11
VO L UM E 26 NUM B ER 6 F EB RUA RY 20 2 008 JOURNAL OF CLINICAL ONCOLOGY BIOLOGY OF NEOPLASIA Early Events in the Pathogenesis of Epithelial Ovarian Cancer Charles N. Landen Jr, Michael J. Birrer, and Anil K. Sood F ro m t h e De par t me nt of G yneco log ic O nco log y and t h e De par t me nt of Can cer Bio log y, U nive r sit y of T exas M .D . An de r son Can cer Cen t er , Hou s- t on , TX ; an d t he Cent e r f or Cance r Re sear ch, Nat i onal Cancer I nst i t ut e, Be t hesd a, M D. ABSTRACT Ovarian carcinogenesis, as in most cancers, involves multiple genetic alterations. A great deal has been learned about proteins and pathways important in the early stages of malignant transformation and metastasis, as derived from studies of individual tumors, microarray data, animal models, and inherited disorders that confer susceptibility. However, a full understand- ing of the earliest recognizable events in epithelial ovarian carcinogenesis is limited by the lack of a well-defined premalignant state common to all ovarian subtypes and by the paucity of data from early-stage cancers. Evidence suggests that ovarian cancers can progress both through a stepwise mutation process (low-grade pathway) and through greater genetic instability that leads to rapid metastasis without an identifiable precursor lesion (high-grade pathway). In this review, we discuss many of the genetic and molecular disorders in each key process that is altered in cancer cells, and we present a model of ovarian pathogenesis that incorporates the role of tumor cell mutations and factors in the host microenvironment important to tumor initiation and progression. S ub mit t e d Janu ar y 8, 200 7; accept e d M ay 25, 2 007 ; p ubl ishe d on lin e ahe ad o f p r int at w w w . jco .o r g on Januar y 1 4, 2 008 . S up por t e d in par t by t h e Rep r odu ct ive S cien t ist De velo pm ent P r og r am t hr o ug h NI H G ran t No. 5 K1 2HD 008 49 an d t he Ovar i an Cance r Resear ch Fun d ( C. N. L. ); Gr an t s No. CA 11 079 301 and CA 1 092 980 1 f r om t he N at ion al I nst i- t ut e s of He alt h ( A. K. S . ); Gr an t N o. P5 0 CA0 836 39 f r o m t he M. D. And er son Can cer Cen t er O var ian Can cer S peci al- iz ed Pr o gr am o f Rese ar ch E xcell ence ; a P r og ram Pr o ject D evel op men t Gr ant f r om t he O var ian Cance r R esear ch F und In c; t h e M ar cus Fo und at ion ( A. K. S .) ; and t h e I nt r amu r al Rese ar ch P r og ram of t he Nat io nal I nst i t ut es o f He al t h, Na t io na l C a nc er Ins t it u te (M .J . B. ) . J Clin Oncol 26:995-1005. © 2008 by American Society of Clinical Oncology INTRODUCTION ETIOLOGY OF SPORADIC EOC Ovarian cancer is the fifth leading cause of can- cer deaths among women, and it is the most common cause among gynecologic malignan- cies.1 The poor ratio of survival to incidence in epithelial ovarian cancer (EOC) results from the high percentage of cases diagnosed at an advanced stage. Despite advances in surgery and chemotherapy, survival of patients with EOC stands at just 45% at 5 years.1 Although the age of biologic therapies holds the potential of improved responses in advanced and recur- rent EOC, a greater impact could be made by recognition of high-risk patients and by offer- ing risk-reducing surgery, a strategy that has demonstrated effectiveness in patients with ge- netic predispositions.2 However, there is signif- icant heterogeneity within the EOC group. For example, histologically defined subtypes such as serous, endometrioid, mucinous, and low- and high-grade malignancies all have variable clinical manifestations and underlying molecu- lar signatures.3 Substantial advances have been made in understanding the genetic alterations and biologic processes in ovarian cancer; how- ever, the etiology remains poorly understood. In this article, we will focus on the current understanding of the early events in EOC. The ovary is surrounded by a single-cell layer of peritoneal mesothelium, which is derived from the coelomic layer during development and which has the potential to undergo metaplastic transformation to a more differentiated state.4 Unlike most malig- nancies, as this epithelium transforms into a malig- nant phenotype, it becomes more differentiated, and it can differentiate toward many of the different cell types found in the mu¨llerian tract, including those in the fallopian tube, uterus, cervix, and ovarian stroma.5 It is widely thought that most ovarian cancers develop from the surface epithe- lium or postovulatory inclusion cysts that were subjected to prolonged exposure to hormones or other chemokines.4 Au t hor s’ di sclosu r es of p ot en t ial co n- ict s of in t er est an d aut h or co nt r ib u- t io ns ar e f o un d at t he end of t hi s ar t icl e. Co r re spo ndi ng au t hor : Ani l K . S oo d, M D, Pr o f essor , De par t me nt s of G yne- co log ic O ncol ogy an d Cance r Biol ogy, Un ive rsi t y of Te xas M. D. And er so n Can cer Cen t er , 115 5 He r man P r essl er , Un it 13 62 , H oust o n, TX 770 30; e- mai l: aso od@ md ande r son. or g . © 20 08 b y Amer i can S ocie t y of Cli nical O nco log y 0 732 -1 83X / 08/ 26 06- 99 5/ $20 .0 0 Primary peritoneal and fallopian tube carcino- DO I : 1 0. 12 00/ JCO .2 006 .0 7. 997 0 mas have similar clinical, molecular, and genetic profiles to ovarian cancers, though some small dif- ferences in frequency of specific protein expression have been described.6 -11 Primary peritoneal carci- nomas may,in fact, have a multifocal and polyclonal origin.12 Therefore, although these entities are often lumped together with ovarian cancer, there may be some significant, but currently poorly defined, dif- ferences. In fact, recent pathologic examination of consecutive cases of ovarian, primary peritoneal, and fallopian tube cancers suggests that a greater 995 Information downloaded from jco.ascopubs.org and provided by UNIVERSITEITSBIBLIOTHEEK on March 25, 2010 from 131.174.244.21. Copyright © 2008 by the American Society of Clinical Oncology. All rights reserved.

Early Events in the Pathogenesis of Epithelial Ovarian Cancer-printed - Copy

  • Upload
    doddydr

  • View
    214

  • Download
    0

Embed Size (px)

DESCRIPTION

epitelial ovarium

Citation preview

Page 1: Early Events in the Pathogenesis of Epithelial Ovarian Cancer-printed - Copy

VO L UM E 26 NUM B ER 6 F EB RUA RY 20 2 008

JOURNAL OF CLINICAL ONCOLOGY BIOLOGY OF NEOPLASIA

Early Events in the Pathogenesis of EpithelialOvarian CancerCharles N. Landen Jr, Michael J. Birrer, and Anil K. Sood

F ro m t h e De par t me nt of G yneco log ic

O nco log y and t h e De par t me nt of

Can cer Bio log y, U nive r sit y of T exas

M .D . An de r son Can cer Cen t er , Hou s-

t on , TX ; an d t he Cent e r f or Cance r

Re sear ch, Nat i onal Cancer I nst i t ut e,

Be t hesd a, M D.

ABSTRACT

Ovarian carcinogenesis, as in most cancers, involves multiple genetic alterations. A great dealhas been learned about proteins and pathways important in the early stages of malignanttransformation and metastasis, as derived from studies of individual tumors, microarray data,animal models, and inherited disorders that confer susceptibility. However, a full understand-ing of the earliest recognizable events in epithelial ovarian carcinogenesis is limited by the lackof a well-defined premalignant state common to all ovarian subtypes and by the paucity of datafrom early-stage cancers. Evidence suggests that ovarian cancers can progress both througha stepwise mutation process (low-grade pathway) and through greater genetic instability thatleads to rapid metastasis without an identifiable precursor lesion (high-grade pathway). In thisreview, we discuss many of the genetic and molecular disorders in each key process that isaltered in cancer cells, and we present a model of ovarian pathogenesis that incorporates therole of tumor cell mutations and factors in the host microenvironment important to tumorinitiation and progression.

S ub mit t e d Janu ar y 8, 200 7; accept e d

M ay 25, 2 007 ; p ubl ishe d on lin e ahe ad

o f p r int at w w w . jco .o r g on Januar y 1 4,

2 008 .S up por t e d in par t by t h e Rep r odu ct ive

S cien t ist De velo pm ent P r og r am

t hr o ug h NI H G ran t No. 5 K1 2HD 008 49

an d t he Ovar i an Cance r Resear ch Fun d

( C. N. L. ); Gr an t s No. CA 11 079 301 and

CA 1 092 980 1 f r om t he N at ion al I nst i-

t ut e s of He alt h ( A. K. S . ); Gr an t N o. P5 0

CA0 836 39 f r o m t he M. D. And er son

Can cer Cen t er O var ian Can cer S peci al-

iz ed Pr o gr am o f Rese ar ch E xcell ence ; a

P r og ram Pr o ject D evel op men t Gr ant

f r om t he O var ian Cance r R esear ch

F und In c; t h e M ar cus Fo und at ion

( A. K. S .) ; and t h e I nt r amu r al Rese ar ch

P r og ram of t he Nat io nal I nst i t ut es o f

He al t h, Na t io na l C a nc er Ins t it u te (M .J . B. ) .

J Clin Oncol 26:995-1005. © 2008 by American Society of Clinical Oncology

INTRODUCTION ETIOLOGY OF SPORADIC EOCOvarian cancer is the fifth leading cause of can-

cer deaths among women, and it is the mostcommon cause among gynecologic malignan-cies.1 The poor ratio of survival to incidence inepithelial ovarian cancer (EOC) results fromthe high percentage of cases diagnosed at anadvanced stage. Despite advances in surgeryand chemotherapy, survival of patients withEOC stands at just 45% at 5 years.1 Althoughthe age of biologic therapies holds the potentialof improved responses in advanced and recur-rent EOC, a greater impact could be made byrecognition of high-risk patients and by offer-ing risk-reducing surgery, a strategy that hasdemonstrated effectiveness in patients with ge-netic predispositions.2 However, there is signif-icant heterogeneity within the EOC group. Forexample, histologically defined subtypes suchas serous, endometrioid, mucinous, and low-and high-grade malignancies all have variableclinical manifestations and underlying molecu-lar signatures.3 Substantial advances have beenmade in understanding the genetic alterationsand biologic processes in ovarian cancer; how-ever, the etiology remains poorly understood.In this article, we will focus on the currentunderstanding of the early events in EOC.

