofattention with several molecules found that are able toantagonise activity in response to CXCL12 (Barbieri et al.2010). In two separate studies involving mouse modelsof EOC, AMD3100 (a clinically approved CXCR4inhibitor) significantly reduced ovarian cancer cellgrowth (Ray et al. 2011, Righi et al. 2011). In mice,administration of AMD3100 significantly reduced

www.reproduction-online.org

Page 11: The emerging role of CXC chemokines in epithelial ovarian cancer

CXC chemokines in epithelial ovarian cancer 313

intraperitoneal dissemination and angiogenesis(measured by vessel density with the tumour), increasedT-cell-mediated anti-tumour immune responses andapoptosis and reduced FoxP3C Treg cell numbers withinthe tumour (Righi et al. 2011). Subsequently, it wasshown that AMD3100 blocked CXCL12/CXCR4 binding,resulting in prolonged survival and reduced tumourgrowth in mice with disseminated ovarian cancer (Rayet al. 2011). The use of this important chemokineinhibitor in human trials is yet to be reported.

Conclusion

A significant body of evidence indicates the involvementof chemokines in EOC and suggests that initiation,progression and metastasis are strongly influenced by theCXC chemokines. In particular, their roles in inflam-mation, angiogenesis and immune cell recruitment haveall been shown to exert profound influence in the tumourmicroenvironment; moreover, there is a complex inter-play of these factors that influences clinical outcomes.Research to exploit these chemokine-signallingpathways, and translate the findings to a clinicalenvironment, remains at a very early stage. Relevantstudies in murine models using the inhibitor AMD3100targeting the CXCR4/CXCL12 axis have been reported,but there are no human trials yet reported. Similarly, theenhancement of CXCL9 and CXCL10 expression throughstimulation of Th17 cells, or CXCL5 inhibitor develop-ment to prevent neutrophil adhesion and potentialmetastasis, is also at an early stage.

The groundwork for understanding the combinedaction and mechanisms of CXC chemokine activity inEOC is underway. Exploitation of CXC chemokine-signalling pathways to reduce EOC morbidity andmortality remains as a future avenue to potentiallyimprove outcomes for patients with this disease.

Declaration of interest

The authors declare that there is no conflict of interest thatcould be perceived as prejudicing the impartiality of theresearch reported.

Funding

This work was supported by the Ovarian Cancer ResearchFoundation and the Victorian Government’s OperationalInfrastructure Support Program.

References

Agarwal A, Covic L, Sevigny LM, Kaneider NC, Lazarides K, Azabdaftari G,Sharifi S & Kuliopulos A 2008 Targeting a metalloprotease-PAR1signaling system with cell-penetrating pepducins inhibits angiogenesis,ascites, and progression of ovarian cancer. Molecular CancerTherapeutics 7 2746–2757. (doi:10.1158/1535-7163.MCT-08-0177)

www.reproduction-online.org

Agarwal A, Tressel SL, Kaimal R, Balla M, Lam FH, Covic L & Kuliopulos A2010 Identification of a metalloprotease-chemokine signaling system inthe ovarian cancer microenvironment: implications for antiangiogenictherapy. Cancer Research 70 5880–5890. (doi:10.1158/0008-5472.CAN-09-4341)

Aitokallio-Tallberg AM, Viinikka LU & Ylikorkala RO 1988 Increasedsynthesis of prostacyclin and thromboxane in human ovarian malig-nancy. Cancer Research 48 2396–2398.

Akishima-Fukasawa Y, Nakanishi Y, Ino Y, Moriya Y, Kanai Y & Hirohashi S2009 Prognostic significance of CXCL12 expression in patients withcolorectal carcinoma. American Journal of Clinical Pathology 132202–210 (quiz 307). (doi:10.1309/AJCPK35VZJEWCUTL)

Archibald KM, Kulbe H, Kwong J, Chakravarty P, Temple J, Chaplin T,Flak MB, McNeish IA, Deen S, Brenton JD et al. 2012 Sequentialgenetic change at the TP53 and chemokine receptor CXCR4 locusduring transformation of human ovarian surface epithelium. Oncogene[in press]. (doi:10.1038/onc.2011.653)

Arnogiannaki N, Grigoriadis C, Zygouris D, Terzakis E, Sebastiadou M &Tserkezoglou A 2011 Proliferative Brenner tumor of the ovary.Clinicopathological study of two cases and review of the literature.European Journal of Gynaecological Oncology 32 576–578.

Auersperg N, Edelson MI, Mok SC, Johnson SW & Hamilton TC 1998 Thebiology of ovarian cancer. Seminars in Oncology 25 281–304.

