You are on page 1of 14

u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132

available at www.sciencedirect.com

journal homepage: www.updateoncancer.com

Development of novel agents for ovarian cancer

B.T. Hennessy ∗ , M. Markman


Departments of Gynecology Medical Oncology and Systems Biology, The University of Texas MD Anderson Cancer Center,
1515 Holcombe Boulevard, Houston, TX 77030, USA

a r t i c l e i n f o a b s t r a c t

Keywords: Epithelial ovarian cancer, the fourth leading cause of cancer deaths in American women,
Ovarian is a devastating disease. Although the combined intraperitoneal and intravenous adminis-
Cancer tration of first-line chemotherapy does produce some improvement in patient outcomes
Molecular in comparison with intravenous administration alone, the results of completed clinical
Genomic trials suggest that a plateau has been reached in the benefits that can be achieved with
Chemotherapy cytotoxic chemotherapy alone in ovarian cancer. Epithelial ovarian cancer is currently clas-
Novel sified by surgical and histologic criteria. However, the predictive value of this classification
Targeted is limited. Moreover, the complexity of the different subtypes of ovarian cancer presents
Therapy challenges to our understanding of the origin and pathogenesis of this disease. The sequen-
tial acquisition of molecular anomalies as occurs in colorectal carcinogenesis has not been
described for ovarian cancer. However, increasing evidence indicates that multiple genetic
and epigenetic events contribute to epithelial ovarian carcinogenesis. These events include
aberrations in the expression of patterning genes, genomic instability, and resultant chro-
mosomal changes, and may be triggered by excessive exposure of surface epithelial cells
to autocrine/paracrine stimulation by sex steroids and other growth factors. As the dis-
ease progresses, activation of kinase pathways and continued abnormal autocrine/paracrine
stimulation contribute to genomic instability and chemoresistance but also identify novel
potential targets for therapeutic intervention. Although the list of genomic and other aber-
rations in ovarian cancer is daunting, a systems approach and the integration of therapies
targeting multiple component ‘driver’ genes of important genetic aberrations has potential
in treatment and for potentiation of cytotoxic chemotherapy efficacy.
© 2009 Elsevier Ltd. All rights reserved.

1. Introduction ment therapy, a family history of ovarian and/or breast cancer,


and nulliparity, whereas the oral contraceptive pill (OCP)
Epithelial ovarian cancer is the fourth leading cause of can- decreases risk. Consistent with an association with ovula-
cer deaths in American women [1]. Although referred to as tion and endocrine factors, 40–60% of ovarian cancers express
epithelial ovarian cancer, neoplasms with similar morphology estrogen receptor (ER)␣ , although only a minor proportion of
and behavior can also arise from endometriosis, endosalp- patients, usually those possessing low-grade tumors, respond
ingiosis, the fallopian tube and peritoneum. The disease is to endocrine manipulation [2]. Loss of ER␤ and progesterone
currently thought to arise from the ovarian surface epithelium receptor expression may be important events leading to ovar-
(OSE). Risk factors include increasing age, estrogen replace- ian carcinogenesis.


Corresponding author at: Departments of Gynecology Medical Oncology and Systems Biology, CPB6.3444, The University of Texas MD
Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA. Tel.: +1 713 563 1792; fax: +1 713 745 1541.
E-mail address: bhennessy@mdanderson.org (B.T. Hennessy).
1872-115X/$ – see front matter © 2009 Elsevier Ltd. All rights reserved.
doi:10.1016/j.uct.2009.02.001
120 u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132

Table 1 – Conventional staging of ovarian cancer.


Stage Description

1 Tumor limited to ovary or ovaries


A One ovary involved
B Both ovaries involved
C Tumor limited to ovary or ovaries with ascites, positive peritoneal washings, surface involvement, or rupture present
2 Ovarian tumor with pelvic extension
3 Tumor involving the upper abdomen or lymph nodes
4 Distant organ involvement (includes a malignant pleural effusion and parenchymal involvement of liver or spleen)

Three interrelated theories have been proposed to explain cancer associated with talc/asbestos exposure, endometriosis,
the epidemiologic data associated with susceptibility to ovar- pelvic inflammatory disease, and mumps infection [4]. Since
ian cancer [3]. The incessant ovulation hypothesis postulates the inflammation-like setting in which ovulation occurs is
that repetitive wounding and cell proliferation in postovu- cyclooxygenase 2 (COX2)-dependent, this provides support for
latory repair of the OSE result in a stepwise accumulation exploration of the chemopreventive and therapeutic potential
of genomic abnormalities. Ovarian epithelial inclusion cysts of COX2 inhibitors in ovarian cancer. However, while chemo-
occur as a result and may increase the risk of carcino- prevention and screening remain extremely attractive, the low
genesis by trapping cells in an environment of aberrant prevalence of ovarian cancer in the general population reduces
autocrine/paracrine stimulation by growth factors including enthusiasm for such approaches. Most current efforts that
hormones, phospholipids, and vascular endothelial growth attempt to improve patient outcomes thus focus on therapy
factor (VEGF), which activate intracellular processes such as for women with established ovarian cancer.
kinase signaling. The gonadotropin theory postulates that Table 1 shows epithelial ovarian cancer staging and Fig. 1
surges of pituitary gonadotrophins at ovulation and persis- represents current treatment guidelines. The majority of
tent high levels following menopause stimulate OSE cells, patients present with advanced (stage III/IV) high-grade dis-
resulting in accumulation of genetic changes and carcinogen- ease and, as a result, most of these patients ultimately relapse
esis. The third theory suggests a role for inflammation and and die from ovarian cancer within 5 years of diagnosis despite
changes in redox potential in the setting of ovulation and an improved median survival associated with standard first-
OSE repair and may account for the increased risk of ovarian line therapy (paclitaxel or docetaxel/carboplatin combination

Fig. 1 – Simplified schema of current NCCN treatment guidelines for epithelial ovarian cancer. Cytoreductive surgery
generally consists of a total abdominal hysterectomy, salpingo-oophorectomy, and omentectomy. Unilateral
salpingo-oophorectomy for those desiring fertility preservation may be considered in patients with stage 1A or 1C disease
of any grade (*). Generally, adjuvant taxane/carboplatin-based chemotherapy is recommended in patients following
debulking of grade 3 stage 1A/B tumors or of any stage IC-IV tumors. With grade 2 stage 1A/B disease, observation may also
be an appropriate option (**). Although data regarding interval cytoreductive surgery during primary chemotherapy are
somewhat conflicting, most studies agree in terms of the benefit of an extensive attempt at surgical cytoreduction during
primary therapy. Several studies have also shown a survival advantage for intraperitoneal chemotherapy in patients with
advanced (stage III) ovarian cancer (***) although there has thus far been reticence to move this into standard of care.
u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132 121

Table 2 – Current concepts regarding the origins and molecular pathology of ovarian cancer. PIK3CA is the gene at
chromosome 3q26 which specifically encodes the p110␣ subunit of phosphatidylinositol-3-kinase (PI3K) [25].
Histology Precursor Molecular features

Low-grade serous carcinoma Cystadenoma-borderline Mutations in K-ras and/or b-raf


tumor-carcinoma sequence
High-grade serous carcinoma ‘De novo’ in epithelial inclusion cysts p53 mutation and BRCA1 dysfunction
(usually promoter methylation)
PIK3CA amplification (25–40%)
Low-grade endometrioid Endometriosis and endometrial-like Mutations in CTNNB1 (␤-catenin gene) and
carcinoma hyperplasiaa PTEN with microsatellite instability
High-grade endometrioid Epithelial inclusion glands/cysts p53 mutation and BRCA1 dysfunction
carcinoma (usually promoter methylation)
PIK3CA mutation
Mucinous carcinoma Cystadenoma-borderline Mutations in K-ras; ? p53 mutation
tumor-carcinoma sequence associated with transition from borderline
tumor to carcinoma
Clear cell carcinoma ?endometriosis ?PTEN mutation/loss of heterozygosity
PIK3CA mutation

The statement following the ? is controversial and not conclusively shown to date.
a
Endometriosis and adjacent low-grade endometrioid carcinoma share common genetic events such as loss of heterozygosity at the same loci
involving the same allele (e.g. PTEN). In contrast, high-grade and poorly differentiated endometrioid carcinomas are similar to high-grade
serous carcinomas.