The ovary is surrounded by a single-cell layer ofperitoneal mesothelium, which is derived from thecoelomic layer during development and which hasthe potential to undergo metaplastic transformationto a more differentiated state.4 Unlike most malig-nancies, as this epithelium transforms into a malig-nant phenotype, it becomes more differentiated,and it can differentiate toward many of the differentcell types found in the mu¨llerian tract, includingthose in the fallopian tube, uterus, cervix, andovarian stroma.5 It is widely thought that mostovarian cancers develop from the surface epithe-lium or postovulatory inclusion cysts that weresubjected to prolonged exposure to hormones orother chemokines.4

Au t hor s’ di sclosu r es of p ot en t ial co n-

ict s of in t er est an d aut h or co nt r ib u-

t io ns ar e f o un d at t he end of t hi s

ar t icl e.Co r re spo ndi ng au t hor : Ani l K . S oo d,

M D, Pr o f essor , De par t me nt s of G yne-

co log ic O ncol ogy an d Cance r Biol ogy,

Un ive rsi t y of Te xas M. D. And er so n

Can cer Cen t er , 115 5 He r man P r essl er ,

Un it 13 62 , H oust o n, TX 770 30; e- mai l:

aso od@ md ande r son. or g .© 20 08 b y Amer i can S ocie t y of Cli nical

O nco log y0 732 -1 83X / 08/ 26 06- 99 5/ $20 .0 0

Primary peritoneal and fallopian tube carcino-DO I : 1 0. 12 00/ JCO .2 006 .0 7. 997 0mas have similar clinical, molecular, and geneticprofiles to ovarian cancers, though some small dif-ferences in frequency of specific protein expressionhave been described.6 -11 Primary peritoneal carci-nomas may,in fact, have a multifocal and polyclonalorigin.12 Therefore, although these entities are oftenlumped together with ovarian cancer, there may besome significant, but currently poorly defined, dif-ferences. In fact, recent pathologic examination ofconsecutive cases of ovarian, primary peritoneal,and fallopian tube cancers suggests that a greater

995Information downloaded from jco.ascopubs.org and provided by UNIVERSITEITSBIBLIOTHEEK on March 25, 2010 from

131.174.244.21. Copyright © 2008 by the American Society of Clinical Oncology. All rights reserved.

Page 2: Early Events in the Pathogenesis of Epithelial Ovarian Cancer-printed - Copy

Landen, Birrer, and Sood

percentage of ovarian cancers than originally thought may actuallyhave a fallopian origin with metastasis to the ovary.13 However, be-cause of the changes in definition, inconsistent reporting of subtypes,and the paucity of direct comparative studies, these entities will beconsidered as variations within a disease and will be considered to-gether in this review.

increased risk of EOC by a factor of 2.8, and of borderline tumors by4.0,compared with infertile women who were not usingfertility drugs.However, subsequent case-control and cohort studies demonstratedinconsistent associations between gonadotropin use and epithelialovarian carcinoma.25 These studies collectivelysuggest that the condi-tion of infertility (or the predisposing condition), rather than fertilitydrug use, is responsible for the increased risk.2 6 From a basic scienceperspective, receptors for FSH and LH have been found on 100% ofnormal ovarian surface epithelial cells and on 60% of malignant tu-mor cells.27 FSH, LH, and human chorionic gonadotropin (hCG) allstimulate proliferation of EOCs and may activate mitogen-activatedkinase (MAPK).28 Furthermore, induced overexpression of the FSHreceptor led to upregulation of epidermal growth factor receptor(EGFR), human epidermal growth factor receptor 2 (HER2), and

There have been several proposed hypotheses about the under-

lying physiological processes that increase the risk of malignant trans-

formation of the ovarian epithelium in the 90% of EOCs that do not

have a known genetic component (Table 1). Importantly, these may

also play a role in the 10% of cases in women with a genetic suscepti-

bility through BRCA or mismatch-repair gene mutations. These hy-

potheses will be reviewed brie y, and they are discussed in greater

depth in other excellent reviews.14, 14a ,1 4bThe observation that women with a greater number of ovulatoryC-MYC.29 Other potential oncogenes upregulated by FSH or LH

treatment in vitro include -catenin, Meis-1, cyclin G2, insulin-like

growth factor 1 (IGF-1), and -1 integrin.30, 31 To date, no study has

demonstrated that exposure to gonadotropins is capable of inducing

transformation of ovarian surface epithelium (OSE) cells to a malig-

nant phenotype. However, in animal models of implanted tumors,

exposure to gonadotropins promotes tumor growth,32 angiogenesis,32

cycles have an increased risk of ovarian cancer led to the incessant

ovulation hypothesis by Fathalla in 1971.15 According to this hypoth-

esis, as ovulation occurs, ovarian surface epithelial cells are internal-

ized and damaged, and the subsequent repair mechanisms place the

cells at an increased risk of developing mutations and subsequent

malignancies. Consistent with this hypothesis, women with a history

of multiple pregnancies,16-1 8 increased time of lactation,19 and oral

contraceptive use16, 20 are all at a decreased risk. Moreover, the risk for

ovarian cancer decreases further with the increased occurrence of

each of these factors. There is also experimental evidence from

primate and other animal models that supports the incessant ovu-

lation hypothesis.21, 22 However, this theory is somewhat weakened

by observations that progesterone-only oral contraceptives, which

do not inhibit ovulation, are at least as effective as ovulation-

inhibiting contraceptives.23 Moreover, women with polycystic

ovarian syndrome, who have decreased ovulatory cycles, are at an

increased risk of EOC.24

vascular endothelial growth factor (VEGF) expression,33 and adhesion.34Collectively, these studies suggest a role for gonadotropins in promotingthe progression of ovarian cancer, rather than of the causation.

Notable hormones have also been implicated in ovarian carcino-genesis. On the basis of epidemiologic studies, progestin-only contra-ceptives are as effective as combined oral contraceptive pills in thereduction of ovarian cancer risk,23 ,35 and progesterone is the domi-nant hormone during pregnancy, which also reduces risk.23 Interest-ingly, use of progestin contraceptives can also decrease ovariantestosterone levels.36 In vitro studies have not, however,demonstrateda clear inhibition of cancer cell growth.37 Conditions of increasedandrogens (eg, polycystic ovarian syndrome, hirsutism, acne) are as-sociated with an increased risk of EOC.24 Androgens represent thegreatest hormone concentration within a developing follicle,3 8whichprolongs exposure to the epithelial cells. Androgen receptors arepresent on human OSEcells,and they stimulate proliferation.39 Thereis no strong evidence, however, that exposure to androgens inducesmalignant transformation.

Weaknesses in the incessant ovulation theoryand observations ofan increased risk in infertile women who use fertility drugs led to thegonadotropin hypothesis, which theorizes that stimulation of theovarian surface epithelium by follicle-stimulating hormone (FSH)and by luteinizing hormone (LH) may place the cells at an increasedrisk of developing EOC. In 1992, Whittemore et al16 reported a case-control studyin which infertile patients who used fertilitydrugshad an

Table 1. Hypotheses on Physiologic Susceptibilities to Epithelial Ovarian Cancer

Hypothesis Proposed Mechanism Best Evidence

Incessant ovulation OSE damaged during ovulation, with repair making Risk of EOC decreases with decreased number ofcells susceptible to mutations cycles, such as pregnancy, lactation, and OCP use

Gonadotropin stimulation Stimulatory effect of FSH and LH promote growth, Increased EOC risk with infertility, PCOS; Decreasedincreased cell divisions, and mutationsrisk with progesterone-only OCPs; FSH upregulates

many oncogenes and promotes growth in preclinicalmodelsHormonal stimulation High concentrations of androgens in the tumor Conditions of high circulating androgens (PCOS,

microenvironment promote carcinogenesis,whereas progestins decrease risk

hirsutism, acne) associated with increased risk,androgens are the dominant hormone in theinclusion cyst; progestin use decreases risk of EOC,induces OSE apoptosisIn ammation Damaged OSE with ovulation induces in ammation, Possible reduced risk with NSAID use; increased risk

which promotes reconstruction and mutationsusceptibility

with talc or asbestos; abundance of in ammatorymediators in tumors

Abbreviations: OSE, ovarian surface epithelium; EOC, epithelial ovarian cancer; OCP, oral contraceptive pill; FSH, follicle-stimulating hormone; LH, luteinizinghormone; PCOS, polycystic ovarian syndrome.

996 JO URN AL OF CL I N ICA L O N COL O GYInformation downloaded from jco.ascopubs.org and provided by UNIVERSITEITSBIBLIOTHEEK on March 25, 2010 from

131.174.244.21. Copyright © 2008 by the American Society of Clinical Oncology. All rights reserved.

Page 3: Early Events in the Pathogenesis of Epithelial Ovarian Cancer-printed - Copy

Carcinogenesis of Ovarian Cancer

There is growing interest in the etiologic role of in ammation, Earliest Recognizable Events in Tumor ProgressionEOCs, like most cancers, are thought to arise from a singlewhich accompanies each ovulation, with an associated cytokine re-

lease, in ux of in ammatory cells, and tissue reconstruction.26 Thismechanism has been postulated to stress OSE cells such that theyare predisposed to genetic damage and malignant transformation.Consistent with this hypothesis,patients with chronic aspirin,nonste-roidal anti-in ammatory drug, or acetaminophen use have a re-duced risk of EOC.40 Downstream effectors of the nonsteroidalanti-in ammatory drug pathway, such as nitric oxide synthase,cyclooxygenase-2, VEGF, and NF- B, have been implicated in carci-nogenic pathways.40 Patients exposed to in ammation-inducingagents, such as talc and asbestos, have been shown in some studies tobe at an increased risk.26 Although talc particles have been foundon human and murine ovaries after perineum exposure,41 noanimal model of ovarian carcinogenesis has been proven with talcor asbestos exposure.

multidysfunctional cell in 90% of occurrences. Evidence for theclonality of ovarian cancer lies in the similarity between primaryand metastatic lesions during the examination of the loss of het-erozygosity (LOH), X-chromosome inactivation, and specific genemutations.4 2 The difficulty in describing the earliest events in ovar-ian cancer is in the limited availability of early-stage tumors, theheterogeneity among individuals, and the genetic instability oftumors, which makes it difficult to know if detected mutations areearly or late occurrences.

Genomic comparison of early- versus late-stage, high-grade ovariancancers. Genomic analysis of high-grade tumors has identified am-plification and/or over-expression of numerous genes thought to beimportant in the development of ovarian cancer.However,the preciserole of these genes in earlycarcinogenesis remains unclear. The appli-cation of new genomic technologies, such as comparative genomehybridization (CGH) and microarray expression profiling, has helpedelucidate many of the important genetic events that may lead toovarian cancer. The ability of these technologies to simultaneouslymeasure thousands of genes allows not only the identification ofindividual genes but also the delineation of dominant pathways thatmay be responsible for cancer pathogenesis43 ,44 (Fig 1).