Balkwill F 2004 Cancer and the chemokine network. Nature Reviews.Cancer 4 540–550. (doi:10.1038/nrc1388)

Balkwill FR 2012 The chemokine system and cancer. Journal of Pathology226 148–157. (doi:10.1002/path.3029)

Barbieri F, Bajetto A & Florio T 2010 Role of chemokine network inthe development and progression of ovarian cancer: a potentialnovel pharmacological target. Journal of Oncology 2010 426956.(doi:10.1155/2010/426956)

Boimel PJ, Smirnova T, Zhou ZN, Wyckoff J, Park HI, Coniglio SJ, Patel P,Qian BZ, Stanley ER, Bresnick AR et al. 2012 Contribution of CXCL12secretion to invasion of breast cancer cells. Breast Cancer Research 14R23. (doi:10.1186/bcr3108)

Bolitho C, Hahn MA, Baxter RC & Marsh DJ 2010 The chemokine CXCL1induces proliferation in epithelial ovarian cancer cells by transactivationof the epidermal growth factor receptor. Endocrine-Related Cancer 17929–940. (doi:10.1677/ERC-10-0107)

Callesen AK, Madsen JS, Iachina M, Vach W, Kruse TA, Jensen ON &Mogensen O 2010 Serum peptide/protein profiling by mass spectrometryprovides diagnostic information independently of CA125 in womenwith an ovarian tumor. Cancer Biomarkers 6 73–82. (doi:10.3233/CBM-2009-0120)

Campanella GS, Colvin RA & Luster AD 2010 CXCL10 can inhibitendothelial cell proliferation independently of CXCR3. PLoS ONE 5e12700. (doi:10.1371/journal.pone.0012700)

Cannon MJ, Goyne H, Stone PJ & Chiriva-Internati M 2011 Dendritic cellvaccination against ovarian cancer – tipping the Treg/TH17 balance totherapeutic advantage? Expert Opinion on Biological Therapy 11441–445. (doi:10.1517/14712598.2011.554812)

CGARN 2011 Integrated genomic analyses of ovarian carcinoma. Nature474 609–615. (doi:10.1038/nature10166)

de Chaisemartin L, Goc J, Damotte D, Validire P, Magdeleinat P, Alifano M,Cremer I, Fridman WH, Sautes-Fridman C & Dieu-Nosjean MC 2011Characterization of chemokines and adhesion molecules associated with Tcell presence in tertiary lymphoid structures in human lung cancer. CancerResearch 71 6391–6399. (doi:10.1158/0008-5472.CAN-11-0952)

Charo IF & Ransohoff RM 2006 The many roles of chemokines andchemokine receptors in inflammation. New England Journal of Medicine354 610–621. (doi:10.1056/NEJMra052723)

Cho KR 2009 Ovarian cancer update: lessons from morphology, molecules,and mice. Archives of Pathology & Laboratory Medicine 133 1775–1781.(doi:10.1043/1543-2165-133.11.1775)

Cho KR & Shih Ie M 2009 Ovarian cancer. Annual Review of Pathology 4287–313. (doi:10.1146/annurev.pathol.4.110807.092246)

Clarke B, Tinker AV, Lee CH, Subramanian S, van de Rijn M, Turbin D,Kalloger S, Han G, Ceballos K, Cadungog MG et al. 2009 IntraepithelialT cells and prognosis in ovarian carcinoma: novel associations withstage, tumor type, and BRCA1 loss. Modern Pathology 22 393–402.(doi:10.1038/modpathol.2008.191)

Reproduction (2012) 144 303–317

Page 12: The emerging role of CXC chemokines in epithelial ovarian cancer

314 A Rainczuk, J Rao and others

Clark-Lewis I, Mattioli I, Gong JH & Loetscher P 2003 Structure–functionrelationship between the human chemokine receptor CXCR3 and itsligands. Journal of Biological Chemistry 278 289–295. (doi:10.1074/jbc.M209470200)

Clendenen TV, Lundin E, Zeleniuch-Jacquotte A, Koenig KL, Berrino F,Lukanova A, Lokshin AE, Idahl A, Ohlson N, Hallmans G et al. 2011Circulating inflammation markers and risk of epithelial ovarian cancer.Cancer Epidemiology, Biomarkers & Prevention 20 799–810.(doi:10.1158/1055-9965.EPI-10-1180)

Coffelt SB & Scandurro AB 2008 Tumors sound the alarmin(s).Cancer Research 68 6482–6485. (doi:10.1158/0008-5472.CAN-08-0044)

Crum CP, Drapkin R, Kindelberger D, Medeiros F, Miron A & Lee Y 2007Lessons from BRCA: the tubal fimbria emerges as an origin for pelvicserous cancer. Clinical Medicine & Research 5 35–44. (doi:10.3121/cmr.2007.702)

Cuenca RE, Azizkhan RG & Haskill S 1992 Characterization of GROa,b and g expression in human colonic tumours: potential significance ofcytokine involvement. Surgical Oncology 1 323–329. (doi:10.1016/0960-7404(92)90094-2)

Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P,Evdemon-Hogan M, Conejo-Garcia JR, Zhang L, Burow M et al. 2004Specific recruitment of regulatory T cells in ovarian carcinoma fostersimmune privilege and predicts reduced survival. Nature Medicine 10942–949. (doi:10.1038/nm1093)

Darash-Yahana M, Gillespie JW, Hewitt SM, Chen YY, Maeda S, Stein I,Singh SP, Bedolla RB, Peled A, Troyer DA et al. 2009 The chemokineCXCL16 and its receptor, CXCR6, as markers and promoters ofinflammation-associated cancers. PLoS ONE 4 e6695. (doi:10.1371/journal.pone.0006695)

De Larco JE, Wuertz BR & Furcht LT 2004 The potential role of neutrophilsin promoting the metastatic phenotype of tumors releasing interleukin-8.Clinical Cancer Research 10 4895–4900. (doi:10.1158/1078-0432.CCR-03-0760)

Demopoulos RI, Aronov R & Mesia A 2001 Clues to the pathogenesis offallopian tube carcinoma: a morphological and immunohistochemicalcase control study. International Journal of Gynecological Pathology 20128–132. (doi:10.1097/00004347-200104000-00003)

Deng L, Chen N, Li Y, Zheng H & Lei Q 2010 CXCR6/CXCL16 functions as aregulator in metastasis and progression of cancer. Biochimica etBiophysica Acta 1806 42–49.