chemotherapy and surgical debulking) [5]. Although the ovarian cancer is a heterogeneous disease and its clinical and
combined intraperitoneal and intravenous administration cellular biology have implications for the molecular pathogen-
of first-line chemotherapy does produce some improve- esis of specific tumors (Table 2) and thus for novel potential
ment in patient outcomes in comparison with intravenous targets and therapies that will likely prove useful in patient
administration alone at the expense of increased toxicity therapy. These novel therapies may have their greatest impact
[6], the incorporation of additional cytotoxic chemotherapy on patient outcomes when used in combination with cytotoxic
drugs into first-line treatment has not resulted in signifi- chemotherapy drugs such as paclitaxel and carboplatin.
cant improvements in patient outcomes in comparison with
paclitaxel/carboplatin therapy alone [7]. This suggests that a
plateau has been reached in the benefits that can be achieved 2. Pathogenesis of ovarian cancer
with cytotoxic chemotherapy alone in ovarian cancer. Thus,
although novel chemotherapy drugs (e.g. epothilones and hali- 2.1. Clinical and cellular biology
chondrins, both novel microtubule inhibitors and karenitecin,
a camptothecin analogue) and formulations (e.g. nanopar- The cellular and clinical biology of ovarian cancers have impli-
ticle albumin-bound (nab)-paclitaxel (Abraxane® ), liposomal cations for patient outcomes after current standard therapies
lurtotecan (OSI-211) and antibody-drug conjugates such as [12–17]. Epithelial ovarian neoplasms are classified based on
cBR96-doxorubicin that delivers doxorubicin to tumor tissues their histologic appearance into serous, mucinous, endometri-
expressing the Lewis-y (Ley ) antigen) are being evaluated (Clin- oid, and, less commonly, clear cell, transitional, squamous,
icalTrials.gov) [8], most enthusiasm and efforts to improve mixed, and undifferentiated types. The serous subtype is
treatment are moving towards novel targeted therapies. most common. In a series of more than 8000 cases pub-
Therefore, the bulk of this review will focus on the current and lished between 1982 and 1986, mucinous and endometrioid
future development of novel targeted therapeutic approaches carcinomas were found to be associated with a relatively
in ovarian cancer. To date, novel inhibitors such as epidermal favorable prognosis, serous carcinomas less so, and undif-
growth factor receptor (EGFR) family inhibitors or inhibitors of ferentiated carcinoma was the most aggressive subtype of
Kit have not had a major impact on the treatment of women ovarian carcinoma [12,18]. The data regarding clear cell (or
with ovarian cancer likely because EGFR and Kit gene aber- “mesonephroid”) carcinoma appeared somewhat conflicting,
rations, which are associated with responsiveness to these with about half of reviewed publications placing it in a rel-
targeted therapies in other tumor types, are rare in epithe- atively favorable category and the other half finding it to be
lial ovarian cancer [9]. More enthusiasm surrounds the use associated with an unfavorable prognosis [13]. These discrep-
of inhibitors of VEGF signaling such as the monoclonal anti- ancies may be due in part to the fact that, although clear cell
body bevacizumab (Avastin® ); indeed, bevacizumab is now carcinoma is inherently aggressive, it is frequently detected
being assessed in randomized phase III trials in women with at Stage I. The histopathologic grade of epithelial ovarian
newly diagnosed ovarian cancer [10,11]. To advance the devel- carcinoma is also important to tumor behavior and has con-
opment of novel agents for the treatment of ovarian cancer, sistently been found to be of prognostic significance [12,13]. In
an in depth and improved understanding of the molecular virtually all published series’, histopathologic subtype had a
biology of the disease is required so that rational molecu- less significant impact on prognosis than either clinicopatho-
lar targets can be identified and exploited. Further, epithelial logic stage or histopathologic grade [12,13]. Emerging data now
122 u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132

suggest that low- and high-grade carcinomas of the ovary con on chromosome 17q, although multiple other nearby
develop along different pathogenetic pathways, particularly genes are frequently co-amplified with HER2-neu. Whether
with serous carcinomas, and that low-grade carcinomas and these co-amplified genes are “innocent” passengers or act
tumors of low malignant potential may emerge along a sim- cooperatively with HER2-neu remains unknown. Theoreti-
ilar developmental sequence of molecular changes [19–22]. cally, genes selected for mutation as well as ‘key drivers’ of
The molecular heterogeneity associated with ovarian can- chromosomal copy number alterations are most important to
cers of different grades and subtypes will be discussed in the carcinogenesis process and represent the best potential
detail below as it has profound implications for the design targets for novel therapies because of ‘oncogene addiction’
of novel therapies. For example, many experts feel that grade [24]. Study of the transcriptional and proteomic effects of
is an unreliable criterion on which to base a therapy decision aberrant genes in ovarian cancer is thus essential to deter-
presently but this is likely to change as novel therapies that mining their importance to the carcinogenesis process and to
target specific molecular anomalies are introduced. Currently, exploring the antitumor efficacy and proteomic effects of ther-
the mitogen activated protein kinase (MAPK) kinase (MEK) apies designed to specifically target the protein products of
inhibitor AZD6244 is being evaluated specifically in the treat- these aberrant genes. Further, as there are too many candidate
ment of women with low-grade ovarian cancer by the United genes for systematic analysis of all potential drivers at areas of
States (U.S.) National Cancer Institute (NCI-ClinicalTrials.gov) genomic abnormality, maintaining focus specifically on those
(see below). genomic changes that correlate with patient outcomes is an
imperfect filter but one that nonetheless allows us to focus
2.2. Molecular biology on genes that are most likely to provide effective treatment
targets.
An in-depth and improved understanding of the molecular
biology of epithelial ovarian cancer is likely to efficiently facil- 2.3. Mutations
itate the identification of targets and the development of novel
agents for ovarian cancer treatment. Current understand- Genomic mutations are felt to play a frequent and key
ing of the molecular complexity of ovarian cancer suggests role in the pathogenesis of multiple forms of cancer. High
that novel treatment strategies likely need to focus on more prevalence mutations (>5%) have been found in only a lim-
than single molecules or targets. Like many solid tumors, ited number of genes in epithelial ovarian cancer, occur
epithelial ovarian cancer has a high degree of chromoso- in most cases in a histologic subtype- and grade-specific
mal instability, and both total and regional instability are fashion (Table 2) and identify genes whose functional per-
associated with tumor grade and altered patient outcomes turbation likely plays a key role in ovarian carcinogenesis.
[23]. Ovarian carcinogenesis is thought to evolve through These genes include p53, CTNNB1, PTEN (all inactivated), K-
the multistep acquisition of genomic and epigenetic aberra- ras and PIK3CA (the gene that encodes the p110␣ protein
tions that initially deregulate normal cell growth control and subunit of the phosphatidylinositol-3-kinase (PI3K) protein)
subsequently result in autonomous proliferation and other (both activated) [25–29]. Activating AKT1 mutations also occur
“hallmark” features of malignancy [24]. Although there are but at a frequency of under 5% [28]. Although p53 is a tumor
only a few “hallmarks”, many genomic aberrations act in con- suppressor gene, due to the dominant negative activity of
cert to contribute to the carcinogenesis process. Indeed, it is mutant p53 protein, p53 function can be lost with a single
known that aberrations in single oncogenes frequently result genetic event. Further, germline mutations of BRCA1, BRCA2,
in senescence or oncogene-induced cell death. Combined with and the hereditary non-polyposis coli (Lynch syndrome II)
the evidence for multiple different non-overlapping defects in genes, all associated with DNA repair processes, are asso-
cancer cells, this suggests a major degree of complexity. An ciated with a markedly increased risk of epithelial ovarian
improved understanding of the underlying genomic aberra- cancer and account for 13.2% of all ovarian cancers [30]. Hered-
tions that drive epithelial ovarian cancer and how they disrupt itary BRCA1/2-related ovarian cancers tend to occur at an
protein function leading to tumorigenesis is needed to design earlier age than sporadic tumors, possibly by predisposing to
optimal therapy strategies. Thus, a comprehensive character- genomic instability, and as compared with sporadic ovarian
ization of genomic anomalies in epithelial ovarian cancer is tumors are more commonly high-grade serous tumors with
a useful first step in studies that attempt to advance under- p53 dysfunction [31]. Although BRCA1 and BRCA2 are rarely
standing of ovarian carcinogenesis and thereby to discover mutated by sporadic mutation in ovarian cancers, these genes
promising new targets and agents for therapy. can also be silenced by methylation. Indeed, transcriptional
The selection of epithelial ovarian cancer as one of three profiles demonstrate similar changes in sporadic and BRCA1-
tumor types for extensive characterization as part of the associated tumors suggesting that BRCA1 function may be
cancer genome atlas (TCGA) project offers an incredible oppor- compromised in most ovarian cancers. As poly (ADP-ribose)
tunity to extend our understanding of this disease. Although polymerase 1 (PARP) inhibitors may be selectively active in
mutations frequently affect only a limited number of genes cancer patients with aberrations in BRCA1 and BRCA2 (see
in epithelial ovarian cancer, chromosomal copy number aber- below), this likely has clinical significance.
rations typically involve large numbers of genes and further Novel high-throughput approaches to sequencing of spe-
investigative techniques are required to determine a more cific functional groups of genes (e.g. the kinome) have not
limited list of those target genes that act as ‘key drivers’ of thus far revealed other commonly mutated genes in ovarian
carcinogenesis. For example, in breast cancer, the HER2-neu cancer but have probed only limited parts of the genome [32].
oncogene is believed to be a ‘key driver’ of the HER2-neu ampli- These approaches have, however, identified a large number of
u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132 123