Although any of the above mechanisms may play a role in ovar-ian carcinogenesis in some patients, the modest association with eachsuggests that multiple other processes are involved, which cannot bepredicted by clinically recognizable conditions such as nulliparity,infertility, or hormone exposure. To detect EOC early or to identifyat-risk patients, a search must therefore continue for genetic or epige-netic conditions that predispose patients to the development of EOCor for proteins that may allow for early detection.

A study that compared normal ovarian epithelial cells to early-and late-stage cancers found several differentially expressed genes

Thrombin

Beta-3integrin Alpha-5

MT-SP1 ExtracellularintegrinPAR1 matrix

MAGP2G GPAR2

GHEF1

SNX1 GPRK5FAK YES

Fig 1. Pathway identification by microar-ARHIray analysis. Schematic representation of

potential signaling pathways in ovarian

cancer, identified by incorporating the mi-

croarray results (genes differentially ex-

pressed between normal and malignant

ovarian epithelial cells) into PathwayAssist.

Genes in red are upregulated in cancer

compared with normal ovarian epithelium;

genes in green are downregulated in can-

cer; genes in yellow did not show a signif-

icant difference between specimens.

Reproduced with permission.43

Cell cycle RACI CDC42ERKprogression GD P GTP GD P G TP

VAV3 G

CCND1C CND 1 CytoskeletonGmodulation and IAP

enhanced GATA6 MMPmotilityproductionD OC-2 DOC-2 TSP-1

MTI-MMP

RECK InvasionETAR

ET-1

www.jco.org 997Information downloaded from jco.ascopubs.org and provided by UNIVERSITEITSBIBLIOTHEEK on March 25, 2010 from

131.174.244.21. Copyright © 2008 by the American Society of Clinical Oncology. All rights reserved.

Page 4: Early Events in the Pathogenesis of Epithelial Ovarian Cancer-printed - Copy

Landen, Birrer, and Sood

been well studied beyond a description of mismatch repair defects.Other familial syndromes associated with an increased risk of ovariancancer include Peutz-Jeghers Syndrome (ie, mutation in the STK11

between normal and malignant tissues.4 5 However, the early- and

late-stage tumors were remarkably similar. This seems to be at odds

with the concept of early-stage tumors that evolve into late-stage ones.

However, in the same study, CGH analysis demonstrated acquired

gene abnormalities in late-stage tumors, which was more consistent

with tumor evolution.Another study that compared tumors collected

from the ovary or the omentum identified a 27-gene signature that

could differentiate between the primary and metastatic tumor.46

gene; 21% lifetime risk) and Gorlin Syndrome (ie, mutation in PTCH;20% lifetime risk), but these tumors are usually stromal cancers andfibromas, respectively.

Animal models. In an attempt to better understand ovariancarcinogenesis, several animals models have been developed. Orsulicet al5 9 introduced various oncogenes into transgenic ovarian surfaceepithelial cells that expressed the avian receptor TVA. These cellsbecame tumorigenic when two of three genes (C-MYC, K-RAS,or

Many of the genes are involved in the p53 pathway, which suggeststhat this pathway is important for the peritoneal metastasis. The chal-lenge is in determining whether a noted difference is truly responsiblefor a particular function, such as malignant transformation or metas-tasis.For example, metastasized tumors with genetic instability wouldcontinue to acquire genetic mutations that could be erroneously as-signed to causingmetastasis. Additionally, early genetic perturbationswould persist in metastasized tumors and would not be identified asan earlyevent when comparing early-and late-stage tumors. However,with validation by additional studies that use larger sample sizes,various array platforms to account for methodologic inconsistencies,and microdissected samples to differentiate tumoral and stromal al-terations, these technologies willallowmore information to be gainedon the earliest events in ovarian cancer.

AKT) were overexpressed in p53-deficient cells. After inducingchanges in vitro, they were implanted into the bursal sac that sur-rounds the ovary of recipient mice, and they developed a carcinoma-tosis pattern similar to human ovarian cancer. Subsequently,Connolly et al60 generated de novo ovary-specific tumors in trans-genic mice that expressed the transforming region of the SV40T-antigen under control of the ovary-specific Mu

¨llerian inhibitorysubstance type II receptor gene promoter.In these mice, poorlydiffer-entiated tumorsof both ovaries developed in 50% of transfected miceand often led to carcinomatosis and ascites formation. A model ofendometrioid ovarian carcinogenesis was described by Dinulescu etal,6 1 in which adenoviral vectors were injected into the bursal sac thatinduced K-RAS overexpression and PTEN inactivation.61 Although

Inherited disorders. A study of genetic disorders can providegreat insight into the etiology and early events in carcinogenesis. He-reditary genetic disorders account for approximately 10% of ovariancancers, and 90% of these are either BRCA1 or BRCA2 mutations.Evaluation of BRCA1 and BRCA2 mutant and sporadic tumors withgene expression profiling has demonstrated that the greatest contrastin expression patterns wasbetween that of BRCA1 and BRCA2 mutanttumorsand that sporadic tumorsshared characteristicsof both.47 Thisintriguing finding suggests that BRCA1 and BRCA2 tumors may havevariable pathways in carcinogenesis and that even sporadic tumorsmay develop as a result of alterations in either pathway. Clinically,patients with BRCA mutations tend to have highly proliferative tu-morsbut more favorable outcomes when adjusted for stage.48 Border-line tumors have a much less frequent incidence of BRCA mutations(4.3% v 24.2% in a Jewish population),49 which also suggests a differ-ent molecular origin.

K-RAS overexpression alone induced lesions that were histologicallycompatible with endometriosis, the combination of both mutationsled to the rapid development of carcinomatosis of endometrioid his-tology. Although these models have limited applicability to de novohuman ovarian cancers because of their different genetic composition,such as greater homogeneity, diploid status (rather than aneuploid),and progression with few mutations, they can provide useful insightsinto specific gene functions.

TWO-PATHWAY MODEL OF OVARIAN CANCER

With the recognition that ovarian tumors are heterogeneous and

generate a wide spectrum of disease states, there is growing clinical,

translational, and genetic evidence to support at least two broad cate-

gories of carcinogenesis.62 High-grade malignanciesare rapidlygrow-

ing, relatively chemosensitive, and without a definitive precursor

lesion. In contrast, low-grade tumors grow more slowly, are less re-

sponsive to chemotherapy, and share molecular characteristics with

low-malignant potential (LMP) neoplasms. Clinically, in a large series

of 112 low-grade patients observed for a median of 71 months, the

average age at diagnosis was 43 years (compared with 61 years for all

ovarian cancers), and the median survival was 81 months63—much

longer than the 57- to 65-month survival observed in phase III trials

that define the standard of care in EOC.64 ,65 Pathologic analysis has

found that approximately 60% of low-grade serous carcinomas also con-

tain areas of serous LMP tumors compared with just 2% of high- grade,66

Other than in hereditary syndromes, BRCA genes are rarely mu-tated in sporadic ovarian cancers,50 although epigenetic changes, al-ternate splicing,and othergenetic factors mayaffect BRCA function inas manyas 82% of sporadic occurrences.5 1-53 The BRCA1 and BRCA2proteins are considered caretakers of the genome,and playkey roles in

the signaling of DNA damage, the activation of DNA repair, the

induction of apoptosis, and the monitoring of cell cycle check-

points.54 -56 Cells that lack functional BRCA have increased aneu-

ploidy, centrosome amplification, and chromosomal aberrations,57which make them susceptible to further mutations. BRCA appears to

function as a cofactor for a variety of transcription factors, including

p53, STAT1, c-Myc, JunB, ATF-1, and others.57Defects in mismatch repair in patients with Lynch syndrome orand LMP tumors recur as a low-grade carcinoma in 75% of cases.67hereditary nonpolyposis colon cancer (HNPCC) account for approx-

imately10% of hereditaryovarian cancers and for 1% to 2% of overallcases. Patients with this syndrome, however, individually carry anapproximately 12% risk of developing ovarian cancer.58 The mecha-nism of increased risk is through defects in the mismatch-repair ma-chinery and its resulting genetic instability that places cells at risk ofmultiple mutations; however,carcinogenesis in ovarian cancerhas not

Molecular and protein analyses of tumors of these two differentsubtypes also suggest different pathogenesis (Table 2). Analyses ofindividual genes have found that K-RAS and BRAF mutations arerarely detected in high-grade invasive carcinomas but are present in30% to 50% of LMP tumors, in low-grade adenocarcinomas, andoften in the adjacent benign epithelium.62 ,68 -70 The P53 gene is mu-tated in 50% to 80% of high-grade invasive carcinomas, but rarely in

998 JO URN AL OF CL I N ICA L O N COL O GYInformation downloaded from jco.ascopubs.org and provided by UNIVERSITEITSBIBLIOTHEEK on March 25, 2010 from

131.174.244.21. Copyright © 2008 by the American Society of Clinical Oncology. All rights reserved.

Page 5: Early Events in the Pathogenesis of Epithelial Ovarian Cancer-printed - Copy

Carcinogenesis of Ovarian Cancer

GENETIC AND PROTEIN ABERRATIONS IN OVARIAN CANCER

Table 2. Variability in Biology of Low- and High-Grade TumorsCharacteristic LMP/Low-Grade (%) High-Grade (%)The majority of evidence on genetic or protein alterations in ovarian

cancer is based on studies of late-stage cancers. However, currentunderstanding of these processes allows speculation that many alter-ations must occur early to achieve a clinically recognized tumor. It isbelieved that, for the majority of malignancies, a cancer cell mustovercome many protective mechanisms to develop into a clinicallyevident tumor.84 These include unchecked proliferation, inhibition ofapoptosis, angiogenesis, stromal invasion, separation and survivalaway from the primary tumor, and implantation and growthwithin new tissues. We examine the evidence for many of theever-increasing recognized participants in each of these processesin ovarian cancer (Table 3).

p53 inactivity Rare 50-80HLA-G overexpression Rare 61HER2 overexpression Rare 20-66AKT overexpression Rare 12-30Apolipoprotein E expression 12 66B-RAF mutation 30-50 RareK-RAS mutation 30-50 RarePTEN mutation 20* RareMSI 50* 8-28

Abbreviations: LMP, low malignant potential; HER2, human epidermalgrowth factor receptor 2; MSI, microsatellite instability.

*Endometrioid.