Eck M, Schmausser B, Scheller K, Brandlein S & Muller-Hermelink HK2003 Pleiotropic effects of CXC chemokines in gastric carcinoma:differences in CXCL8 and CXCL1 expression between diffuse andintestinal types of gastric carcinoma. Clinical and ExperimentalImmunology 134 508–515. (doi:10.1111/j.1365-2249.2003.02305.x)

Erreni M, Bianchi P, Laghi L, Mirolo M, Fabbri M, Locati M, Mantovani A &Allavena P 2009 Expression of chemokines and chemokine receptors inhuman colon cancer. Methods in Enzymology 460 105–121.(doi:full_text)

Fader AN, Rasool N, Vaziri SA, Kozuki T, Faber PW, Elson P, Biscotti CV,Michener CM, Rose PG, Rojas-Espaillat L et al. 2010 CCL2 expression inprimary ovarian carcinoma is correlated with chemotherapy responseand survival outcomes. Anticancer Research 30 4791–4798.

Fernandez EJ & Lolis E 2002 Structure, function, and inhibition ofchemokines. Annual Review of Pharmacology and Toxicology 42469–499. (doi:10.1146/annurev.pharmtox.42.091901.115838)

Fleming JS, Beaugie CR, Haviv I, Chenevix-Trench G & Tan OL 2006Incessant ovulation, inflammation and epithelial ovarian carcinogenesis:revisiting old hypotheses. Molecular and Cellular Endocrinology 2474–21. (doi:10.1016/j.mce.2005.09.014)

Frick VO, Rubie C, Wagner M, Graeber S, Grimm H, Kopp B, Rau BM &Schilling MK 2008 Enhanced ENA-78 and IL-8 expression inpatients with malignant pancreatic diseases. Pancreatology 8 488–497.(doi:10.1159/000151776)

Furuya M, Suyama T, Usui H, Kasuya Y, NishiyamaM, Tanaka N, Ishiwata I,Nagai Y, Shozu M & Kimura S 2007 Up-regulation of CXC chemokinesand their receptors: implications for proinflammatory microenviron-ments of ovarian carcinomas and endometriosis. Human Pathology 381676–1687. (doi:10.1016/j.humpath.2007.03.023)

Furuya M, Yoneyama T, Miyagi E, Tanaka R, Nagahama K, Miyagi Y,Nagashima Y, Hirahara F, Inayama Y & Aoki I 2011 Differential

Reproduction (2012) 144 303–317

expression patterns of CXCR3 variants and corresponding CXCchemokines in clear cell ovarian cancers and endometriosis. GynecologicOncology 122 648–655. (doi:10.1016/j.ygyno.2011.05.034)

Gacci M, Serni S, Lapini A, Vittori G, Alessandrini M, Nesi G, Palli D &Carini M 2009 CXCR3-B expression correlates with tumor necrosisextension in renal cell carcinoma. Journal of Urology 181 843–848.(doi:10.1016/j.juro.2008.10.063)

Gelmini S, Mangoni M, Castiglione F, Beltrami C, Pieralli A, Andersson KL,Fambrini M, Taddei GL, Serio M & Orlando C 2009 The CXCR4/CXCL12axis in endometrial cancer. Clinical & Experimental Metastasis 26261–268. (doi:10.1007/s10585-009-9240-4)

Gijsbers K, Gouwy M, Struyf S, Wuyts A, Proost P, Opdenakker G,Penninckx F, Ectors N, Geboes K & Van Damme J 2005 GCP-2/CXCL6synergizes with other endothelial cell-derived chemokines in neutrophilmobilization and is associated with angiogenesis in gastrointestinaltumors. Experimental Cell Research 303 331–342. (doi:10.1016/j.yexcr.2004.09.027)

Gilks CB, Ionescu DN, Kalloger SE, Kobel M, Irving J, Clarke B, Santos J,Le N, Moravan V & Swenerton K 2008 Tumor cell type can bereproducibly diagnosed and is of independent prognostic significance inpatients with maximally debulked ovarian carcinoma. Human Pathology39 1239–1251. (doi:10.1016/j.humpath.2008.01.003)

Giuliani N, Bonomini S, Romagnani P, Lazzaretti M, Morandi F, Colla S,Tagliaferri S, Lasagni L, Annunziato F, Crugnola M et al. 2006 CXCR3and its binding chemokines in myeloma cells: expression of isoforms andpotential relationships with myeloma cell proliferation and survival.Haematologica 91 1489–1497.