“rare” events that may act coordinately during tumorigenesis. critical markers and potential treatment targets. However, it
Thus, high-throughput sequencing [33] by the Sanger Cen- is not clear if each genomic aberration harbors a single “key
ter (www.sanger.ac.uk) and as proposed by TCGA in epithelial driver” or whether multiple genes within an aberrant region
ovarian cancer will likely identify a large number of low may contribute to “cooperative carcinogenesis”.
frequency mutations. A systems biology approach with path- Novel high throughput technologies such as comparative
way or network analyses may demonstrate that these “rare” genomic hybridization are capable of profiling copy number
mutations result in coordinate aberrations in common critical changes across the entire genome in cancer [40–43] and have
processes. become a conventional first step in the identification of poten-
It is possible by ectopically manipulating genes in the ovar- tially important markers and targets in epithelial ovarian
ian surface epithelial (OSE) cells of mice to induce a very cancer. Subsequent approaches to distinguishing candidate
poorly differentiated form of ovarian cancer by combining p53 ‘drivers’ from other less important co-amplified or co-deleted
loss with activation/overexpression of two of three oncogenes genes in areas of chromosomal copy number change have
(myc, K-ras, and AKT1) [34,35]. In mice that are deficient for focused on finding those aberrations that induce significant
both p53 and BRCA1, myc alone is sufficient to induce trans- changes in both RNA and protein levels and function and
formation and resultant cancers have increased sensitivity to result in alterations in the phenotypic behavior of manipu-
the DNA damaging agent cisplatin. Although the relevance of lated cells in vitro and/or in significant changes to patient
these models to human ovarian cancer remains disputable, outcome [42]. These ‘drivers’ are susceptible to exploitation
they do have the potential to allow exploration for the role through small molecule inhibition, antibody targeting, small
of specific oncogenes in ovarian carcinogenesis and to allow interfering (si)-RNA technology, or transfection and overex-
potential evaluation of targeted therapies. Further, OSE cells pression in the case of deleted genes.
provide potent reagents to determine mechanisms underlying Since inhibition of protein function is generally believed to
immortalization and transformation [36–38]. be more achievable than restoration of function, a perception
Mutations identify logical potential markers and tar- reinforced by the clinical failure in ovarian cancer to date to
gets to facilitate better treatment [24]. However, successful exploit p53 loss and by the success in breast cancer of the
targeting of the consequences of mutations in human ovar- anti-HER2 monoclonal antibody trastuzumab, most studies
ian cancer treatment has not yet been achieved. Despite focus on areas of chromosomal gain (amplicons) rather than
promising preclinical and clinical data leading to an interna- on areas of deletion for identification of novel potential ther-
tional randomized phase II/III trial of p53 gene-therapy using apy targets. Candidate ‘drivers’ at areas of copy number gain
replication-deficient adenoviral vectors in combination with that have been identified to date in ovarian cancer include the
first-line chemotherapy for epithelial ovarian cancer, no ben- small G protein RAB25 at 1q22, ecotropic viral integration site-
efit was shown [39]. However, there were multiple potential 1 (EVI1), protein kinase C iota (PKCi), SnoN, BCL6, the initiation
problems including some that may have affected the success- factor eIF-5A2 and PIK3CA at 3q26.2, c-myc and PVT1 at 8q24.2,
ful intratumoral delivery of p53. However, a number of novel remodeling and spacing factor 1 (rsf-1, HBXAP) and PAK1 at
therapeutics targeting aberrations relevant to selected ovarian 11q13, HER2-neu at 17q12, AKT2 at 19q13.2 and ZNF217, BTAK
cancer histiotypes such as mutations in PIK3CA are currently (Aurora Kinase), the tyrosine kinase BRK and EEF1A2 at 20q13.2
entering clinical trials (e.g. the AKT inhibitor perifosine (Keryx [41,44–51]. Some of these abnormalities are targetable with
Pharmaceuticals, New York, NY) and the PI3K inhibitor SF1126 novel agents that are currently undergoing preclinical or early
(Semafore Pharmaceuticals, Indianapolis, IN)). The successful clinical assessment.
exploitation of genomic mutations for ovarian cancer therapy In addition to mutations and DNA copy number changes,
will require not only effective delivery of the agent to tumor rearrangements, epigenetic changes and imprinting also
cells but also a comprehensive understanding of the transcrip- affect cellular function potentially identifying important
tional and functional proteomic effects of these mutations, markers and therapy targets in ovarian cancer. Approximately
changes induced in response to in vitro and in vivo targeting 70 human genes are now known to be imprinted with only
of the protein product of the mutant gene and its interaction one allele expressed at conception, during embryonic devel-
with other genomic changes. opment and in each normal adult cell. Several imprinted genes
including ARHI (NOEY2) and LOT1/ZAC function as tumor sup-
2.4. Gene copy number aberrations pressor genes, and these and other potential tumor suppres-
sors (e.g. RASSF1A, DLEC1, WWOX, HIC1, OVCA1 and OVCA2)
Genomic instability is thought to represent at least two dif- may be lost in ovarian cancer, either by epigenetic mecha-
ferent processes in ovarian carcinogenesis that occur at least nisms and/or deletions [52,53]. New comprehensive profiling
in part due to bridge-fusion-breakage that occurs at telomeric approaches to detect methylation patterns and to explore the
crisis [24]. Of less importance are those genomic areas that functional proteomic and other effects of imprinted genes are
are destabilized as a result of the tumorigenesis process per likely to identify new candidates [54]. To date, however, clin-
se. Likely more important are particular areas of the genome ical studies have not yet begun to assess therapies that are
that may be targeted and selected for amplification or deletion targeted to the majority of these aberrations.
specifically due to their functional importance for tumor ini-
tiation and progression. Although such aberrant areas of DNA 2.5. Proteomic studies
frequently carry multiple genes, it is presently thought that
only a limited number of the multiple genes are ‘key drivers’ Since DNA copy number, mutation, rearrangement and
of the process; these ‘key drivers’ are thought to be the most methylation alterations ultimately act coordinately to alter
124 u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132

critical cellular functions through changes at the protein level, onic pathways involving HOX genes [60]. These genes encode
it is necessary to develop and implement functional pro- transcription factors that control developmental patterning
teomics approaches that can assay post-translational and along the anterior-posterior axis, and the temporal and spatial
other modifications of proteins. New mass spectroscopy plat- expression of these genes is tightly coupled to their clustered
forms and the efforts of the Clinical Proteome Analysis organization. HOX genes are not normally expressed in OSE
Technology Consortium are providing new approaches and [60]. However, expression of HOXA9, HOXA10 and HOXA11 in
opportunities. We have focused our efforts on development of tumorigenic mouse OSE-derived cells has been demonstrated
a novel, sensitive, moderate-throughput and quantitative pro- to induce cells to differentiate along distinct mullerian lin-
teomic technology known as reverse phase tissue lysate array eages, giving rise to tumors that exhibit morphologic features
(RPPA) [55,56]. RPPA is primarily a candidate gene approach that are characteristic of serous, endometrioid and mucinous
based on known proteins with high quality antibodies. It is ovarian tumors, respectively (Table 3). HOXA7, a gene located
ideally suited to determine a ‘global’ proteomic view of sig- 3 of HOXA9, has been additionally found to control the degree
naling changes induced by genomic aberrations as well as of differentiation and grade of ovarian tumors [60]. Since sex
pharmacodynamic signaling changes that occur in response steroids regulate HOX expression throughout the menstrual
to the targeting of aberrant proteins [57]. The global functional cycle in the adult, it is plausible that prolonged exposure of
proteomic information derived from RPPA studies can thus OSE cells to these hormones in the adult female contributes
facilitate definition of optimal combination approaches that to inappropriate HOX activation, perhaps in the context of
will most effectively inhibit important signaling pathways in epithelial inclusion cysts and excessive autocrine/paracrine
ovarian and other cancers. stimulation leading to proliferation and genomic instability.

2.6. Histology-specific molecular changes in ovarian


cancer
3. Novel targets and approaches for ovarian
cancer therapy
Transcriptional profiling of ovarian cancers has revealed
marked differences in gene expression patterns between his- 3.1. Kinases: receptor tyrosine kinases
tologic subtypes [58]. Further, as discussed, there are distinct
genomic aberrations that are clearly important to the patho- EGFR and HER2 are validated as important treatment targets
genesis of specific histologic subtypes (Table 2). For example, in cancers of the lung, head and neck, and breast, diseases
the importance of PTEN loss to endometrioid ovarian carcino- in which these receptors are frequently genomically abnor-
genesis is supported by mouse models [59]. Together [25,29], mal. However, they are less frequently genomically aberrant
these observations provide considerable support to the long- in epithelial ovarian cancer. The prognostic significance of
held notion that the different subtypes of epithelial ovarian HER2 in ovarian cancer is less clear than in breast can-
cancer should be regarded as separate disease entities with cer and responses to therapy with the monoclonal antibody
distinct pathogenesis pathways and potential therapy targets. trastuzumab have been disappointing, probably due to the
Furthermore, genetic analysis indicates that cystadenomas, low frequency of gene amplification [61]. In parallel, although
borderline tumors and carcinomas do not represent a stepwise studies are attempting to exploit other membrane receptors
progression for the serous subtype, although such a colorectal including EGFR and c-kit in ovarian cancer with tyrosine
cancer-type sequence may exist for mucinous tumors [29]. kinase inhibitors such as gefitinib (inhibits EGFR) and imatinib
Although the different subtypes of epithelial ovarian (inhibits c-kit), the rationale is frequently based on little mech-
cancer (serous, endometrioid, mucinous) possess unique anistic consideration other than protein (over)expression [9].
molecular aberrations (Table 2), their morphologic features In the absence of frequent genomic aberrations of these recep-
resemble the specialized epithelia of the reproductive tract tor tyrosine kinases in ovarian cancer and with conflicting
that derive from the mullerian ducts and recent studies sug- data regarding their prognostic significance, it may be that
gest that they may all arise from a single OSE precursor cell therapies designed to specifically exploit them will not sig-
with the specific path of differentiation regulated by embry- nificantly impact the outcome of ovarian cancer patients.