Self-sufficiency in growth signals. A number of oncogenes have

been identified in ovarian cancer that allow cells to grow indepen-

dently from the host’s signals. One of the first oncoproteins described

was src, a nonreceptor tyrosine kinase that participates in multiple

carcinogenic pathways and promotes proliferation,adhesion, cellsur-

vival, and angiogenesis.85-8 7 The overexpression of src has been dem-

onstrated in 93% of advanced-stage ovarian tumors and in more than

80% of cell lines.88 This oncoprotein promotes both platinum and

taxane resistance and survival in ovarian cancer cell lines.89 Further-

more, inhibition of src with antisense or with small molecule inhibi-

tors has reduced ovarian cancergrowth in preclinicalmouse models.85

other subtypes or LMPs.71 -73 HER2 and AKT are overexpressed in

20% to 67% and 12% to 30% of high-grade carcinomas, respectively,

but rarely in low-grade and LMP tumors.74, 75 Overexpression of hu-

man leukocyte antigen-G (HLA-G), which may provide a mechanism

of immune escape for the tumor, hasbeen noted in 61% of high-grade

carcinomas but is absent in low-grade or LMP neoplasms.76Whole-genome approaches have also provided key insights into

the developmental relatedness of various ovarian tumors. Compari-

son of whole-genome expression profiles of ovarian tumors of differ-

ent grades reveals that LMP tumors are quite distinct from invasive

cancers, and hierarchical clustering demonstrates that they group

closer to the normal ovarian epithelium than to invasive cancers.3, 77

The type I tyrosine kinase receptor family HER (ie, Erb) consists

of four known monomers: EGFR (ie, Erb1/HER1), HER2 (encoded

by the proto-oncogene neu), HER3, and HER4. EGFR is expressed on

the normal human ovarian surface epithelium (as detected by immu-

nohistochemistry) and is overexpressed in 35% to 70% of EOCs.90Furthermore, low-grade invasive cancers were indistinguishable from

borderline tumors but were distinct from high-grade tumors. More

detailed analyses have identified specific pathways, which correlate

with each specific tumor type. One predominant pathway present in

LMP tumors and low-grade tumors is a functional wild-type p53

pathway, which is absent in high-grade tumors.3 This suggests that

inactivation of p53 is a key branch point, in which an intact p53

pathway could lead to LMP/low-grade tumors, but disfunctional p53

could lead to high-grade cancers. In other genomic studies, LOH78

HER2 has no extracellular ligand-binding domain, but it is activated

when dimerized with other type I receptors. HER2 expression in

ovarian cancer varies widely; overexpression is found in 20% to 30%

of cases.9 1 Many proliferation pathways mediate signals through the RAS

oncoprotein, a G-protein attached to the cell membrane and activated

by many tyrosine kinase receptors. RAS activates a cascade of serine/

threonine and tyrosine nonreceptorkinases, which leads to phosphor-

ylation and activation of Erk1 and Erk2 transcription factors that

make their way to the nucleus to initiate signals of growth and pro-

gression through the cell cycle. As described above, K-RAS mutations

are common in adenocarcinomas, and frequency is variable in differ-

ent histologic subtypes.7 0,8 2

and CGH79 analyses have found similar profiles in benign adenomasand in LMP tumors, which supports the concept of a transformationfrom benign adenoma to LMP.

Although ovarian adenocarcinomas can be subtyped by grade,

histologic subtypes also differ. Although differences in clinical out-

comes among serous,endometrioid, and mucinous adenocarcinomas

are not as dramatic as those between high- and low-grade cancers,

genomic studies have demonstrated that mucinous adenocarcinomas

often harbor mutations and have differential gene expression similar

to LMP tumors and to benign cystadenomas.80 ,81 Specifically, muta-

tions in K-RAS have been described in 61% of borderline tumors, in

68% of low-grade tumors, and in 50% of mucinous adenocarcinomas,

but only in 5% of high-grade serous carcinomas.7 0,8 2 These studies

suggest that the malignant transformation in mucinous tumors may

follow a sequence of adenoma to LMP tumor to invasive adenocarci-

noma8 0,8 1 more frequently than to high-grade serous carcinomas.

Endometrioid adenocarcinomas more frequently harbor PTEN mu-

tations(similar to endometrioid tumorsof the uterine endometrium)

than do serous or mucinous subtypes.83

Resistance to antigrowth signals. In early-transformed cells, anti-growth signals must be overcome.Although definitive data are lackingregarding the sequence of specific genetic events in carcinogenesis,there is evidence for abnormalities in cell cycle mediators, such ascyclins, cyclin-dependent kinases (CDKs, which complex with thecyclins to allow their activity), CDK inhibitors (CDKIs, which inhibitcyclin/CDK complexes), and other proteins or transcription factorssuch as pRb, p53, and E2F. The restriction point, after which a cell iscommitted to divide, is controlled by Cyclin D and E’s regulation ofE2F release by Rb. Cyclin Eis expressed by only 9% of benign tumorsbut by 48% of borderline and by 70% of malignant tumors, and it isassociated with poor survival.92 Similarly, CDK2, which complexesexclusively with Cyclin E, is expressed more frequently in malignantovarian tumors compared with LMPor benign tumors.92 Cyclin D1 is

www.jco.org 999Information downloaded from jco.ascopubs.org and provided by UNIVERSITEITSBIBLIOTHEEK on March 25, 2010 from

131.174.244.21. Copyright © 2008 by the American Society of Clinical Oncology. All rights reserved.

Page 6: Early Events in the Pathogenesis of Epithelial Ovarian Cancer-printed - Copy

Landen, Birrer, and Sood

Table 3. Select Contributors to Ovarian Carcinogenesis

Protein/Gene Function Rate in EOC (%)

Growth Promotion

EGFR (HER1) Membrane TK receptor, promotes cell growth 35-70HER2 Membrane TK receptor, promotes growth 20-66*Src TK, promotes growth, angiogenesis, survival 80-90CSF-1/fms Ligand/receptor, inhibits anoikis 50-70IGF/IGFR Peptide hormone/receptor, promotes growth 21-25K-RAS G-protein, promotes growth through MAP kinase pathway 30-50†BRAF Promotes growth through MAP kinase pathway 30-50†

Insensitivity to Antigrowth Signals

TGF- Ligand, inhibits growth through Rb activation Lost in 40%C-MYC Transcription factor, cell cycle mediator 30Cyclin D/CDK4/6 Advance from G1 to S phase 30-90Cyclin E/CDK2 Advance from G1 to S phase 30-70Cyclin B/CDK1 Advance cell cycle into M phase 80p16 Inhibits Cyclin D/Cdk4/6 Lost in 30%p27 (kip-1) Inhibits Cyclin E/Cdk2 Lost in 55%p21 (WAF-1) Inhibits Cyclin B/Cdk1 Lost in 25%–40%NF B Transcription factor, effector of many survival pathways UnknownNOEY(ARHI) GTPase tumor suppressor, induces apoptosis through p21 40‡

Inhibition of Apoptosis and Immune Surveillance

PIP3/AKT AKT (activated by PIP3) inhibits apoptosis 12-18*PTEN Decrease AKT 20§p53 Promotes cell cycle arrest/apoptosis with DNA damage 50-90*BRCA1 Cofactor for transcription factors, caretaker of genome 6-82¶BRCA2 Cofactor for transcription factors, caretaker of genome 1-3MLH1/MSH2 Mediates mismatch repair, promotes genetic stability 30§Fas ligand Produced by tumor cells to induce apoptosis of T-cells 50-80HLA-G Secreted by tumor cells to inhibit cytotoxic immune cells 61*

Limitless Replicative Potential

hTERT Subunit of telomerase, maintains telomere length 80-85Enhanced Angiogenesis

VEGF/VEGFR Ligand/receptor complex induces angiogenesis 40-100IL-8 Cytokine promoting angiogenesis UnknownEphA2 TK promoting angiogenesis and vasculogenic mimicry 76

Promotion of Invasion and Metastasis

MMPs Degrade extracellular matrix 40-100v 3 Integrin, promotes survival and angiogenesis 95

FAK Cofactor TK promotes adhesion, proliferation, survival 70E-cadherin Promotes adhesion 90-100

Abbreviations: EOC, epithelial ovarian cancer; EGFR, epidermal growth factor receptor; HER, human epidermal growth factor receptor; TK, tyrosine kinase; IGF,insulin-like growth factor; TGF, transforming growth factor; CDK, cyclin-dependent kinase; VEGF, vascular endothelial growth factor; MMPs, matrix metalloprotein-ases; FAK, focal adhesion kinase.*High-grade serous.

†Low-grade serous.‡40% loss of heterozygocity.§Endometrioid.¶Inherited mutation in 6%–7% of all cancers; may play a role in 82% of sporadic cancers.

cycle regulation that likely provide an unchecked growth advantage toovarian cancer cells.

expressed at low levels in normal ovarian epithelial cells but is prom-

inent in ovarian cancer cells (89% cytoplasmic; 30% nuclear).93 CDK1

complexes with cyclin B to regulate entry into the M phase, and it is

expressed at high levels in 80% of ovarian cancers, although absent

from normal epithelium.94

Evading apoptosis. It has been proposed that a more importantcharacteristic of cancer than increased cell division is the reducedapoptosis and prolonged survival seen in these cells. Indeed, cancercells often divide less frequently than their normal equivalents, espe-cially in epithelial cancers, in which normal epithelial cells have rapidturnover. Many participants in this process are altered in ovariancancer to inhibit cell death. Among these is P53, which normallypromoteseither cell cycle arrest and initiation of repairmechanismsor

Other proteins that control the cell cycle include myc (an onco-genic transcription factor activated by the RAS-RAF pathway andoverexpressed in approximately 30% of ovarian cancers) and AHRI(ie,NOEY2,a GTPase tumor suppressorgene lost in almost all ovariancancers95, 96).Thus,there are multiple aberrations in the geneticsof cell

1000 JO URN AL OF CL I N ICA L O N COL O GYInformation downloaded from jco.ascopubs.org and provided by UNIVERSITEITSBIBLIOTHEEK on March 25, 2010 from

131.174.244.21. Copyright © 2008 by the American Society of Clinical Oncology. All rights reserved.

Page 7: Early Events in the Pathogenesis of Epithelial Ovarian Cancer-printed - Copy

Carcinogenesis of Ovarian Cancer

the shunting of the cell to an apoptotic pathway.97 It has been hypoth-esized that cancers that do not have mutations in the P53 gene likelyhave alterations in the function of p53 in other ways, such as in theproduction of p53-binding proteins or the enhanced degradationthrough ubiquitination. Most P53 mutations in ovarian cancer aremissense,9 8 but specific mutations (ie, null mutations) may play a keyrole in producing a metastatic phenotype, in that they are seen muchless frequently in stage I ovarian cancers.99 Interestingly, P53 muta-tions have been detected in ovarian inclusion cysts adjacent to cysta-denocarcinomas, in microscopic ovarian cancer, and even intubular intraepithelial carcinomas removed prophylactically frompatients with BRCA1 mutations.1 3, 100 The accumulation of evi-dence suggests that p53 inactivation may be a relatively early eventin ovarian cancer pathogenesis.