Gottlieb AB, Luster AD, Posnett DN & Carter DM 1988 Detection of agamma interferon-induced protein IP-10 in psoriatic plaques. Journal ofExperimental Medicine 168 941–948. (doi:10.1084/jem.168.3.941)

Goyne H, Stone PJ & Cannon MJ 2011 Combinatorial strategies foralleviation of tumor-associated immune suppression and therapeuticvaccination against ovarian cancer. Immunotherapy 3 805–807. (doi:10.2217/imt.11.71)

Green A, Purdie D, Bain C, Siskind V, Russell P, Quinn M & Ward B 1997Tubal sterilisation, hysterectomy and decreased risk of ovarian cancer.Survey of Women’s Health Study Group. International Journal of Cancer71 948–951. (doi:10.1002/(SICI)1097-0215(19970611)71:6!948::AID-IJC6O3.0.CO;2-Y)

Griffioen AW & Molema G 2000 Angiogenesis: potentials for pharmaco-logic intervention in the treatment of cancer, cardiovascular diseases,and chronic inflammation. Pharmacological Reviews 52 237–268.

Groom JR & Luster AD 2011 CXCR3 in T cell function. Experimental CellResearch 317 620–631. (doi:10.1016/j.yexcr.2010.12.017)

Gutwein P, Schramme A, Sinke N, Abdel-BakkyMS, Voss B, Obermuller N,Doberstein K, Koziolek M, Fritzsche F, Johannsen M et al. 2009Tumoural CXCL16 expression is a novel prognostic marker of longersurvival times in renal cell cancer patients. European Journal of Cancer45 478–489. (doi:10.1016/j.ejca.2008.10.023)

Ha HK, Lee W, Park HJ, Lee SD, Lee JZ & Chung MK 2011 Clinicalsignificance of CXCL16/CXCR6 expression in patients with prostatecancer. Molecular Medicine Reports 4 419–424. (doi:10.3892/mmr.2011.446)

Hassan S, Ferrario C, Saragovi U, Quenneville L, Gaboury L, Baccarelli A,Salvucci O & Basik M 2009 The influence of tumor–host interactions inthe stromal cell-derived factor-1/CXCR4 ligand/receptor axis in deter-mining metastatic risk in breast cancer. American Journal of Pathology175 66–73. (doi:10.2353/ajpath.2009.080948)

Hojo S, Koizumi K, Tsuneyama K, Arita Y, Cui Z, Shinohara K, Minami T,Hashimoto I, Nakayama T, Sakurai H et al. 2007 High-level expressionof chemokine CXCL16 by tumor cells correlates with a good prognosisand increased tumor-infiltrating lymphocytes in colorectal cancer.Cancer Research 67 4725–4731. (doi:10.1158/0008-5472.CAN-06-3424)

van der Horst PH, Wang Y, Vandenput I, Kuhne LC, Ewing PC, vanIjcken WF, van der Zee M, Amant F, Burger CW & Blok LJ 2012Progesterone inhibits epithelial-to-mesenchymal transition in endometrialcancer. PLoS ONE 7 e30840. (doi:10.1371/journal.pone.0030840)

Hwang WT, Adams SF, Tahirovic E, Hagemann IS & Coukos G 2012Prognostic significance of tumor-infiltrating T cells in ovarian cancer: ameta-analysis. Gynecologic Oncology 124 192–198. (doi:10.1016/j.ygyno.2011.09.039)

www.reproduction-online.org

Page 13: The emerging role of CXC chemokines in epithelial ovarian cancer

CXC chemokines in epithelial ovarian cancer 315

Jiang YP, Wu XH, Shi B, Wu WX & Yin GR 2006 Expression of chemokineCXCL12 and its receptor CXCR4 in human epithelial ovarian cancer: anindependent prognostic factor for tumor progression. GynecologicOncology 103 226–233. (doi:10.1016/j.ygyno.2006.02.036)

Kajiyama H, Shibata K, Terauchi M, Ino K, Nawa A & Kikkawa F 2008Involvement of SDF-1a/CXCR4 axis in the enhanced peritonealmetastasis of epithelial ovarian carcinoma. International Journal ofCancer 122 91–99. (doi:10.1002/ijc.23083)

Kaku T, Ogawa S, Kawano Y, Ohishi Y, Kobayashi H, Hirakawa T &Nakano H 2003 Histological classification of ovarian cancer. MedicalElectron Microscopy 36 9–17. (doi:10.1007/s007950300002)

Karstrom-Encrantz L, Runesson E, Bostrom EK & Brannstrom M 1998Selective presence of the chemokine growth-regulated oncogene a

(GROa) in the human follicle and secretion from cultured granulosa-lutein cells at ovulation. Molecular Human Reproduction 4 1077–1083.(doi:10.1093/molehr/4.11.1077)

Keeley EC, Mehrad B & Strieter RM 2008 Chemokines as mediators ofneovascularization. Arteriosclerosis, Thrombosis, and Vascular Biology28 1928–1936. (doi:10.1161/ATVBAHA.108.162925)

Kelley TW & Parker CJ 2010 CD4 (C)CD25 (C)Foxp3 (C) regulatory Tcells and hematologic malignancies. Frontiers in Bioscience 2 980–992.(doi:10.2741/S114)

Kiefer F & Siekmann AF 2011 The role of chemokines and their receptors inangiogenesis. Cellular and Molecular Life Sciences 68 2811–2830.(doi:10.1007/s00018-011-0677-7)

Kim J, Coffey DM, Creighton CJ, Yu Z, Hawkins SM & Matzuk MM 2012High-grade serous ovarian cancer arises from fallopian tube in a mousemodel. PNAS 109 3921–3926. (doi:10.1073/pnas.1117135109)