Table 3 – Important role for the homeobox genes HOXA7, A9, A10, and A11 in the morphologic heterogeneity of epithelial
ovarian cancers and their assumption of mullerian-like features [60].
Homeobox (HOX) gene Normally expressed ina Epithelial ovarian Stable expression in
cancer expression transformed mouse OSE
cells induces formation of

HOXA9 Fallopian tubes, endometrium, Serous, endometrioid, Cystic, papillary tumors


endocervix mucinous resembling serous carcinomas
HOXA10 Endometrium, endocervix Mucinous, Glandular tumors resembling
endometrioid endometrioid carcinomas
HOXA11 Endocervix, lower uterine Mucinous Tumors resembling mucinous
segment carcinomas

a
Little or no staining for HOX proteins is present in normal human ovarian surface epithelial cells.
u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132 125

Table 4 – Epidermal growth factor receptor (EGFR)/HER2/phosphatidylinositol-3-kinase (PI3K) signaling pathway


abnormalities in ovarian cancer. PIK3CA—the gene encoding the catalytic p110␣ subunit of PI3K; p85 – the regulatory
subunit of PI3K.
Gene Genomic alteration in tumors Epithelial ovarian cancer subtype Alteration frequency in subtype

PIK3CA Amplification Prevalent in all high-grade epithelial 25–40%


ovarian cancers (mostly serous)
PIK3CA Activating mutations Serous epithelial ovarian tumors 2%
Endometrioid ovarian tumors 20%
Clear cell ovarian tumors 20%

PTEN Mutations Endometrioid ovarian tumors only (6–8% 50% (but rare in other subtypes)
of all epithelial ovarian tumors)
p85 Activating Mutations High-grade epithelial ovarian cancers Rare
AKT1 Activating mutations Not defined as of yet 2%
AKT2 Amplification High-grade epithelial ovarian cancers 5%
HER2 Amplification High-grade epithelial ovarian cancers 8%
EGFR Protein expression High-grade epithelial ovarian cancers 30–70%, although genomic
changes (amplification, mutation)
are rare

3.2. Kinases: PI3K/AKT signaling the treatment of borderline and low-grade ovarian tumors.
Although farnesyl transferase inhibitors and sorafenib were
Genomic aberrations commonly activate protein signal- originally developed to inhibit ras signaling, current interest
ing through the oncogenic PI3K/AKT pathway downstream resides with newer more specific inhibitors such as the MEK
from receptor tyrosine kinases in epithelial ovarian cancer inhibitor AZD6244 (AstraZeneca).
(Table 4) [44]. Given the evidence suggesting that cancer
cells with genomic anomalies activating PI3K/AKT signaling
are extremely sensitive to inhibitors of this signaling path- 3.4. Kinases: protein kinase C iota (PKCi)
way (‘oncogene addiction’) [28,44,62,63], PI3K/AKT pathway
PKCi is activated by K-ras and is also located near the peak of
inhibitors have exciting potential for ovarian cancer treat-
the 3q26.2 amplicon in high grade ovarian cancers. PKCi is a
ment. Indeed, the PI3K inhibitor LY294002 decreases ovarian
key catalytic regulator of the Par3-Par6-atypical PKC complex,
cancer cell proliferation and ascites formation and increases
a complex required for localization of E-cadherin to cell junc-
apoptosis induced by paclitaxel and other chemotherapies in
tions, for maintenance of tight and adherens junctions, and for
vitro and in vivo. Although this particular drug is not suitable
the establishment of epithelial cell polarity [65]. Many growth
to clinical development because of poor solubility and a short
factors are normally compartmentalized by tight junctions in
half life, several companies now have PI3K/AKT inhibitors
polarized cells. Loss of polarity is a feature of epithelial ovarian
in early phase clinical trials (SF1126, perifosine, XL147 from
cancer and allows growth factors and signaling complexes to
Exelixis (San Francisco, CA), BEZ235 from Novartis (East
interact aberrantly and to mediate autocrine cell activation.
Hanover, NJ)). Inhibitors of the PI3K/AKT pathway-related
Thus, deregulation of PKCi likely contributes to the patho-
G protein Rab25 [45] are also now available for preclinical
physiology of ovarian cancer. Our group has demonstrated
use. Because of its role in promoting cell survival under
that PKCi is a potential therapeutic target in human ovar-
stressful conditions, inhibition of the PI3K/AKT pathway may
ian tumors, being associated with proliferation and cyclin E
demonstrate greatest efficacy in combination with cytotoxic
induction. Strikingly, coordinate elevation of cyclin E and PKCi
chemotherapy.
identifies a population of patients with a particularly poor out-
come.
3.3. Kinases: Ras signaling

In the mouse ovary, K-ras activation in OSE cells in 3.5. Kinases: Src
combination with PTEN inactivation or TAG and hTERT-
immortalization induces the formation of high-grade cancers Src, a nonreceptor tyrosine kinase, is a key mediator for multi-
[35,64]. In contrast, however, the K-ras gene is commonly acti- ple signaling pathways that regulate critical cellular functions
vated by mutation in human low-grade serous ovarian tumors and is often aberrantly activated in ovarian carcinoma [66].
and in 47% of mucinous lesions, with mutations being more In vivo, Src inhibition by AP23994, an orally bioavailable ana-
common in borderline tumors (27–60%) than in invasive (19%) logue of AP23846, significantly decreases tumor burden in
cancers; there is some evidence that mutations play a role in ovarian cancer xenografts both alone and in combination
the progression of the adenoma-borderline tumor-carcinoma therapy with docetaxel. Src inhibition also has potent antian-
sequence in invasive mucinous and in borderline/low-grade giogenic effects and may be an attractive therapeutic approach
serous ovarian carcinomas [25,29]. Further, b-Raf mutations for patients with ovarian carcinoma. Src inhibitors such as
have been identified in borderline and low-grade ovarian can- dasatinib (Bristol-Myers Squibb, New York, NY) and AZD0530
cers. Thus, there is interest in exploring the clinical role (AstraZeneca) are thus being evaluated in phase 2 clinical tri-
of drugs that inhibit signaling downstream from K-ras in als.
126 u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132