sites. Although metastasis is thought of as a late event in carcinogene-sis, emergingevidence in breast cancer suggests that early tumorsmayalready hold the genetic profile needed for metastasis,108 which furthersuggests that factors other than the tumor cell itself may regulatemetastasis. Similarly, in ovarian cancer, peritoneal and stromal alter-ations may be permissive for cancer spread.1 09 An understanding ofthese factors mayprovide additional insights into tumorpathogenesisand also may offer unique targets for therapy. No cells, cancerous or benign,can exist without oxygen and other

nutrients. Cells must reside within 100 m of a capillary in order to

receive oxygen.110 Therefore, in order for a malignancy to grow be-

yond approximately 1 mm3, it must induce the growth of new vessels

in or around itself. Regulation of angiogenesis is complex, which

re ects a balance between pro- and antiangiogenic in uences within

the tumor microenvironment. The primary mediator of angiogenesis

is VEGF-A,11 1,1 12 which is known to increase vascular permeability,

stimulate endothelial cell proliferation and migration, alter endothe-

lial cell gene expression, and protect endothelial cells from apopto-

sis.113 ,11 4 VEGF expression strongly correlates with ovarian cancer cell

linesthat induce ascites and carcinomatosis,115 and increased circulat-

ingand tumor VEGF levels are associated with the clinical outcome of

ovarian cancer patients.116 ,11 7

The PI3-kinase/AKT pathway is upregulated in approximately

30% of ovarian cancers.74 Activators of thispathway inhibit apoptosis,

but they also have been shown to increase neovascularization, enhance

invasion, and increase resistance to chemotherapeutic agents.101 Con-

trol of the balance in this pathway lies primarily with PTEN, a tumor

suppressor that dephosphorylates PIP3 back into PIP2, promoting

apoptosis. The PTEN mutation is a frequent finding in endometrioid

ovarian cancers, and animal models suggest that it may be an early

event in ovarian carcinogenesis,at least of the endometrioid subtype.61Mediators of angiogenesis include tumor-derived factors and

NF B is the primary member of a family of five transcriptionhost stromal factors. Interleukin-8 plays a significant role in neovas-

cularization and ovarian cancer growth11 8 and is elevated in patients

with both early- and late-stage cancers.119 The v 3 integrin is pri-

marilyexpressed on newlydeveloping vascular endothelial cells, but it

isalso expressed on ovarian tumor cells.12 0 The tyrosine kinase recep-

tor EphA2 is overexpressed by 75% of ovarian cancers,12 1 and its

inhibition reduces tumor growth, at least in part through antiangio-

genic mechanisms.12 2,1 23 From a translational perspective, patient-

specific tumor microenvironment characteristics may in uence the

response to antiangiogenic therapy.1 24, 125

factors that deliver signalsto the nucleus to both increase proliferation

and inhibit apoptosis. NF B activation upregulates expression of

Bcl-2 family members, inhibitor of apoptosis proteins (IAP), and

additionalgenes identified by cDNA microarrayanalysisthat mayplay

a role in ovarian cancerpathogenesis.102 NF Bblockade also decreases

VEGF and interleukin-8 production and decreases tumorigenicity of

ovarian cancer cell lines in mice.103Limitless replicative potential. Normal cells can only divide a setnumber of timesbefore they achieve senescence and undergo apopto-sis. The clock for this pathway lies in telomere caps on the ends ofchromosomes that are made up of DNA and associated proteins.Without the protection provided by telomeres, exposed chromo-somes undergo massive defects, activating p53 and other policingproteins that propela cellinto an apoptotic pathway.Most cancer cells(75% to 90% of all types; 81% to 86% of those in ovarian cancer)maintain telomere length byproduction of telomerase, a reverse tran-scriptase composed of an RNA component (hTR) and a catalyticsubunit (hTERT).104 The hTR subunit is expressed by all cells, buthTERT expression increases with increasing tumorigenicity, whichsuggests that it is the rate-limiting step in telomerase activity.1 05 Find-ings that P53 knockdown and hTERT expression alone can transformovarian surface epithelial cells106 and that functional BRCA inhibitstelomerase activity107 suggest that telomerase activation is an earlyandrequired event for carcinogenesis.

The all-important first step in metastasis, and the primary feature

that defines malignancy,isinvasion through the basement membrane,

which requires interplay between tumor cells and the permissive un-

derlying stroma. Invasion of malignant cells through the basement

membrane and endothelial cell migration for angiogenesis require

degradation of the extracellular matrix. Matrix metalloproteinases

(MMPs) are a family of zinc-dependent endopeptidases that digest

collagen and other extracellular matrix components. They also stim-

ulate proliferation and induce release of VEGF.126 Ovarian tumors

overexpress MMP-2 and MMP-9,1 27 and this increased expression

correlates with clinical stage12 8 and patient survival.1 29 Interest-

ingly, host production of MMPs may be more important than

production by tumor cells, as demonstrated by Huang et al130 in

MMP-knockout mice. Another potentiator of invasion is host

production of catecholamines through chronic stress. A growing

body of preclinical data support the theory that chronic stress

contributes to the initiation and progression of cancer though

activation of adrenergic receptors, which leads to increased inva-

sion and metastasis.131 ,1 32 These mechanistic data support epide-

miologic studies that show that patients with poor social support

and increased stress are at greater risk for cancer progression.1 33

Early events in the tumor microenvironment: angiogenesis, inva-sion, and metastasis. A growing body of evidence suggests that, al-though genetic events in the tumor cells themselves are certainlycrucial, host and stromal factors in the tumor microenvironment areequally important. A clinically significant tumor includes not onlytumor cells but also matrixcomponents, stromal cells, and in amma-tory cells. An interplay between tumor cells and surrounding normaltissue dictates the establishment of a vascular supply through angio-genesis, invasion into the surrounding stroma, penetration of lym-phatic and vascular spaces, and adhesion and growth at metastatic

In ammatory cellsand associated cytokinesplay significant rolesin the tumor microenvironment. Because tumor cells can produceproteins that are recognized as abnormal,theycan induce an immuneresponse that can result in tumor cell death. As such, many functions

www.jco.org 1001Information downloaded from jco.ascopubs.org and provided by UNIVERSITEITSBIBLIOTHEEK on March 25, 2010 from

131.174.244.21. Copyright © 2008 by the American Society of Clinical Oncology. All rights reserved.

Page 8: Early Events in the Pathogenesis of Epithelial Ovarian Cancer-printed - Copy

Landen, Birrer, and Sood

of tumor cells serve to evade recognition and destruction by immune

cells, such as Fas ligand production to induce lymphocyte apoptosis1 34

assess, but positive peritoneal cytologyis detectedinapproximately

30% of stage I cancers.14 1 Given the shedding nature of ovarian

cancer, adhesion molecules in particular have been evaluated for

their role in peritoneal metastasis. Evidence for mediators of this

process playing a role in early carcinogenesis is lacking but may

include such promoters of cell survival as focal adhesion kinase

(FAK) andE-cadherin.142 -1 45 E-cadherin is uniformly expressed in

ovarian cancer, in low–malignant-potential tumors, in benign

neoplasms, and—notably—in inclusion cysts of normal ovaries,

but not in the normal surface epithelium.1 46

and HLA-G secretion to inhibit natural-killer cell activity.7 6,1 35 Cyto-

kine production by mesenchymal cells stimulates ovarian epithelium

and activates processes that may participate in malignant transforma-

tion.13 6 Moreover, cytokine production by tumor cells promotes

growth and inhibits apoptosis.137 As a testament to the importance of

the host antitumor immune response, increased T-cell infiltration

into the tumor is associated with improved survival.138 The role of

specific immune cell populations in controlling versus promoting

tumor growth remains to be fully defined.13 9Although the definition of an advanced stage requires meta-Proposed Model of Ovarian Carcinogenesisstatic spread of cancer cells, recent evidence suggests that metasta-

sis is an earlier event than previously thought.10 8 However, few (0.01%) of shed malignant cells are capable of metastasizing, andeven the persistent presence of cancer cells in the vasculature doesnot necessarily result in seeding to distant sites.14 0 The patterns ofmetastasis with EOC are different than those of most cancers.Release of malignant cells by early-stage cancers is difficult to

The increasing knowledge about early genetic events in ovariancarcinoma cells has provided a better understanding of factors thatmay induce malignant transformation of the normal ovarian epithe-lium. However,we propose that, in a comprehensive model of ovariancarcinogenesis, components that arise in (or are deposited to) thestroma, such as in ammatory cells and immune modulators, MMPs,

Normal ovarian Fig 2. Proposed model of ovarian carcino-surface epithelium

and inclusion cystsgenesis. Normal ovarian epithelium is ex-posed to physiologic processes that maypredispose to malignant transformation,such as prolonged androgen exposure. Anumber of characteristics must be ob-tained, primarily through mutations orother genetic changes, to be transformedto a malignant state. These include unreg-ulated growth, resistance to antigrowthsignals, inhibition of apoptosis, evasion ofrecognition by the immune system,achieving limitless replicative potential, in-duction of angiogenesis, and invasion ofthe basement membrane. Examples ofspecific proteins known to play a role ineach of these processes in ovarian cancerare listed in italics. The order in whichthese mutations may occur is not wellunderstood, but the timing and specificprotein affected may be significant in pro-ducing different histologic subtypes andgrades of ovarian cancer. For example, ifmutations favoring growth and resistanceto apoptosis occurred early, before achiev-ing the potential for invasion and metasta-sis, an intermediate pathologic subtypewould be noted more often, such as K-RAS

Predisposing events suchas androgen exposure

High-grade Low-grade EGFR, KRAS,HER2 BRAFpathway pathway

GrowthfactorsAKT2 PTEN

Inhibition of No perceivable apoptosis

intermediate LMPhistology

Tum orAngiogenesis – VEGF, IL-8

Limitless replicative potential – hTERT

Genetic instabilityp53, BRCA MSI

Immune escape – FasL, HLA-G

Microenvironment effects – MMP’smutations in low malignant potential (LMP)tumors. A mutation leading to genetic in-stability, such as P53, that occurred earlywould predispose cells to other mutations,and rapid progression to a metastatic phe-notype, as seen in high-grade malignan-cies. Permissive or contributing factors ofthe microenvironment, such as productionof matrix metalloproteinases (MMPs) byfibroblasts (pictured in red), infiltration ofin ammatory cells (pictured in blue), andproliferation of endothelial cells for angio-genesis, may be just as important as mu-tations in the tumor cells.