Klink M, Jastrzembska K, Nowak M, Bednarska K, Szpakowski M, Szyllo K& Sulowska Z 2008 Ovarian cancer cells modulate human bloodneutrophils response to activation in vitro. Scandinavian Journal ofImmunology 68 328–336. (doi:10.1111/j.1365-3083.2008.02139.x)

Kobel M, Kalloger SE, Boyd N, McKinney S, Mehl E, Palmer C, Leung S,Bowen NJ, Ionescu DN, Rajput A et al. 2008 Ovarian carcinomasubtypes are different diseases: implications for biomarker studies. PLoSMedicine 5 e232. (doi:10.1371/journal.pmed.0050232)

Kryczek I, Lange A, Mottram P, Alvarez X, Cheng P, Hogan M, Moons L,Wei S, Zou L, Machelon V et al. 2005 CXCL12 and vascular endothelialgrowth factor synergistically induce neoangiogenesis in human ovariancancers. Cancer Research 65 465–472.

Kryczek I, Banerjee M, Cheng P, Vatan L, Szeliga W, Wei S, Huang E,Finlayson E, Simeone D, Welling TH et al. 2009 Phenotype, distribution,generation, and functional and clinical relevance of Th17 cells in thehuman tumor environments. Blood 114 1141–1149. (doi:10.1182/blood-2009-03-208249)

Kulbe H, Chakravarty P, Leinster DA, Charles KA, Kwong J, Thompson RG,Coward JI, Schioppa T, Robinson SC, Gallagher WM et al. 2012 Adynamic inflammatory cytokine network in the human ovarian cancermicroenvironment. Cancer Research 72 66–75. (doi:10.1158/0008-5472.CAN-11-2178)

Lasagni L, Francalanci M, Annunziato F, Lazzeri E, Giannini S, Cosmi L,Sagrinati C, Mazzinghi B, Orlando C, Maggi E et al. 2003 Analternatively spliced variant of CXCR3 mediates the inhibition ofendothelial cell growth induced by IP-10, Mig, and I-TAC, and acts asfunctional receptor for platelet factor 4. Journal of ExperimentalMedicine 197 1537–1549. (doi:10.1084/jem.20021897)

Li A, Dubey S, Varney ML, Dave BJ & Singh RK 2003 IL-8 directly enhancedendothelial cell survival, proliferation, and matrix metalloproteinasesproduction and regulated angiogenesis. Journal of Immunology 1703369–3376.

Li A, King J, Moro A, Sugi MD, Dawson DW, Kaplan J, Li G, Lu X,Strieter RM, Burdick M et al. 2011 Overexpression of CXCL5 isassociated with poor survival in patients with pancreatic cancer.American Journal of Pathology 178 1340–1349. (doi:10.1016/j.ajpath.2010.11.058)

Lokshin AE, Winans M, Landsittel D, Marrangoni AM, Velikokhatnaya L,Modugno F, Nolen BM & Gorelik E 2006 Circulating IL-8 and anti-IL-8autoantibody in patients with ovarian cancer. Gynecologic Oncology102 244–251. (doi:10.1016/j.ygyno.2005.12.011)

Ludwig A, Schulte A, Schnack C, Hundhausen C, Reiss K, Brodway N,Held-Feindt J & Mentlein R 2005 Enhanced expression and shedding of

www.reproduction-online.org

the transmembrane chemokine CXCL16 by reactive astrocytes andglioma cells. Journal of Neurochemistry 93 1293–1303. (doi:10.1111/j.1471-4159.2005.03123.x)

Maccio A & Madeddu C 2012 Inflammation and ovarian cancer. Cytokine58 133–147. (doi:10.1016/j.cyto.2012.01.015)

Machelon V, Gaudin F, Camilleri-Broet S, Nasreddine S, Bouchet-Delbos L,Pujade-Lauraine E, Alexandre J, Gladieff L, Arenzana-Seisdedos F,Emilie D et al. 2011 CXCL12 expression by healthy and malignantovarian epithelial cells. BMC Cancer 11 97. (doi:10.1186/1471-2407-11-97)

Mantovani A, Savino B, Locati M, Zammataro L, Allavena P & Bonecchi R2010 The chemokine system in cancer biology and therapy. Cytokine &Growth Factor Reviews 21 27–39. (doi:10.1016/j.cytogfr.2009.11.007)

Masoumi Moghaddam S, Amini A, Morris DL & Pourgholami MH 2012Significance of vascular endothelial growth factor in growth andperitoneal dissemination of ovarian cancer. Cancer and MetastasisReviews 31 143–162. (doi:10.1007/s10555-011-9337-5)

Matsubara J, Honda K, Ono M, Tanaka Y, Kobayashi M, Jung G,Yanagisawa K, Sakuma T, Nakamori S, Sata N et al. 2011 Reducedplasma level of CXC chemokine ligand 7 in patients with pancreaticcancer. Cancer Epidemiology, Biomarkers & Prevention 20 160–171.(doi:10.1158/1055-9965.EPI-10-0397)

Medeiros F, Muto MG, Lee Y, Elvin JA, Callahan MJ, Feltmate C, Garber JE,Cramer DW & Crum CP 2006 The tubal fimbria is a preferred site forearly adenocarcinoma in women with familial ovarian cancer syndrome.American Journal of Surgical Pathology 30 230–236. (doi:10.1097/01.pas.0000180854.28831.77)

Merritt MA & Cramer DW 2011 Molecular pathogenesis of endometrialand ovarian cancer. Cancer Biomarkers 9 287–305. (doi:10.3233/CBM-2011-0167)

Milner BJ, Allan LA, Eccles DM, Kitchener HC, Leonard RC, Kelly KF,Parkin DE & Haites NE 1993 p53 mutation is a common genetic event inovarian carcinoma. Cancer Research 53 2128–2132.