3.6. Kinases: signal transduction and activation of 3.9. Autocrine signaling and angiogenesis: VEGF
transcription (STAT) signaling
VEGF production is increased in ovarian cancer cells as a result
STAT3 is phosphorylated by Janus kinase (JAK) and Src. Phos- of mechanisms including PI3K/AKT signaling activation and
phorylated STAT3 forms dimers that can be translocated LPA (auto)stimulation [75]. VEGF stimulates angiogenesis, acts
to the nucleus, binding DNA and inducing transcription of as an autocrine/paracrine growth factor through cancer cell
genes resulting in proliferation (cyclin D, c-myc, c-Fos), sur- VEGF receptors, and is thought to be a major mediator of
vival (Bcl-2, Bcl-XL, XIAP, Survivin) and angiogenesis. STAT3 ascites production. The STAT pathway may be particularly
can be activated by growth factor (EGFR) and cytokine recep- important in mediating the effects of VEGF [75]. VEGF inhi-
tors (Interleukin-6R) or by direct interaction with Src or bition reduces ascites formation in mouse models and the
Abl. Nuclear localization of phosphorylated STAT3 has been ability of ovarian cancer cell lines to migrate in vitro [68].
observed in 71% of ovarian cancers and is associated with The anti-VEGF antibody bevacizumab has produced exciting
decreased overall survival [67]. Autocrine and paracrine acti- antitumor activity in phase 2 clinical trials in women with
vation of STAT3 stimulates cell proliferation and motility in recurrent chemoresistant ovarian cancer both alone and in
vitro. JAK and STAT inhibitors have demonstrated antitumor combination with cytotoxic drugs such as oral cyclophos-
activity in ovarian cancer preclinical models [68,69]. phamide [10,11,76]. Thus, it is currently being evaluated by
the GOG and other groups in phase III clinical trials in com-
bination with paclitaxel and carboplatin as first-line therapy
3.7. PARP
for women with epithelial ovarian cancer. Other inhibitors of
angiogenesis including VEGF Trap (AVE0005), small molecule
PARP inhibitors such as AZD2281 (AstraZeneca) induce cancer-
inhibitors of VEGF receptors (e.g. AZD2171/Cediranib (Astra
specific synthetic lethality in homologous recombination
Zeneca)) and angiopoietin inhibitors (e.g. AMG386) are also
repair-defective cells, including BRCA1/2-mutant tumors [70].
in clinical trials in ovarian cancer (ClinicalTrials.gov). Pre-
In the expansion phase of a phase 1 clinical trial in women
clinical data suggest that inhibitors of both VEGF receptors
with BRCA-deficient ovarian cancer, toxicities were mild and
and platelet-derived frowth factor receptor (PDGFR) may have
14 of 32 patients, the majority of whom had platinum resis-
more antitumor activity than VEGF inhibitors alone [77].
tant/refractory disease, achieved a partial response. Thus,
AZD2281 is well tolerated and demonstrates compelling
3.10. Autocrine signaling: endothelins
activity in patients with BRCA-deficient ovarian cancer. Com-
bination studies of PARP inhibitors with cytotoxic drugs are
The endothelin A peptides (ET-1, ET-2, and ET-3) are potent
now proceeding (ClinicalTrials.gov).
mitogens for several human tumors. ET-1 and its ETA receptor
(ETAR) are overexpressed in primary and metastatic ovar-
3.8. Autocrine signaling: phospholipids and cytokines ian cancers, providing the potential for autocrine stimulation
[78]. Interaction of ET-1 with ETAR activates a pertussis toxin-
PKCi can also activate phospholipase D2 (PLD2), thus increas- insensitive G protein that stimulates phospholipase C gamma
ing phospholipid release from the membrane [71]. In addition, activity, increases intracellullar calcium and activates PKC,
PLD1 may be located in the 3q26 amplicon in epithelial ovarian MAPK, and p125 focal adhesion kinase (FAK) phosphorylation.
cancer. Our group has also demonstrated that mRNA lev- Interaction with ET-1 also transactivates EGFR, stimulates pro-
els of lipid phosphate phosphatase, an enzyme involved in liferation, blocks apoptosis, activates integrin-like kinase (ILK),
lysophospholipid degradation which markedly reduces ovar- upregulates matrix metalloproteinases (MMPs), and increases
ian cancer growth when reintroduced into cells, is decreased vascular endothelial growth factor (VEGF) expression enhanc-
in the majority of ovarian cancers and cell lines [72]. Thus, ing angiogenesis. Clinical trials targeting endothelin signaling
lysophospholipid levels are elevated in the ascitic fluid and in ovarian cancer are thus underway.
blood of ovarian cancer patients. Lysophosphatidic acid (LPA),
an autocrine/paracrine phospholipid growth factor synthe- 3.11. Adhesion and invasion of cancer cells
sized from lysophosphatidylcholine by lysophospholipase D
(autotaxin), is found at high concentrations in ascites and at Normal OSE cells bind to laminin and collagen in the base-
detectable levels in the plasma of most ovarian cancer patients ment membrane [79]. Adhesion to collagen appears to be
and is a potent inducer of cell proliferation, survival, resis- maintained in ovarian cancer cells. In cell culture, ovarian
tance to cisplatin, activation of proteases, invasiveness, VEGF cancer cell lines bind preferentially to type I collagen through
production, glycogen synthase kinase 3 (GSK-3) inhibition, IL- alpha2beta1 integrin. Alteration in adhesion to laminin may
6, and IL-8 transcription in ovarian cancer cells [73]. Indeed, be an important step in ovarian oncogenesis. The distinc-
cytokines such as IL-6, IL-8, TNF␣, IL-1␣, CCL2, CXCL8 and M- tive pattern of metastasis to the peritoneal surface involves
CSF and their receptors have all been implicated in ovarian binding of ovarian cancer cells to peritoneal mesothelial cells,
cancer growth and proliferation [74]. In addition, increased mediated by B1 integrin as well as CD44 and the mucin MUC16
levels of LPA receptors (LPA2 and LPA3) in ovarian cancer cells (CA125) on the surface of ovarian cancer cells [80]. Mesothe-
contribute to growth and metastatic capability. Thus, LPA and lial cells express fibronectin, laminin and type IV collagen
several cytokines represent highly valid autocrine targets in that bind to integrins that contain the B1 subunit, hyaluronic
ovarian cancer and a number of specific antagonists are cur- acid that binds to CD44 and mesothelin that binds to MUC16
rently in preclinical evaluation. (CA125) in 80% of ovarian cancers [81]. The expression of
u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132 127

multiple splice variant forms of CD44 occurs in ovarian can- venous paclitaxel in platinum-resistant ovarian cancer at MD
cer but the significance of this is unclear [82]. Anti-integrin, Anderson Cancer Center (MDACC). The E1A gene, conveyed by
anti-mesothelin and anti-CD44 antibodies as well as MUC16 adenovirus Type 5, was shown to downregulate HER-2/neu and
knockdown can block the binding of ovarian cancer cells to increase cancer cell apoptosis in patients with ovarian cancer.
mesothelial cells and reduce invasion and the frequency of siRNAs incorporated into neutral liposomes are being
peritoneal metastases [83,84]. Oregovomab, a murine mon- developed to facilitate delivery of siRNAs that can target virtu-
oclonal antibody to CA125, is currently in Phase III testing ally any oncogene to cancer cells in vivo. This technology has
[85]. However, this antibody has recently been found not to already been used successfully in animal models [88] and will
be effective as maintenance therapy after front-line treatment soon be introduced into phase 1 clinical trials at MDACC.
of patients with advanced ovarian cancer [86]. The authors of
this phase III study concluded that future studies of this and
3.13. Immune therapy approaches
other tumor antigen-specific immunization strategies should
seek ways to augment induced immunity.
Pemtumomab, a human milk fat globule membrane (HMFG),
is a 90Yttrium-radiolabeled murine IgG1 monoclonal anti-
3.12. Gene therapy body against polymorphic epithelial mucin, which is highly
expressed in ovarian cancer. Despite promising phase II stud-
Despite modest activity in preclinical models, adenovirus- ies [89], however, a phase III study (Study of Monoclonal
mediated gene therapy for ovarian cancer remains limited in Antibody RadioimmunoTherapy [SMART]) using this agent
vivo by inefficient and nonspecific gene transfer, and phase III was closed after failing to meet superiority criteria. Current
studies were stopped early. Adenoviral targeting of mesothe- agents being evaluated include the trifunctional anti-CD3
lin, a cell surface glycoprotein which is overexpressed in antibody catumaxomab, the investigational gene transfer
ovarian cancer but not in normal tissues except mesothelial agent EGEN-001 (phIL-12-005/PPC) that contains the human
cells, makes it a candidate for transcriptional and transduc- interleukin-12 gene in a special carrier system designed to
tional gene therapy targeting [87]. The NCI has supported an enter cancer cells, and MORAb-003 (farletuzumab), a human-
ongoing Phase I/II study of intraperitoneal tgDCC-E1 and intra- ized IgG1 antibody that targets over-expressed folate receptor

Fig. 2 – A systems approach to the integration of genomic and functional proteomic data for identification of rational
molecular strategies for the treatment of cancer. A comprehensive list of aberrant genes identified in chromosomal copy
number and sequence studies can be filtered using expression arrays and functional proteomic assays (e.g. reverse phase
protein array). The resultant targets are likely to represent important ‘drivers’ of oncogenesis and can be mapped using
network analysis approaches into interconnecting pathways. The latter can then be used for the design of rational therapy
approaches in individual patients (from ref. 94 with permission).
128 u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132

alpha in ovarian cancer (ClinicalTrials.gov). MORAb-003 has MMP inhibitors (MMPI) enjoyed a brief limelight with some
received orphan drug designation by the Food & Drug Admin- effect on decreasing CA125 when given to patients with ovar-
istration (FDA) for ovarian cancer. ian cancer and ascites. However, joint toxicities discouraged
further development [90]. A less toxic MMPI, BAY 12-9566, was
studied in Phase III both in small cell lung cancer and ovarian
cancer in combination with first-line chemotherapy. Unfortu-
3.14. Cell cycle inhibitors and extracellular matrix
nately, no additive effect could be documented.
inhibitors

Flavopiridol was the prototypic cyclin inhibitor, but its further 3.15. DNA methylation and acetylation
development was discontinued in favor of newer drugs [90].
Ispinesib selectively inhibits the mitotic motor protein kinesin Preclinical models have established the rationale for using
spindle protein (KSP) and phase II studies are underway DNA demethylating agents including azacitidine to reverse
(ClinicalTrials.gov). Also under development are a telomerase chemoresistance in ovarian cancer [91]. Indeed, a phase IIa
inhibitor, GRN163L (Geron Corporation) and N-cadherin antag- study of a sequential regimen using azacitidine with carbo-
onists (Adherex Technologies, Inc.). platin in patients with platinum resistant epithelial ovarian