Anoikis resistance – FAK, av 3

Reattachment and growth

Metastasis tobowel and omentum

1002 JO URN AL OF CL I N ICA L O N COL O GYInformation downloaded from jco.ascopubs.org and provided by UNIVERSITEITSBIBLIOTHEEK on March 25, 2010 from

131.174.244.21. Copyright © 2008 by the American Society of Clinical Oncology. All rights reserved.

Page 9: Early Events in the Pathogenesis of Epithelial Ovarian Cancer-printed - Copy

Carcinogenesis of Ovarian Cancer

and integrin ligands, may be equally important for tumor establish-ment and growth (Fig 2). Within the broad dual pathway model, it isclear that several characteristics must be acquired by the tumor celland its environment. Although the order in which these occur is likelyvariable, early alterations in dominant genes may dictate the specificpath that is followed, such as K-RAS leading to an LMP tumor andearly occurrence of a P53 or BRCA alteration leading to genetic insta-bility and rapid progression to a high-grade phenotype. Characteris-tics common to both pathways include the evasion of immunesurveillance, the invasion into the stroma, survival in the peritonealcavity, attachment to intraperitoneal sites, and continued growth andangiogenesis. These additional steps likely require a longer period oftime in LMPand low-grade malignancies, but theyalso occur eventu-ally and lead to relentless growth and metastasis. Despite many over-lapping features, every malignancy is unique, and myriad yet-unidentified genetic alterations probably participate in ovariancarcinogenesis. The challenge remains to identify the most importantinitial alterations in ovarian cancer to allow the development of better

methods for early detection and for the targeting of key pathwayswhile patients are still amenable to a cure.

AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

The authors indicated no potential con icts of interest.

AUTHOR CONTRIBUTIONS

Conception and design:Charles N. Landen, Anil K. SoodAdministrative support: Anil K. SoodCollection and assembly of data: Charles N. Landen, Michael J. Birrer,Anil K. SoodData analysis and interpretation: Charles N. Landen, Anil K. SoodManuscript writing: Charles N. Landen,Michael J. Birrer, Anil K. SoodFinal approval of manuscript: Charles N. Landen, Michael J. Birrer,Anil K. Sood

serous carcinoma: Evidence for a causal relation-ship. Am J Surg Pathol 31:161-169, 2007

25. Brinton LA, Lamb EJ, Moghissi KS, et al:

REFERENCES Ovarian cancer risk after the use of ovulation-stimulating drugs. Obstet Gynecol 103:1194-1203,2004

14. Fleming JS, Beaugie CR, Haviv I, et al: Inces-sant ovulation, in ammation and epithelial ovarian

carcinogenesis: Revisiting old hypotheses. Mol CellEndocrinol 247:4-21, 2006

1. Jemal A, Siegel R, Ward E, et al: Cancer26. Ness RB, Cottreau C: Possible role of ovarianstatistics, 2007. CA Cancer J Clin 57:43-66, 2007

epithelial in ammation in ovarian cancer. J NatlCancer Inst 91:1459-1467, 1999

2. Rebbeck TR, Lynch HT, Neuhausen SL, et al:14a. Gadducci A, Cosio S, Gargini A, et al: Sex-

Prophylactic oophorectomy in carriers of BRCA1 orBRCA2 mutations. N Engl J Med 346:1616-1622,2002

27. Zheng W, Lu JJ, Luo F, et al: Ovarian epithelial

steroid hormones, gonadotropin and ovarian carcino-genesis: A review of epidemiological and experimentaldata. Gynecol Endocrinol 19:216-228, 2004

tumor growth promotion by follicle-stimulating hor-mone and inhibition of the effect by luteinizinghormone. Gynecol Oncol 76:80-88, 2000

3. Bonome T, Lee JY, Park DC, et al: Expression14b. Capen CC: Mechanisms of hormone-profiling of serous low malignant potential, low-

grade, and high-grade tumors of the ovary. CancerRes 65:10602-10612, 2005

28. Choi KC, Kang SK, Tai CJ, et al: Follicle-

mediated carcinogenesis of the ovary. ToxicolPathol 32:1-5, 2004 (suppl 2)

stimulating hormone activates mitogen-activatedprotein kinase in preneoplastic and neoplastic ovar-ian surface epithelial cells. J Clin Endocrinol Metab87:2245-2253, 2002

4. Auersperg N, Wong AS, Choi KC, et al: Ovarian 15. Fathalla MF: Incessant ovulation: a factor insurface epithelium: Biology, endocrinology, and pa-thology. Endocr Rev 22:255-288, 2001

ovarian neoplasia? Lancet 2:163, 197116. Whittemore AS, Harris R, Itnyre J: Character-

29. Choi JH, Choi KC, Auersperg N, et al: Overex-

5. Naora H: Developmental patterning in theistics relating to ovarian cancer risk: Collaborativeanalysis of 12 US case-control studies. II. Invasiveepithelial ovarian cancers in white women. Am JEpidemiol 136:1184-1203, 1992

pression of follicle-stimulating hormone receptoractivates oncogenic pathways in preneoplastic ovar-ian surface epithelial cells. J Clin Endocrinol Metab89:5508-5516, 2004

wrong context: The paradox of epithelial ovariancancers. Cell Cycle 4:1033-1035, 2005

6. Kowalski LD, Kanbour AI, Price FV, et al: Acase-matched molecular comparison of extraovarian

versus primary ovarian adenocarcinoma. Cancer 79:1587-1594, 1997

17. Risch HA, Marrett LD, Howe GR: Parity, con-30. Tashiro H, Katabuchi H, Begum M, et al: Rolestraception, infertility, and the risk of epithelial ovar-

ian cancer. Am J Epidemiol 140:585-597, 1994of luteinizing hormone/chorionic gonadotropin re-ceptor in anchorage-dependent and -independentgrowth in human ovarian surface epithelial cell lines.Cancer Sci 94:953-959, 2003

7. Halperin R, Zehavi S, Hadas E, et al: Immuno-

18. Riman T, Dickman PW, Nilsson S, et al: Riskhistochemical comparison of primary peritoneal andprimary ovarian serous papillary carcinoma. Int JGynecol Pathol 20:341-345, 2001

factors for invasive epithelial ovarian cancer: Resultsfrom a Swedish case-control study. Am J Epidemiol156:363-373, 2002

31. Ji Q, Liu PI, Chen PK, et al: Follicle stimulatinghormone-induced growth promotion and gene ex-

pression profiles on ovarian surface epithelial cells.Int J Cancer 112:803-814, 2004

8. Halperin R, Zehavi S, Langer R, et al: Primary

19. Gwinn ML, Lee NC, Rhodes PH, et al: Preg-peritoneal serous papillary carcinoma: A new epide-miologic trend? A matched-case comparison withovarian serous papillary cancer. Int J Gynecol Cancer11:403-408, 2001

nancy, breast feeding, and oral contraceptives andthe risk of epithelial ovarian cancer. J Clin Epidemiol43:559-568, 1990

32. Schiffenbauer YS, Abramovitch R, Meir G, etal: Loss of ovarian function promotes angiogenesisin human ovarian carcinoma. Proc Natl Acad Sci U SA 94:13203-13208, 1997

20. Nasca PC, Greenwald P, Chorost S, et al: An9. Chen LM, Yamada SD, Fu YS, et al: Molecular epidemiologic case-control study of ovarian cancerand reproductive factors. Am J Epidemiol 119:705-713, 1984

similarities between primary peritoneal and primaryovarian carcinomas. Int J Gynecol Cancer 13:749-755, 2003

33. Wang J, Luo F, Lu JJ, et al: VEGF expressionand enhanced production by gonadotropins in ovar-

ian epithelial tumors. Int J Cancer 97:163-167, 200221. Fredrickson TN: Ovarian tumors of the hen.10. Lacy MQ, Hartmann LC, Keeney GL, et al:

Environ Health Perspect 73:35-51, 1987 34. Schiffenbauer YS, Meir G, Maoz M, et al:C-erbB-2 and p53 expression in fallopian tube carci-noma. Cancer 75:2891-2896, 1995

22. Land JA: Ovulation, ovulation induction andGonadotropin stimulation of MLS human epithelialovarian carcinoma cells augments cell adhesionmediated by CD44 and by alpha(v)-integrin. GynecolOncol 84:296-302, 2002

ovarian carcinoma. Baillieres Clin Obstet Gynaecol7:455-472, 1993

11. Pere H, Tapper J, Seppala M, et al: Genomicalterations in fallopian tube carcinoma: Comparisonto serous uterine and ovarian carcinomas revealssimilarity suggesting likeness in molecular patho-genesis. Cancer Res 58:4274-4276, 1998

23. Risch HA: Hormonal etiology of epithelialovarian cancer, with a hypothesis concerning therole of androgens and progesterone. J Natl CancerInst 90:1774-1786, 1998

35. Rosenberg L, Palmer JR, Zauber AG, et al: Acase-control study of oral contraceptive use and

invasive epithelial ovarian cancer. Am J Epidemiol139:654-661, 1994

12. Muto MG, Welch WR, Mok SC, et al: Evidence24. Schildkraut JM, Schwingl PJ, Bastos E, et al:for a multifocal origin of papillary serous carcinoma of

the peritoneum. Cancer Res 55:490-492, 1995 Epithelial ovarian cancer risk among women withpolycystic ovary syndrome. Obstet Gynecol 88:554-559, 1996

36. Gaspard UJ, Romus MA, Gillain D, et al:13. Kindelberger DW, Lee Y, Miron A, et al:

Plasma hormone levels in women receiving neworal contraceptives containing ethinyl estradiol plusIntraepithelial carcinoma of the fimbria and pelvic

www.jco.org 1003Information downloaded from jco.ascopubs.org and provided by UNIVERSITEITSBIBLIOTHEEK on March 25, 2010 from

131.174.244.21. Copyright © 2008 by the American Society of Clinical Oncology. All rights reserved.

Page 10: Early Events in the Pathogenesis of Epithelial Ovarian Cancer-printed - Copy

Landen, Birrer, and Sood

levonorgestrel or desogestrel. Contraception 27:577-590, 1983

cycle in response to DNA damage. Cancer Sci95:866-871, 2004

75. Ross JS, Yang F, Kallakury BV, et al: HER-2/neu oncogene amplification by uorescence in situ

hybridization in epithelial tumors of the ovary. Am JClin Pathol 111:311-316, 1999

37. Seeger H, Wallwiener D, Mueck AO: Is there 56. Boulton SJ: Cellular functions of the BRCAa protective role of progestogens on the prolifera-tion of human ovarian cancer cells in the presence ofgrowth factors? Eur J Gynaecol Oncol 27:139-141,2006

tumour-suppressor proteins. Biochem Soc Trans34:633-645, 2006 76. Singer G, Rebmann V, Chen YC, et al: HLA-

G57. Rosen EM, Fan S, Pestell RG, et al: BRCA1is a potential tumor marker in malignant ascites. ClinCancer Res 9:4460-4464, 2003

gene in breast cancer. J Cell Physiol 196:19-41,200338. McNatty KP, Smith DM, Makris A, et al: The 77. Ouellet V, Guyot MC, Le Page C, et al: Tissue

microenvironment of the human antral follicle: Inter-relationships among the steroid levels in antral uid,the population of granulosa cells, and the status ofthe oocyte in vivo and in vitro. J Clin EndocrinolMetab 49:851-860, 1979