Miyanishi N, Suzuki Y, Simizu S, Kuwabara Y, Banno K & Umezawa K 2010Involvement of autocrine CXCL12/CXCR4 system in the regulationof ovarian carcinoma cell invasion. Biochemical and BiophysicalResearch Communications 403 154–159. (doi:10.1016/j.bbrc.2010.11.007)

Moore SW & Enterline HT 1975 Significance of proliferative epitheliallesions of the uterine tube. Obstetrics and Gynecology 45 385–390.

Mortier A, Loos T, Gouwy M, Ronsse I, Van Damme J & Proost P 2010Posttranslational modification of the NH2-terminal region of CXCL5 byproteases or peptidylarginine deiminases (PAD) differently affects itsbiological activity. Journal of Biological Chemistry 285 29750–29759.(doi:10.1074/jbc.M110.119388)

Mortier A, Gouwy M, Van Damme J & Proost P 2011 Effect ofposttranslational processing on the in vitro and in vivo activity ofchemokines. Experimental Cell Research 317 642–654. (doi:10.1016/j.yexcr.2010.11.016)

Mukaida N & Baba T 2012 Chemokines in tumor development andprogression. Experimental Cell Research 318 95–102. (doi:10.1016/j.yexcr.2011.10.012)

Munn DH 2009 Th17 cells in ovarian cancer. Blood 114 1134–1135.(doi:10.1182/blood-2009-06-224246)

MurdochWJ & McDonnel AC 2002 Roles of the ovarian surface epitheliumin ovulation and carcinogenesis. Reproduction 123 743–750. (doi:10.1530/rep.0.1230743)

Murdoch WJ, Murphy CJ, Van Kirk EA & Shen Y 2010 Mechanisms andpathobiology of ovulation. Society of Reproduction and Fertility.Supplement 67 189–201.

Musha H, Ohtani H, Mizoi T, Kinouchi M, Nakayama T, Shiiba K,Miyagawa K, Nagura H, Yoshie O & Sasaki I 2005 Selective infiltration ofCCR5(C)CXCR3(C) T lymphocytes in human colorectal carcinoma.International Journal of Cancer 116 949–956. (doi:10.1002/ijc.21135)

Naldini A, Morena E, Belotti D, Carraro F, Allavena P & Giavazzi R 2011Identification of thrombin-like activity in ovarian cancer associatedascites and modulation of multiple cytokine networks. Thrombosis andHaemostasis 106 705–711. (doi:10.1160/TH11-05-0311)

Ness RB & Cottreau C 1999 Possible role of ovarian epithelial inflammationin ovarian cancer. Journal of the National Cancer Institute 911459–1467. (doi:10.1093/jnci/91.17.1459)

Reproduction (2012) 144 303–317

Page 14: The emerging role of CXC chemokines in epithelial ovarian cancer

316 A Rainczuk, J Rao and others

Ness RB & Modugno F 2006 Endometriosis as a model for inflammation–hormone interactions in ovarian and breast cancers. European Journal ofCancer 42 691–703. (doi:10.1016/j.ejca.2006.01.009)

Ness RB, Grisso JA, Cottreau C, Klapper J, Vergona R, Wheeler JE,Morgan M & Schlesselman JJ 2000 Factors related to inflammation of theovarian epithelium and risk of ovarian cancer. Epidemiology 11111–117. (doi:10.1097/00001648-200003000-00006)

Ohtani H, Jin Z, Takegawa S, Nakayama T & Yoshie O 2009Abundant expression of CXCL9 (MIG) by stromal cells that includedendritic cells and accumulation of CXCR3CT cells in lymphocyte-richgastric carcinoma. Journal of Pathology 217 21–31. (doi:10.1002/path.2448)

Ray P, Lewin SA, Mihalko LA, Schmidt BT, Luker KE & Luker GD 2011Noninvasive imaging reveals inhibition of ovarian cancer by targetingCXCL12–CXCR4. Neoplasia 13 1152–1161.

Righi E, Kashiwagi S, Yuan J, Santosuosso M, Leblanc P, Ingraham R,Forbes B, Edelblute B, Collette B, Xing D et al. 2011 CXCL12/CXCR4blockade induces multimodal antitumor effects that prolong survival inan immunocompetent mouse model of ovarian cancer. Cancer Research71 5522–5534. (doi:10.1158/0008-5472.CAN-10-3143)

Roett MA & Evans P 2009 Ovarian cancer: an overview. American FamilyPhysician/GP 80 609–616.