Fig. 3 – Our current working concept regarding the mechanisms of ovarian carcinogenesis. During the menstrual lifetime of
a female, ovarian surface epithelial (OSE) cell damage and repair occur with each ovulation to facilitate release of the ovum.
This happens under the control of local paracrine hormone and growth factor release from the ovarian follicle and corpus
luteum in addition to cycling levels of gonadotrophins. Prolonged and aberrant exposure of OSE cells to local growth factors
leads to a risk of genomic changes; this may be facilitated by ‘cell trapping’ within epithelial inclusion cysts. Hereditary
predisposition to genomic damage increases the risk of early-onset ovarian cancer as occurs, for example, in BRCA1
mutation carriers. Early genomic changes include the histology-specific aberrations such as p53 mutation and lead to loss
of growth control. Aberrant OSE cells acquire the ability to control their own growth in an autocrine fashion with activation
of kinase signaling pathways and begin to differentiate inappropriately under the possible influence of excessive and
inappropriate hormone and growth factor exposure. The nature and duration of exposure may control the exact pattern of
homeobox gene expression as in embryonic development. Increasing genomic instability then ensues with further
autocrine growth stimulation and kinase activation.
u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132 129

cancer conducted at MDACC has demonstrated the ability of interrogation of large numbers of stored cancer samples with
a hypomethylating agent to reverse platinum resistance [92]. long term follow up data. Ultimately, an improved ability to
combine information from global genomic, transcriptional,
proteomic and functional technologies (Fig. 2) to identify and
4. Future directions characterize cooperating events between multiple genes and
genomic aberrations will be necessary to address the com-
A systems approach to the comprehensive integration of plexity of ovarian tumorigenesis.
information derived from studies of gene copy number aber-
rations (CNA) and from other high-throughput technologies
has the potential to improve our understanding of ovarian 5. Conclusion
carcinogenesis and to facilitate the development of effec-
tive combinatorial therapies to rational targets. Although the Ovarian cancers are complex malignancies with significant
identification of the ‘drivers’ of genomic aberrations remains chromosomal instability which partly occurs as a result of
challenging, it represents an important effort that should aberrant autocrine/paracrine stimulation and activation of
improve our understanding of ovarian cancer pathophysiol- kinase signaling pathways. Fig. 3 shows our current con-
ogy and lead to improved patient management. Our group and cept regarding the mechanisms of ovarian carcinogenesis. An
others have validated potentially important ‘driver’ genes in understanding of the relationship between histology-specific
ovarian cancer genomic aberrations [41,44–51]. Such valida- genetic aberrations and the regulation of tissue histogene-
tion requires the demonstration of alterations at the DNA, RNA sis by HOX gene control is vital to the development of novel
and protein levels, functional effects on the phenotypic behav- agents for ovarian cancer treatment. It appears that we have
ior of ovarian tumor cells and an impact on patient outcomes, reached a plateau in the benefits associated with cytotoxic
all observations that support a critical role for genes in ovarian chemotherapy drugs in ovarian cancer. A comprehensive com-
carcinogenesis and as potential targets for novel therapies. pilation of aberrant events and processes in ovarian cancer
It is likely that multiple ‘drivers’ exist for most genomic will increase understanding of pathophysiology and provide
aberrations in ovarian cancer and that these ‘drivers’ func- novel therapy targets. Although the list of genomic aberra-
tion in concert with other abnormalities such as mutations tions in ovarian cancer is daunting, a systems approach and
during the development and selection of CNA structure. Fur- the integration of therapies targeting multiple component ‘tar-
ther, given the number and complexity of CNAs in ovarian and get’ genes of important genetic aberrations has potential in
other solid tumors, it is also likely that ‘driver’ genes act in a treatment and in the potentiation of chemotherapy efficacy.
coordinate fashion both within and between amplicons to gov- Indeed, synchronous targeting of coamplified oncogenes in
ern the natural history and therapy responsiveness of these the 3q26 amplicon may more profoundly inhibit ovarian tumor
tumors. A recent study of glioblastoma proposed coordinate proliferation than the targeting of single genes. In the future, it
high-resolution analysis of CNAs, mutations, methylation, will be vital that information from new high-throughput tech-
and transcriptional profiles with integration of these data into nologies be integrated in the determination and validation of
functional pathways or networks as a very efficient approach potentially important therapy targets. This will require shar-
to identify drivers and potential targets [93]. ing of databases and development of coordinating consortia. It
The generation of comprehensive data integrating genomic will also be necessary to define the optimal route of adminis-
aberrations with the transcriptional and functional pro- tration for novel therapies and how best to schedule them with
teomic effects of these aberrations unearths the possibility of cytotoxic chemotherapy. Our covenant with ovarian cancer
applying a systems approach to understanding the complex patients demands that we develop a new paradigm to make
molecular interactions that contribute to ovarian carcino- more efficient progress towards improved management of this
genesis (Fig. 2) [94]. Indeed, recent studies have begun to devastating disease.
demonstrate that an integrated view of complex copy number
and sequence alterations in cancer can identify a limited num-
ber of pathways that could prove useful for cancer diagnosis references
and therapy [93,95]. Such advances will allow the development
of novel therapy strategies in cancer that exploit global path-
way activation patterns rather than the function of individual [1] Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ, et
targets. Since the development of resistance to novel treat- al. Cancer Statistics, 2008. CA Cancer J Clin 2008;58:71–96.
[2] Lindgren PR, Cajander S, Bäckström T, Gustafsson JA, Mäkelä
ments frequently involves reactivation of the pathway that
S, Olofsson JI. Estrogen and progesterone receptors in
mediated the oncogenic effects of the inhibited target [96], ovarian epithelial tumors. Mol Cell Endocrinol
this approach may be less susceptible to the development of 2004;221:97–104.
resistance than current targeted therapy approaches. [3] Fathalla MF. Incessant ovulation—a factor in ovarian
Further advances in CGH and sequencing technologies as neoplasia? Lancet 1971;17:163.
well as in the comprehensive assessment of large numbers [4] Cramer DW, Welch WR. Determinants of ovarian cancer risk.
II: Inferences regarding pathogenesis. J Natl Cancer Inst
of tumors have the potential to provide higher resolution of
1983;71:717–21.
genomic aberrations. Technological advances such as molec-
[5] González-Martín A, GEICO Group. Treatment of recurrent
ular inversion probe (MIP) profiling are also beginning to allow disease: randomized trials of monotherapy versus
comprehensive study of the genome in formaldehyde-fixed combination chemotherapy. Int J Gynecol Cancer
paraffin-embedded (FFPE) samples since this will facilitate the 2005;15(Suppl. 3):241–6.
130 u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132