58. Aarnio M, Sankila R, Pukkala E, et al: Cancer

array analysis of expression microarray candidatesidentifies markers associated with tumor grade andoutcome in serous epithelial ovarian cancer. Int JCancer 119:599-607, 2006

risk in mutation carriers of DNA-mismatch-repairgenes. Int J Cancer 81:214-218, 1999

59. Orsulic S, Li Y, Soslow RA, et al: Induction ofovarian cancer by defined multiple genetic changes

in a mouse model system. Cancer Cell 1:53-62,2002

78. Dodson MK, Hartmann LC, Cliby WA, et al:39. Edmondson RJ, Monaghan JM, Davies BR:Comparison of loss of heterozygosity patterns in

invasive low-grade and high-grade epithelial ovariancarcinomas. Cancer Res 53:4456-4460, 1993

The human ovarian surface epithelium is an andro-gen responsive tissue. Br J Cancer 86:879-885,2002

60. Connolly DC, Bao R, Nikitin AY, et al: Femalemice chimeric for expression of the simian virus 40

TAg under control of the MISIIR promoter developepithelial ovarian cancer. Cancer Res 63:1389-1397,2003

79. Meinhold-Heerlein I, Bauerschlag D, Hilpert F,40. Altinoz MA, Korkmaz R: NF-kappaB, macro-et al: Molecular and prognostic distinction betweenserous ovarian carcinomas of varying grade andmalignant potential. Oncogene 24:1053-1065, 2005

phage migration inhibitory factor and cyclooxygenase-inhibitions as likely mechanisms behind theacetaminophen- and NSAID-prevention of the ovar-ian cancer. Neoplasma 51:239-247, 2004

61. Dinulescu DM, Ince TA, Quade BJ, et al: Role80. Heinzelmann-Schwarz VA, Gardiner-Gardenof K-ras and Pten in the development of mouse

models of endometriosis and endometrioid ovariancancer. Nat Med 11:63-70, 2005

M, Henshall SM, et al: A distinct molecular profileassociated with mucinous epithelial ovarian cancer.Br J Cancer 94:904-913, 2006

41. Heller DS, Westhoff C, Gordon RE, et al: Therelationship between perineal cosmetic talc usageand ovarian talc particle burden. Am J Obstet Gy-necol 174:1507-1510, 1996

62. Shih Ie M, Kurman RJ: Ovarian tumorigene-81. Wamunyokoli FW, Bonome T, Lee JY, et al:sis: A proposed model based on morphological and

molecular genetic analysis. Am J Pathol 164:1511-1518, 2004

Expression profiling of mucinous tumors of theovary identifies genes of clinicopathologic impor-tance. Clin Cancer Res 12:690-700, 2006

42. Duggan BD, Dubeau L: Genetics and biologyof gynecologic cancer. Curr Opin Oncol 10:439-446,1998 63. Gershenson DM, Sun CC, Lu KH, et al:

Clinical 82. Suzuki M, Saito S, Saga Y, et al: Mutation of43. Donninger H, Bonome T, Radonovich M, et al: behavior of stage II-IV low-grade serous carcinomaof the ovary. Obstet Gynecol 108:361-368, 2006K-RAS protooncogene and loss of heterozygosity on

6q27 in serous and mucinous ovarian carcinomas.Cancer Genet Cytogenet 118:132-135, 2000

Whole genome expression profiling of advancestage papillary serous ovarian cancer reveals acti-vated pathways. Oncogene 23:8065-8077, 2004

64. Ozols RF, Bundy BN, Greer BE, et al: Phase IIItrial of carboplatin and paclitaxel compared with

cisplatin and paclitaxel in patients with optimallyresected stage III ovarian cancer: A GynecologicOncology Group study. J Clin Oncol 21:3194-3200,2003

44. Bild AH, Yao G, Chang JT, et al: Oncogenic

83. Obata K, Morland SJ, Watson RH, et al:pathway signatures in human cancers as a guide totargeted therapies. Nature 439:353-357, 2006

Frequent PTEN/MMAC mutations in endometrioidbut not serous or mucinous epithelial ovarian tu-mors. Cancer Res 58:2095-2097, 1998

45. Shridhar V, Lee J, Pandita A, et al: Geneticanalysis of early- versus late-stage ovarian tumors.

Cancer Res 61:5895-5904, 200165. Armstrong DK, Bundy B, Wenzel L, et al: 84. Hanahan D, Weinberg RA: The hallmarks of

Intraperitoneal cisplatin and paclitaxel in ovarian can-cer. N Engl J Med 354:34-43, 2006

cancer. Cell 100:57-70, 200046. Lancaster JM, Dressman HK, Clarke JP, et al: 85. Han L, Landen C, Trevino J, et al:

Anti-Identification of genes associated with ovarian can-cer metastasis using microarray expression analy-sis. Int J Gynecol Cancer 16:1733-1745, 2006

66. Malpica A, Deavers MT, Lu K, et al: Grading

angiogenic and anti-tumor effects of Src inhibition inovarian carcinoma. Cancer Res 66:8633-8639, 2006ovarian serous carcinoma using a two-tier system.

Am J Surg Pathol 28:496-504, 2004 86. Ishizawar R, Parsons SJ: C-Src and cooperat-47. Jazaeri AA, Yee CJ, Sotiriou C, et al: Gene

67. Crispens MA, Bodurka D, Deavers M, et al: ing partners in human cancer. Cancer Cell 6:209-214, 2004

expression profiles of BRCA1-linked, BRCA2-linked,and sporadic ovarian cancers. J Natl Cancer Inst94:990-1000, 2002

Response and survival in patients with progressiveor recurrent serous ovarian tumors of low malignantpotential. Obstet Gynecol 99:3-10, 2002

87. Silva CM: Role of STATs as downstreamsignal transducers in Src family kinase-mediatedtumorigenesis. Oncogene 23:8017-8023, 2004

48. Cass I, Baldwin RL, Varkey T, et al: Improved

68. Mok SC, Bell DA, Knapp RC, et al: Mutation ofsurvival in women with BRCA-associated ovarian

carcinoma. Cancer 97:2187-2195, 2003K-ras protooncogene in human ovarian epithelialtumors of borderline malignancy. Cancer Res 53:1489-1492, 1993

88. Wiener JR, Windham TC, Estrella VC, et al:Activated SRC protein tyrosine kinase is overex-pressed in late-stage human ovarian cancers. Gy-necol Oncol 88:73-79, 2003

49. Gotlieb WH, Chetrit A, Menczer J, et al:Demographic and genetic characteristics of patientswith borderline ovarian tumors as compared to earlystage invasive ovarian cancer. Gynecol Oncol 97:780-783, 2005

69. Teneriello MG, Ebina M, Linnoila RI, et al: p53and Ki-ras gene mutations in epithelial ovarian neo-

plasms. Cancer Res 53:3103-3108, 199389. Pengetnze Y, Steed M, Roby KF, et al: Src

tyrosine kinase promotes survival and resistance tochemotherapeutics in a mouse ovarian cancer cellline. Biochem Biophys Res Commun 309:377-383,2003

70. Singer G, Oldt R, 3rd, Cohen Y, et al: Muta-50. Merajver SD, Pham TM, Caduff RF, et al: tions in BRAF and KRAS characterize the develop-

ment of low-grade ovarian serous carcinoma. J NatlCancer Inst 95:484-486, 2003

Somatic mutations in the BRCA1 gene in sporadicovarian tumours. Nat Genet 9:439-443, 1995 90. Bartlett JM, Langdon SP, Simpson BJ, et al:51. Baldwin RL, Nemeth E, Tran H, et al:

BRCA171. Kohler MF, Marks JR, Wiseman RW, et al:

The prognostic value of epidermal growth factorreceptor mRNA expression in primary ovarian can-cer. Br J Cancer 73:301-306, 1996

promoter region hypermethylation in ovarian carci-noma: A population-based study. Cancer Res 60:5329-5333, 2000

Spectrum of mutation and frequency of allelic deletionof the p53 gene in ovarian cancer. J Natl Cancer Inst85:1513-1519, 1993 91. Leary JA, Edwards BG, Houghton CR, et al:52. Esteller M, Silva JM, Dominguez G, et al: 72. Kupryjanczyk J, Thor AD, Beauchamp R, et al:

Amplification of HER-2/neu oncogene in humanovarian cancer. Int J Gynecol Cancer 2:291-294,1992

Promoter hypermethylation and BRCA1 inactivationin sporadic breast and ovarian tumors. J Natl CancerInst 92:564-569, 2000

p53 gene mutations and protein accumulation inhuman ovarian cancer. Proc Natl Acad SciUSA90:4961-4965, 1993 92. Sui L, Dong Y, Ohno M, et al: Implication

of53. Hilton JL, Geisler JP, Rathe JA, et al: Inacti-

73. Skilling JS, Sood A, Niemann T, et al: Anmalignancy and prognosis of p27(kip1), Cyclin E, andCdk2 expression in epithelial ovarian tumors. Gy-necol Oncol 83:56-63, 2001

vation of BRCA1 and BRCA2 in ovarian cancer.J Natl Cancer Inst 94:1396-1406, 2002

abundance of p53 null mutations in ovarian carci-noma. Oncogene 13:117-123, 1996

54. Venkitaraman AR: Cancer susceptibility and 74. Cheng JQ, Godwin AK, Bellacosa A, et al:the functions of BRCA1 and BRCA2. Cell 108:171-182, 2002

AKT2, a putative oncogene encoding a member of asubfamily of protein-serine/threonine kinases, is am-plified in human ovarian carcinomas. Proc Natl AcadSciUSA89:9267-9271, 1992

93. Dhar KK, Branigan K, Parkes J, et al: Expres-sion and subcellular localization of cyclin D1 protein

in epithelial ovarian tumour cells. Br J Cancer 81:1174-1181, 1999

55. Yoshida K, Miki Y: Role of BRCA1 and BRCA2as regulators of DNA repair, transcription, and cell

1004 JO URN AL OF CL I N ICA L O N COL O GYInformation downloaded from jco.ascopubs.org and provided by UNIVERSITEITSBIBLIOTHEEK on March 25, 2010 from

131.174.244.21. Copyright © 2008 by the American Society of Clinical Oncology. All rights reserved.

Page 11: Early Events in the Pathogenesis of Epithelial Ovarian Cancer-printed - Copy