Romagnani P, Annunziato F, Lasagni L, Lazzeri E, Beltrame C,Francalanci M, Uguccioni M, Galli G, Cosmi L, Maurenzig L et al.2001 Cell cycle-dependent expression of CXC chemokine receptor 3 byendothelial cells mediates angiostatic activity. Journal of ClinicalInvestigation 107 53–63. (doi:10.1172/JCI9775)

Romagnani P, Lasagni L, Annunziato F, Serio M & Romagnani S 2004CXC chemokines: the regulatory link between inflammation andangiogenesis. Trends in Immunology 25 201–209. (doi:10.1016/j.it.2004.02.006)

Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, Jungbluth AA,Frosina D, Gnjatic S, Ambrosone C et al. 2005 Intraepithelial CD8C

tumor-infiltrating lymphocytes and a high CD8C/regulatory T cell ratioare associated with favorable prognosis in ovarian cancer. PNAS 10218538–18543. (doi:10.1073/pnas.0509182102)

Schutyser E, Struyf S, Proost P, Opdenakker G, Laureys G, Verhasselt B,Peperstraete L, Van de Putte I, Saccani A, Allavena P et al. 2002Identification of biologically active chemokine isoforms from ascitic fluidand elevated levels of CCL18/pulmonary and activation-regulatedchemokine in ovarian carcinoma. Journal of Biological Chemistry 27724584–24593. (doi:10.1074/jbc.M112275200)

Scotton CJ, Wilson JL, Milliken D, Stamp G & Balkwill FR 2001 Epithelialcancer cell migration: a role for chemokine receptors? Cancer Research61 4961–4965.

Scotton CJ, Wilson JL, Scott K, Stamp G,Wilbanks GD, Fricker S, Bridger G& Balkwill FR 2002 Multiple actions of the chemokine CXCL12 onepithelial tumor cells in human ovarian cancer. Cancer Research 625930–5938.

Seidman JD, Sherman ME, Bell KA, Katabuchi H, O’Leary TJ & Kurman RJ2002 Salpingitis, salpingoliths, and serous tumors of the ovaries: is therea connection? International Journal of Gynecological Pathology 21101–107. (doi:10.1097/00004347-200204000-00001)

Sheng XJ, Zhou YQ, Song QY, Zhou DM & Liu QC 2012 Loss of breastcancer metastasis suppressor 1 promotes ovarian cancer cell metastasisby increasing chemokine receptor 4 expression. Oncology Reports 271011–1018. (doi:10.3892/or.2011.1596)

Smirnova T, Zhou ZN, Flinn RJ, Wyckoff J, Boimel PJ, Pozzuto M,Coniglio SJ, Backer JM, Bresnick AR, Condeelis JS et al. 2012Phosphoinositide 3-kinase signaling is critical for ErbB3-drivenbreast cancer cell motility and metastasis. Oncogene 31 706–715.(doi:10.1038/onc.2011.275)

Son DS, Parl AK, Rice VM & Khabele D 2007 Keratinocyte chemoattractant(KC)/human growth-regulated oncogene (GRO) chemokines and pro-inflammatory chemokine networks in mouse and human ovarianepithelial cancer cells. Cancer Biology and Therapy 6 1302–1312.(doi:10.4161/cbt.6.8.4506)

Spinetti G, Camarda G, Bernardini G, Romano Di Peppe S, Capogrossi MC& Napolitano M 2001 The chemokine CXCL13 (BCA-1) inhibits FGF-2effects on endothelial cells. Biochemical and Biophysical ResearchCommunications 289 19–24. (doi:10.1006/bbrc.2001.5924)

Reproduction (2012) 144 303–317

Strandell A, Thorburn J & Wallin A 2004 The presence of cytokinesand growth factors in hydrosalpingeal fluid. Journal of AssistedReproduction and Genetics 21 241–247. (doi:10.1023/B:JARG.0000042009.93520.15)

Strieter RM, Belperio JA, Phillips RJ & Keane MP 2004 CXC chemokines inangiogenesis of cancer. Seminars in Cancer Biology 14 195–200.(doi:10.1016/j.semcancer.2003.10.006)

Strieter RM, Burdick MD, Mestas J, Gomperts B, Keane MP & Belperio JA2006 Cancer CXC chemokine networks and tumour angiogenesis.European Journal of Cancer42768–778. (doi:10.1016/j.ejca.2006.01.006)

Struyf S, Burdick MD, Peeters E, Van den Broeck K, Dillen C, Proost P, VanDamme J & Strieter RM 2007 Platelet factor-4 variant chemokineCXCL4L1 inhibits melanoma and lung carcinoma growth and metastasisby preventing angiogenesis. Cancer Research 67 5940–5948.(doi:10.1158/0008-5472.CAN-06-4682)

Subbaramaiah K, Telang N, Bansal MB,Weksler BB & Dannenberg AJ 1997Cyclooxygenase-2 gene expression is upregulated in transformedmammary epithelial cells. Annals of the New York Academy of Sciences833 179–185. (doi:10.1111/j.1749-6632.1997.tb48606.x)

Tomsova M, Melichar B, Sedlakova I & Steiner I 2008 Prognostic significanceof CD3C tumor-infiltrating lymphocytes in ovarian carcinoma. GynecologicOncology 108 415–420. (doi:10.1016/j.ygyno.2007.10.016)

Vang R, Shih Ie M & Kurman RJ 2009 Ovarian low-grade and high-gradeserous carcinoma: pathogenesis, clinicopathologic and molecularbiologic features, and diagnostic problems. Advances in AnatomicPathology 16 267–282. (doi:10.1097/PAP.0b013e3181b4fffa)

Wang Y, Yang J, Gao Y, Du Y, Bao L, Niu W & Yao Z 2005 Regulatory effectof e2, IL-6 and IL-8 on the growth of epithelial ovarian cancer cells.Cellular & Molecular Immunology 2 365–372.