[6] Armstrong DK, Bundy B, Wenzel L, Huang HQ, Baergen R, [24] Hanahan D, Weinberg RA. The hallmarks of cancer. Cell
Lele S, et al. Intraperitoneal cisplatin and paclitaxel in 2000;100:57–70.
ovarian cancer. N Engl J Med 2006;354:34–43. [25] Hennessy BT, Mills GB. Ovarian cancer: homeobox genes,
[7] Bookman MA for the Gynecologic Cancer InterGroup (GCIG). autocrine/paracrine growth, and kinase signaling. Int J
GOG0182-ICON5: 5-arm phase III randomized trial of Biochem Cell Biol 2006;38:1450–6.
paclitaxel (P) and carboplatin (C) vs combinations with [26] Levine DA, Bogomolniy F, Yee CJ, Lash A, Barakat RR, Borgen
gemcitabine (G), PEG-lipososomal doxorubicin (D), or PI, et al. Frequent mutation of the PIK3CA gene in ovarian
topotecan (T) in patients (pts) with advanced-stage and breast cancers. Clin Cancer Res 2005;11:2875–8.
epithelial ovarian (EOC) or primary peritoneal (PPC) [27] Campbell IG, Russell SE, Choong DY, Montgomery KG,
carcinoma. J Clin Oncol 2006;24:5002. Ciavarella ML, Hooi CS, et al. Mutation of the PIK3CA
[8] Rivera E, Lee J, Davies A. Clinical development of ixabepilone gene in ovarian and breast cancer. Cancer Res 2004;64:
and other epothilones in patients with advanced solid 7678–81.
tumors. Oncologist 2008;13:1207–23. [28] Carpten JD, Faber AL, Horn C, Donoho GP, Briggs SL, Robbins
[9] Gordon AN, Finkler N, Edwards RP, Garcia AA, Crozier M, CM, et al. A transforming mutation in the pleckstrin
Irwin DH, et al. Efficacy and safety of erlotinib HCl, an homology domain of AKT1 in cancer. Nature
epidermal growth factor receptor (HER1/EGFR) tyrosine 2007;448:439–44.
kinase inhibitor, in patients with advanced ovarian [29] Shih IeM, Kurman RJ. Ovarian tumorigenesis: a proposed
carcinoma: results from a phase II multicenter study. Int J model based on morphological and molecular genetic
Gynecol Cancer 2005;15:785–92. analysis. Am J Pathol 2004;164:1511–8.
[10] Wright JD, Hagemann A, Rader JS, Viviano D, Gibb RK, Norris [30] Risch HA, McLaughlin JR, Cole DE, Rosen B, Bradley L, Fan I,
L, et al. Bevacizumab combination therapy in recurrent, et al. Population BRCA1 and BRCA2 mutation frequencies
platinum-refractory, epithelial ovarian carcinoma: a and cancer penetrances: a kin-cohort study in Ontario,
retrospective analysis. Cancer 2006;107:83–9. Canada. J Natl Cancer Inst 2006;98:1694–706.
[11] Chura JC, Van Iseghem K, Downs Jr LS, Carson LF, Judson PL. [31] Shaw PA, Zweemer RP, McLaughlin J, et al. Characteristics of
Bevacizumab plus cyclophosphamide in heavily pretreated genetically determined ovarian cancer. Mod Pathol
patients with recurrent ovarian cancer. Gynecol Oncol 1999;12:124A [Abstract].
2007;107:326–30. [32] Stephens P, Edkins S, Davies H, Greenman C, Cox C, Hunter
[12] Silverberg S. Histopathologic grading of ovarian carcinoma: C, et al. A screen of the complete protein kinase gene family
a review and proposal. Int J Gynecol Pathol 2000;19:7–15. identifies diverse patterns of somatic mutations in human
[13] International Federation of Gynecology and Obstetrics. breast cancer. Nat Genet 2005;37:590–2.
Classification and staging of malignant tumours in the [33] Sjoblom T, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, et
female pelvis. Acta Obstet Gynecol Scand 1971;50:1–7. al. The consensus coding sequences of human breast and
[14] Histological typing of ovarian tumours.Serov SF, Scully RE, colorectal cancers. Science 2006;314:268–74.
Sobin LH, editors. International histological classification of [34] Orsulic S, Li Y, Soslow RA, Gutkind JS, Varmus HE. Induction
tumours, No. 9. Geneva: World Health Organization; 1973. p. of ovarian cancer by defined multiple genetic changes in a
17–54. mouse model system. Cancer Cell 2002;1:53–62.
[15] Benda JA, Zaino R. GOG pathology manual. Buffalo, NY: [35] Dinulescu DM, Ince TA, Quade BJ, Shafer SA, Crowley D,
Gynecologic Oncology Group; 1994. Jacks T. Role of K-ras and Pten in the development of mouse
[16] Shimizu Y, Kamoi S, Amada S, Akiyama F, Silverberg SG. models of endometriosis and endometrioid ovarian cancer.
Toward the development of a universal grading system for Nat Med 2005;11:63–70.
ovarian epithelial carcinoma: testing of a proposed system [36] Yang G, Rosen DG, Mercado-Uribe I, Colacino JA, Mills GB,
in a series of 461 patients with uniform treatment and Bast Jr RC, et al. Knockdown of p53 combined with
follow-up. Cancer 1998;82:893–901. expression of the catalytic subunit of telomerase is
[17] Shimizu Y, Kamoi S, Amada S, Akiyama F, Hasumi K, sufficient to immortalize primary human ovarian surface
Silverberg SG. The grader and the typer should be friends: a epithelial cells. Carcinogenesis 2007;28:174–82.
study of comparative value of histopathologic grading and [37] Alvero AB, Fishman DA, Qumsiyeh MB, Garg M, Kacinski BM,
typing of ovarian epithelial carcinoma [Abstract]. Mod Sapi E. Telomerase prolongs the lifespan of normal human
Pathol 1998;11:115A. ovarian surface epithelial cells without inducing neoplastic
[18] Silverberg SG. Prognostic significance of pathologic features. phenotype. J Soc Gynecol Investig 2004;11:553–61.
Curr Top Pathol 1989;78:85–109. [38] Auersperg N, Wong AS, Choi KC, Kang SK, Leung PC. Ovarian
[19] Gershenson D, Wong KK. The continuum of serous tumors surface epithelium: biology, endocrinology and pathology.
of low malignant potential and low-grade serous carcinomas Endocr Rev 2001;22:255–88.
of the ovary. In: ASCO educational book; 2007. p. 313–7. [39] Zeimet AG, Marth C. Why did p53 gene therapy fail in
[20] Hernandez E, Bhagavan BS, Parmley TH, Rosenshein NB. ovarian cancer? Lancet Oncol 2003;4:415–22.
Interobserver variability in the interpretation of epithelial [40] Wang TL, Maierhofer C, Speicher MR, Lengauer C, Vogelstein
ovarian cancer. Gynecol Oncol 1984;16:117–23. B, Kinzler KW, et al. Digital karyotyping. Proc Natl Acad Sci U
[21] Baak JPA, Langley FA, Talerman A, Delemarre JFM. S A 2002;99:16156–61.
Interpathologist and intrapathologist disagreement in [41] Shih IeM, Sheu JJ, Santillan A, Nakayama K, Yen MJ, Bristow
ovarian tumor grading and typing. Anal Quant Cytol Histol RE, et al. Amplification of a chromatin remodeling gene,
1986;8:354–7. Rsf-1/HBXAP, in ovarian carcinoma. Proc Natl Acad Sci U S A
[22] Stalsberg H, Abeler V, Blom GP, Bostad L, Skarland E, 2005;102:14004–9.
Westgaard G. Observer variation in histologic classification [42] Hennessy BT, Nanjunden M, Cheng KW, Nolden L, Mills GB.
of malignant and borderline ovarian tumors. Hum Pathol Ovarian cancer: identification of remodeling and spacing
1988;19:1030–5. factor 1 (rsf-1, HBXAP) at chromosome 11q13 as a putative
[23] Suzuki S, Moore 2nd DH, Ginzinger DG, Godfrey TE, Barclay J, oncogene in ovarian cancer (News and Commentary). Eur J
Powell B, et al. An approach to analysis of large-scale Human Genet 2006;14:381–3.
correlations between genome changes and clinical [43] Watanabe T, Imoto I, Kosugi Y, Ishiwata I, Inoue S, Takayama
endpoints in ovarian cancer. Cancer Res 2000;60:5382–5. M, et al. A novel amplification at 17q21-23 in ovarian cancer
u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132 131

cell lines detected by comparative genomic hybridization. anti-HER2 antibody, trastuzumab, in patients with recurrent
Gynecol Oncol 2001;81:172–7. or refractory ovarian or primary peritoneal carcinoma with
[44] Shayesteh L, Lu Y, Kuo WL, Baldocchi R, Godfrey T, Collins C, overexpression of HER2: a phase II trial of the Gynecologic
et al. PIK3CA is implicated as an oncogene in ovarian cancer. Oncology Group. J Clin Oncol 2003;21:283–90.
Nat Genet 1999;21:99–102. [62] Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB. Exploiting
[45] Cheng KW, Lahad JP, Kuo WL, Lapuk A, Yamada K, Auersperg the PI3K/AKT pathway for cancer drug discovery. Nat Rev
N, et al. The Rab 25 small GTPase determines aggressiveness Drug Discov 2005;4:988–1004.
of ovarian and breast cancers. Nat Med 2004;10:1251–6. [63] Mills GB, Kohn E, Lu Y, Eder A, Fang X, Wang H, et al. Linking
[46] Eder A, Sui X, Rosen D, Nolden LK, Cheng KW, Lahad JP, et al. molecular diagnostics to molecular therapeutics: targeting
Atypical PKCiota contributes to poor prognosis through loss the PI3K pathway in breast cancer. Semin Oncol
of apical-basal polarity and Cyclin E overexpression in 2003;30:93–104.
ovarian cancer. Proc Natl Acad Sci U S A 2005;102:12519–24. [64] Liu J, Yang G, Thompson-Lanza JA, Glassman A, Hayes K,
[47] Brown LA, Irving J, Parker R, Kim H, Press JZ, Longacre TA, et Patterson A, et al. A genetically defined model for human
al. Amplification of EMSY, a novel oncogene on 11q13, in ovarian cancer. Cancer Res 2004;64:1655–63.
high grade ovarian surface epithelial carcinomas. Gynecol [65] Macara IG. Parsing the polarity code. Nat Rev Mol Cell Biol
Oncol 2005;100:264–70. 2004;5:220–31.
[48] Hughes-Davies L, Huntsman D, Ruas M, Fuks F, Bye J, Chin [66] Han LY, Landen CN, Trevino JG, Halder J, Lin YG, Kamat AA,
SF, et al. EMSY links the BRCA2 pathway to sporadic breast et al. Antiangiogenic and antitumor effects of SRC inhibition
and ovarian cancer. Cell 2003;115:523–35. in ovarian carcinoma. Cancer Res 2006;66:8633–9.
[49] Anand N, Murthy S, Amann G, Wernick M, Porter LA, Cukier [67] Rosen DG, Mercado-Uribe I, Yang G, Bast Jr RC, Amin HM, Lai
IH, et al. Protein elongation factor EEF1A2 is a putative R, et al. The role of constitutively active signal transducer
oncogene in ovarian cancer. Nat Genet 2002;31:301–5. and activator of transcription 3 in ovarian tumorigenesis
[50] Schraml P, Schwerdtfeger G, Burkhalter F, Raggi A, Schmidt and prognosis. Cancer 2006;107:2730–40.
D, Ruffalo T, et al. Combined array comparative genomic [68] Silver DL, Naora H, Liu J, Cheng W, Montell DJ. Activated
hybridization and tissue microarray analysis suggest PAK1 signal transducer and activator of transcription (STAT) 3:
at 11q13.5-q14 as a critical oncogene target in ovarian localization in focal adhesions and function in ovarian
carcinoma. Am J Pathol 2003;163:985–92. cancer cell motility. Cancer Res 2004;64:3550–8.
[51] Li P, Maines-Bandiera S, Kuo WL, Guan Y, Sun Y, Hills M, et [69] Yu Y, Luo R, Lu Z. Biochemistry and biology of ARHI
al. Multiple roles of the candidate oncogene ZNF217 in (DIRAS3), an imprinted tumor suppressor gene whose
ovarian epithelial neoplastic progression. Int J Cancer expression is lost in ovarian and breast cancers. In: Balch
2007;120:1863–73. WE, Der C, Hall A, editors. Regulators and effectors of small
[52] Kamikihara T, Arima T, Kato K, Matsuda T, Kato H, Douchi T, GTPases. Part D. Ras proteins. Methods in enzymology, vol.
et al. Epigenetic silencing of the imprinted gene ZAC by DNA 407. 2006. p. 455–67.
methylation is an early event in the progression of human [70] Fong PC, Boss DS, Carden CP, Roelvink M, De Greve J, Gourley
ovarian cancer. Int J Cancer 2005;115:690–700. CM, et al. AZD2281 (KU-0059436), a PARP (poly ADP-ribose
[53] Strathdee G, Appleton K, Illand M, Millan DW, Sargent J, Paul polymerase) inhibitor with single agent anticancer activity
J, et al. Primary ovarian carcinomas display multiple in patients with BRCA deficient ovarian cancer: Results from
methylator phenotypes involving known tumor suppressor a phase I study. J Clin Oncol 2008;26:5510.
genes. Am J Pathol 2001;158:1121–7. [71] Mwanjewe J, Spitaler M, Ebner M, Windegger M, Geiger M,
[54] Wei SH, Balch C, Paik HH, Baldwin RL, Liyanarachchi S, Li L, Kampfer S, et al. Regulation of phospholipase D isoenzymes
et al. Prognostic DNA methylation biomarkers in ovarian by transforming Ras and atypical protein kinase C-iota.
cancer. Clin Cancer Res 2006;12:2788–94. Biochem J 2001;359:211–7.
[55] Sheehan KM, Calvert VS, Kay EW, Lu Y, Fishman D, Espina V, [72] Tanyi JL, Morris AJ, Wolf JK, Fang X, Hasegawa Y, Lapushin R,
et al. Use of reverse phase protein microarrays and reference et al. The human lipid phosphate phosphatase-3 decreases
standard development for molecular network analysis of the growth, survival, and tumorigenesis of ovarian cancer
metastatic ovarian carcinoma. Mol Cell Proteomics cells: validation of the lysophosphatidic acid signaling
2005;4:346–55. cascade as a target for therapy in ovarian cancer. Cancer Res
[56] Tibes R, Qiu Y, Lu Y, Hennessy B, Andreeff M, Mills GB, et al. 2003;63:1073–82.
Reverse phase protein array (RPPA): validation of a novel [73] Fang X, Yu S, Tanyi JL, Lu Y, Woodgett JR, Mills GB.
proteomic technology and utility for analysis of primary Convergence of multiple signaling cascades at glycogen
leukemia specimens and hematopoetic stem cells (HSC). synthase kinase 3: Edg receptor-mediated phosphorylation
Mol Cancer Ther 2006;5:2512–21. and inactivation by lysophosphatidic acid through a protein
[57] Hennessy BT, Lu Y, Poradosu E, Yu Q, Yu S, Hall H, et al. kinase C-dependent intracellular pathway. Mol Cell Biol
Pharmacodynamic markers of perifosine efficacy. Clin Can 2002;22:2099–110.
Res 2007;13:7421–31. [74] Szosarek PW, Grimshaw MJ, Kulbe H, Wilson JL, Wilbanks
[58] Smith DI. Transcriptional profiling develops molecular GD, Burke F, et al. Expression and regulation of tumor
signatures for ovarian tumors. Cytometry 2002;47:60–2. necrosis factor alpha in normal and malignant ovarian
[59] Wu R, Hendrix-Lucas N, Kuick R, Zhai Y, Schwartz DR, Akyol epithelium. Mol Cancer Ther 2006;5:382–90.
A, et al. Mouse model of human ovarian endometrioid [75] Naora H, Montell DJ. Ovarian cancer metastasis: integrating
adenocarcinoma based on somatic defects in the insights from disparate model organisms. Nat Rev Cancer
Wnt/beta-catenin and PI3K/Pten signaling pathways. Cancer 2005;5:355–66.
Cell 2007;11:321–33. [76] Monk BJ, Han E, Josephs-Cowan CA, Pugmire G, Burger RA.
[60] Cheng W, Liu J, Yoshida H, Rosen D, Naora H. Lineage Salvage bevacizumab (rhuMAB VEGF)-based therapy after
infidelity of epithelial ovarian cancers is controlled by HOX multiple prior cytoxic regimens in advanced refractory
genes that specify regional identity in the reproductive tract. epithelial ovarian cancer. Gynecol Oncol 2006;102:140–4.
Nat Med 2005;11:531–7. [77] Lu C, Thaker PH, Lin YG, Spannuth W, Landen CN, Merritt
[61] Bookman MA, Darcy KM, Clarke-Pearson D, Boothby RA, WM, et al. Impact of vessel maturation on antiangiogenic
Horowitz IR. Evaluation of monoclonal humanized therapy in ovarian cancer. Am J Obstet Gynecol 2008;198:477.
132 u p d a t e o n c a n c e r t h e r a p e u t i c s 3 ( 2 0 0 9 ) 119–132