Carcinogenesis of Ovarian Cancer

94. Barrette BA, Srivatsa PJ, Cliby WA, et al: 111. Folkman J: The role of angiogenesis in tumor 129. Kamat AA, Fletcher M, Gruman LM, et al:Overexpression of p34cdc2 protein kinase in epithe-lial ovarian carcinoma. Mayo Clin Proc 72:925-929,1997

growth. Semin Cancer Biol 3:65-71, 1992The clinical relevance of stromal matrix metallopro-teinase expression in ovarian cancer. Clin CancerRes 12:1707-1714, 2006

112. Frumovitz M, Sood A: Vascular EndothelialGrowth Factor (VEGF) Pathway as a TherapeuticTarget in Gynecologic Malignancies. Gynecol Oncol104:768-778, 2007

95. Bao R, Connolly DC, Murphy M, et al: Activa-

130. Huang S, Van Arsdall M, Tedjarati S, et al:tion of cancer-specific gene expression by the sur-

vivin promoter. J Natl Cancer Inst 94:522-528, 2002Contributions of stromal metalloproteinase-9 to an-giogenesis and growth of human ovarian carcinomain mice. J Natl Cancer Inst 94:1134-1142, 2002

113. Connolly DT, Heuvelman DM, Nelson R, et96. Yu Y, Xu F, Peng H, et al: NOEY2 (ARHI), an

al: Tumor vascular permeability factor stimulatesendothelial cell growth and angiogenesis. J ClinInvest 84:1470-1478, 1989

imprinted putative tumor suppressor gene in ovarianand breast carcinomas. Proc Natl Acad Sci U S A96:214-219, 1999

131. Thaker PH, Han LY, Kamat AA, et al: Chronicstress promotes tumor growth and angiogenesis in

a mouse model of ovarian carcinoma. Nat Med12:939-944, 2006

114. Senger DR, Galli SJ, Dvorak AM, et al: Tumor97. Michalovitz D, Halevy O, Oren M: p53 muta- cells secrete a vascular permeability factor that

promotes accumulation of ascites uid. Science219:983-985, 1983

tions: Gains or losses? J Cell Biochem 45:22-29,1991

132. Sood AK, Bhatty R, Kamat AA, et al: Stresshormone-mediated invasion of ovarian cancer cells.Clin Cancer Res 12:369-375, 2006

98. Skilling JS, Squatrito RC, Connor JP, et al: p53

115. Yoneda J, Kuniyasu H, Crispens MA, et al:gene mutation analysis and antisense-mediatedgrowth inhibition of human ovarian carcinoma celllines. Gynecol Oncol 60:72-80, 1996

Expression of angiogenesis-related genes and pro-gression of human ovarian carcinomas in nude mice.J Natl Cancer Inst 90:447-454, 1998

133. Antoni MH, Lutgendorf SK, Cole SW, et al:The in uence of bio-behavioral factors on tumorbiology: Pathways and mechanisms. Nat Rev Can-cer 6:240-248, 2006

99. Marks JR, Davidoff AM, Kerns BJ, et al: 116. Paley PJ, Staskus KA, Gebhard K, et al:Overexpression and mutation of p53 in epithelialovarian cancer. Cancer Res 51:2979-2984, 1991

Vascular endothelial growth factor expression inearly stage ovarian carcinoma. Cancer 80:98-106,1997

134. Ben-Hur H, Gurevich P, Huszar M, et al:Apoptosis and apoptosis-related proteins (Fas, Fasligand, Blc-2, p53) in lymphoid elements of humanovarian tumors. Eur J Gynaecol Oncol 21:141-145,2000

100. Kerner R, Sabo E, Gershoni-Baruch R, et al:Expression of cell cycle regulatory proteins in ova-ries prophylactically removed from Jewish Ash-kenazi BRCA1 and BRCA2 mutation carriers:Correlation with histopathology. Gynecol Oncol 99:367-375, 2005

117. Cooper BC, Ritchie JM, Broghammer CL, etal: Preoperative serum vascular endothelial growthfactor levels: Significance in ovarian cancer. ClinCancer Res 8:3193-3197, 2002

135. Paul P, Rouas-Freiss N, Khalil-Daher I, et al:HLA-G expression in melanoma: A way for tumor

cells to escape from immunosurveillance. Proc NatlAcad SciUSA95:4510-4515, 1998

118. Xu L, Fidler IJ: Interleukin 8: an autocrine101. Hu L, Hofmann J, Lu Y, et al: Inhibition

ofgrowth factor for human ovarian cancer. Oncol Res12:97-106, 2000phosphatidylinositol 3 -kinase increases efficacy of

paclitaxel in in vitro and in vivo ovarian cancermodels. Cancer Res 62:1087-1092, 2002

136. Bukovsky A: Immune system involvement in119. Lokshin AE, Winans M, Landsittel D, et al:the regulation of ovarian function and augmentationof cancer. Microsc Res Tech 69:482-500, 2006

Circulating IL-8 and anti-IL-8 autoantibody in patientswith ovarian cancer. Gynecol Oncol 102:244-251,2006

102. Deregowski V, Delhalle S, Benoit V, et al:Identification of cytokine-induced nuclear factor-kappaB target genes in ovarian and breast cancercells. Biochem Pharmacol 64:873-881, 2002

137. Nash MA, Ferrandina G, Gordinier M, et al:120. Davidson B, Goldberg I, Reich R, et al:

The role of cytokines in both the normal and malig-nant ovary. Endocr Relat Cancer 6:93-107, 1999AlphaV- and beta1-integrin subunits are commonly

expressed in malignant effusions from ovarian car-cinoma patients. Gynecol Oncol 90:248-257, 2003

103. Huang S, Robinson JB, Deguzman A, et al: 138. Zhang L, Conejo-Garcia JR, Katsaros D, et al:Blockade of nuclear factor-kappaB signaling inhibits

angiogenesis and tumorigenicity of human ovariancancer cells by suppressing expression of vascularendothelial growth factor and interleukin 8. CancerRes 60:5334-5339, 2000

Intratumoral T cells, recurrence, and survival in epi-thelial ovarian cancer. N Engl J Med 348:203-213,2003

121. Thaker PH, Deavers M, Celestino J, et al:EphA2 expression is associated with aggressivefeatures in ovarian carcinoma. Clin Cancer Res 10:5145-5150, 2004

139. Coukos G, Conejo-Garcia JR, Roden RB, etal: Immunotherapy for gynaecological malignancies.Expert Opin Biol Ther 5:1193-1210, 2005

104. Landen CN, Klingelhutz A, Coffin JE, et al:

122. Landen CN, Jr., Chavez-Reyes A, Bucana C,Genomic instability is associated with lack of telom-erase activation in ovarian cancer. Cancer Biol Ther3:1250-1253, 2004

et al: Therapeutic EphA2 Gene Targeting In vivoUsing Neutral Liposomal Small Interfering RNA De-livery. Cancer Res 65:6910-6918, 2005

140. Fidler IJ: The pathogenesis of cancer metas-tasis: The ‘seed and soil’ hypothesis revisited. NatRev Cancer 3:453-458, 2003105. Poremba C, Heine B, Diallo R, et al: Telom- 123. Landen CN, Jr., Lu C, Han LY, et al:

Efficacy141. Emerich J, Konefka T, Dudziak M, et al: [The

erase as a prognostic marker in breast cancer:High-throughput tissue microarray analysis ofhTERT and hTR. J Pathol 198:181-189, 2002

and antivascular effects of EphA2 reduction with anagonistic antibody in ovarian cancer. J Natl CancerInst 98:1558-1570, 2006

value of peritoneal cytology in the staging of ovariancancer]. Ginekol Pol 68:74-77, 1997

142. Hood JD, Cheresh DA: Role of integrins in106. Yang G, Rosen DG, Mercado-Uribe I, et al: 124. Jung YD, Ahmad SA, Akagi Y, et al: Role of

cell invasion and migration. Nat Rev Cancer 2:91-100, 2002

Knockdown of p53 combined with expression of thecatalytic subunit of telomerase is sufficient to im-mortalize primary human ovarian surface epithelialcells. Carcinogenesis 28:174-182, 2007

the tumor microenvironment in mediating responseto anti-angiogenic therapy. Cancer Metastasis Rev19:147-157, 2000

143. Carreiras F, Rigot V, Cruet S, et al: Migrationproperties of the human ovarian adenocarcinoma

cell line IGROV1: importance of alpha(v) beta3 inte-grins and vitronectin. Int J Cancer 80:285-294, 1999

125. Lu C, Bonome T, Li Y, et al: Gene alterations107. Zhou C, Liu J: Inhibition of human

telomer-identified by expression profiling in tumor-associated endothelial cells from invasive ovariancarcinoma. Cancer Res 67:1757-1768, 2007

ase reverse transcriptase gene expression byBRCA1 in human ovarian cancer cells. BiochemBiophys Res Commun 303:130-136, 2003

144. Sundfeldt K: Cell-cell adhesion in the normalovary and ovarian tumors of epithelial origin; anexception to the rule. Mol Cell Endocrinol 202:89-96,2003

126. Belotti D, Paganoni P, Manenti L, et al:108. Sorlie T, Perou CM, Tibshirani R, et al: Gene

Matrix metalloproteinases (MMP9 and MMP2) in-duce the release of vascular endothelial growthfactor (VEGF) by ovarian carcinoma cells: Implica-tions for ascites formation. Cancer Res 63:5224-5229, 2003

expression patterns of breast carcinomas distin-guish tumor subclasses with clinical implications.Proc Natl Acad SciUSA98:10869-10874, 2001

145. Halder J, Kamat AA, Landen CN, Jr., et al:Focal adhesion kinase targeting using in vivo short

interfering RNA delivery in neutral liposomes forovarian carcinoma therapy. Clin Cancer Res 12:4916-4924, 2006

109. Wang E, Ngalame Y, Panelli MC, et al: Peri-toneal and subperitoneal stroma may facilitate re-gional spread of ovarian cancer. Clin Cancer Res11:113-122, 2005

127. Naylor MS, Stamp GW, Davies BD, et al:Expression and activity of MMPS and their regula-tors in ovarian cancer. Int J Cancer 58:50-56, 1994 146. Sundfeldt K, Piontkewitz Y, Ivarsson K, et al:

110. Folkman J: Angiogenesis in cancer, vascular, 128. Herrera CA, Xu L, Bucana CD, et al: Expres-

E-cadherin expression in human epithelial ovariancancer and normal ovary. Int J Cancer 74:275-280,1997

rheumatoid and other disease. Nat Med 1:27-31,1995

sion of metastasis-related genes in human epithelialovarian tumors. Int J Oncol 20:5-13, 2002

¦ ¦¦

www.jco.org 1005Information downloaded from jco.ascopubs.org and provided by UNIVERSITEITSBIBLIOTHEEK on March 25, 2010 from

131.174.244.21. Copyright © 2008 by the American Society of Clinical Oncology. All rights reserved.