Wang Q, Li D, Zhang W, Tang B, Li QQ & Li L 2011 Evaluation ofproteomics-identified CCL18 and CXCL1 as circulating tumor markersfor differential diagnosis between ovarian carcinomas and benignpelvic masses. International Journal of Biological Markers 26 262–273.(doi:10.5301/JBM.2011.8616)

Whitworth JM & Alvarez RD 2011 Evaluating the role of IL-12 basedtherapies in ovarian cancer: a review of the literature. Expert Opinion onBiological Therapy 11 751–762. (doi:10.1517/14712598.2011.566854)

Wilke CM, Bishop K, Fox D & Zou W 2011 Deciphering the role ofTh17 cells in human disease. Trends in Immunology 32 603–611.(doi:10.1016/j.it.2011.08.003)

Wislez M, Philippe C, Antoine M, Rabbe N, Moreau J, Bellocq A,Mayaud C, Milleron B, Soler P & Cadranel J 2004 Upregulation ofbronchioloalveolar carcinoma-derived C-X-C chemokines by tumorinfiltrating inflammatory cells. Inflammation Research 53 4–12.(doi:10.1007/s00011-003-1215-3)

Wong KH, Negishi H & Adashi EY 2002 Expression, hormonal regulation,and cyclic variation of chemokines in the rat ovary: key determinants ofthe intraovarian residence of representatives of the white blood cellseries. Endocrinology 143 784–791. (doi:10.1210/en.143.3.784)

Wu QD, Wang JH, Condron C, Bouchier-Hayes D & Redmond HP 2001Human neutrophils facilitate tumor cell transendothelial migration.American Journal of Physiology. Cell Physiology 280 C814–C822.

Wuyts A, Govaerts C, Struyf S, Lenaerts JP, Put W, Conings R, Proost P &Van Damme J 1999 Isolation of the CXC chemokines ENA-78, GROaand GRO gamma from tumor cells and leukocytes reveals NH2-terminalheterogeneity. Functional comparison of different natural isoforms.European Journal of Biochemistry 260 421–429. (doi:10.1046/j.1432-1327.1999.00166.x)

Yang G, Rosen DG, Zhang Z, Bast RC Jr, Mills GB, Colacino JA,Mercado-Uribe I & Liu J 2006 The chemokine growth-regulatedoncogene 1 (Gro-1) links RAS signaling to the senescence of stromalfibroblasts and ovarian tumorigenesis. PNAS 103 16472–16477.(doi:10.1073/pnas.0605752103)

Yang E, Boire A, Agarwal A, Nguyen N, O’Callaghan K, Tu P, Kuliopulos A &Covic L 2009 Blockade of PAR1 signaling with cell-penetratingpepducins inhibits Akt survival pathways in breast cancer cells andsuppresses tumor survival and metastasis. Cancer Research 696223–6231. (doi:10.1158/0008-5472.CAN-09-0187)

Yigit R, Massuger LF, Zusterzeel PL, Pots J, Figdor CG & Torensma R 2011Cytokine profiles in cyst fluids from ovarian tumors reflect immuno-suppressive state of the tumor. International Journal of GynecologicalCancer 21 1241–1247. (doi:10.1097/IGC.0b013e3182289ab1)

www.reproduction-online.org

Page 15: The emerging role of CXC chemokines in epithelial ovarian cancer

CXC chemokines in epithelial ovarian cancer 317

Yin J, Yu C, Yang Z, He JL, Chen WJ, Liu HZ, Li WM, Liu HT & Wang YX2011 Tetramethylpyrazine inhibits migration of SKOV3 human ovariancarcinoma cells and decreases the expression of interleukin-8 via theERK1/2, p38 and AP-1 signaling pathways. Oncology Reports 26671–679. (doi:10.3892/or.2011.1334)

Yoneda J, Kuniyasu H, Crispens MA, Price JE, Bucana CD & Fidler IJ 1998Expression of angiogenesis-related genes and progression of humanovarian carcinomas in nude mice. Journal of the National CancerInstitute 90 447–454. (doi:10.1093/jnci/90.6.447)

Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M,Regnani G, Makrigiannakis A, Gray H, Schlienger K, Liebman MN et al.2003 Intratumoral T cells, recurrence, and survival in epithelial ovariancancer. New England Journal of Medicine 348 203–213. (doi:10.1056/NEJMoa020177)

www.reproduction-online.org

Zhou C, Borillo J, Wu J, Torres L & Lou YH 2004 Ovarian expression ofchemokines and their receptors. Journal of Reproductive Immunology 631–9. (doi:10.1016/j.jri.2004.03.002)

Zlotnik A, Burkhardt AM & Homey B 2011 Homeostatic chemokinereceptors and organ-specific metastasis. Nature Reviews. Immunology11 597–606. (doi:10.1038/nri3049)

Zou W & Restifo NP 2010 T(H)17 cells in tumour immunity and immuno-therapy. Nature Reviews. Immunology 10 248–256. (doi:10.1038/nri2742)

Received 24 April 2012

First decision 19 June 2012

Accepted 5 July 2012

Reproduction (2012) 144 303–317