[78] Bagnato A, Salani D, Di Castro V, Wu-Wong JR, Tecce R, adenoviral vectors for ovarian cancer gene therapy. Gene
Nicotra MR, et al. Expression of endothelin 1 and endothelin Ther 2005;12:187–93.
A receptor in ovarian carcinoma: evidence for an autocrine [88] Merritt WM, Lin YG, Spannuth WA, Fletcher MS, Kamat AA,
role in tumor growth. Cancer Res 1999;59:720–7. Han LY, et al. Effect of interleukin-8 gene silencing with
[79] Moser TL, Pizzo SV, Bafetti LM, Fishman DA, Stack MS. liposome-encapsulated small interfering RNA on ovarian
Evidence for preferential adhesion of ovarian epithelial cancer cell growth. J Natl Cancer Inst 2008;100:359–72.
carcinoma cells to type I collagen mediated by the [89] Nicholson S, Gooden CS, Hird V, Maraveyas A, Mason P,
alpha2beta1 integrin. Intl J Cancer 1996;67:695–701. Lambert HE, et al. Radioimmunotherapy after chemotherapy
[80] Lessan K, Aguiar DJ, Oegema T, Siebenson L, Skubitz AP. compared to chemotherapy alone in the treatment of
CD44 and b1 integrin mediate ovarian carcinoma cell advanced ovarian cancer: a matched analysis. Oncol Rep
adhesion to peritoneal mesothelial cells. Am J Pathol 1998;5:223–6.
1999;154:1525–37. [90] Muggia F, Kosloff R. Investigational agents for epithelial
[81] Bast Jr RC, Klug TL, St. John E, Jenison E, Niloff JM, Lazarus H, ovarian cancer. Exp Rev Anticancer Ther 2005;5:855–68.
et al. A radioimmunoassay using a monoclonal antibody to [91] Li Y, Hu W, Shen DY, Kavanagh JJ, Fu S. Azacitidine enhances
monitor the course of epithelial ovarian cancer. New Engl J sensitivity of platinum-resistant ovarian cancer cells to
Med 1983;309:883–7. carboplatin through induction of apoptosis. Am J Obstet
[82] Cannistra SA, Abu-Jawdeh G, Niloff J, Strobel T, Swanson L, Gynecol 2008 [epub ahead of print].
Andersen J, et al. CD44 variant expression is a common [92] Bast RC, Iyer RB, Hu W, Kavanagh J, Coleman RL, Levenback
feature of epithelial ovarian cancer: lack of association with C, et al. A phase IIa study of a sequential regimen using
standard prognostic factors. J Clin Oncol 1995;13:1912–21. azacitidine to reverse platinum resistance to carboplatin in
[83] Strobel T, Swanson L, Cannistra SA. In vivo inhibition of patients with platinum resistant or refractory epithelial
CD44 limits intra–abdominal spread of a human ovarian ovarian cancer. J Clin Oncol 2008;26:3500.
cancer xenograft in nude mice: a novel role for CD44 in the [93] Cancer Genome Atlas Research Network. Comprehensive
process of peritoneal implantation. Cancer Res genomic characterization defines human glioblastoma
1997;57:1228–32. genes and core pathways. Nature 2008;455:1061–8.
[84] Rump A, Morikawa Y, Tanaka M. Binding of ovarian cancer [94] Hennessy BT, Gonzalez-Angulo AM, Carey MS, Mills GB. A
antigen CA125/MUC16 to mesothelin mediates cell systems approach to analysis of molecular complexity in
adhesion. J Biol Chem 2004;279:9190–8. breast cancer. Clin Cancer Res 2009;15:417–9.
[85] Braly P, Chu C, Collins Y, Edwards R, Gordon A, McGuire W, et [95] Leary RJ, Lin JC, Cummins J, Boca S, Wood LD, Parsons DW, et
al. Prospective evaluation of front-line al. Integrated analysis of homozygous deletions, focal
chemo-immunotherapy (C-IT) with oregovomab (2 amplifications, and sequence alterations in breast and
alternative dosing schedules) carboplatin-paclitaxel (C-P) in colorectal cancers. Proc Natl Acad Sci U S A
advanced ovarian cancer (OC). J Clin Oncol 2007;25:3024. 2008;105:16224–9.
[86] Berek J, Taylor P, McGuire W, Smith LM, Schultes B, [96] Berns K, Horlings HM, Hennessy BT, Madiredjo M, Hijmans
Nicodemus CF. Oregovomab maintenance EM, Beelen K, et al. A functional genetic approach identifies
monoimmunotherapy does not improve outcomes in the PI3K pathway as a major determinant of trastuzumab
advanced ovarian cancer. J Clin Oncol 2009;27:418–25. resistance in breast cancer. Cancer Cell 2007;12:
[87] Breidenbach M, Rein DT, Everts M, Glasgow JN, Wang M, 395–402.
Passineau MJ, et al. Mesothelin-mediated targeting of

You might also like